Abstract
The gut microbiome has emerged as an important modulator of cancer progression and therapy response, yet the role of its fungal component, the gut mycobiome, remains poorly understood. In this review, we summarize current knowledge on the relationship between intestinal fungi and cancer, with a particular focus on the potential influence of the gut mycobiome on the efficacy of immune checkpoint inhibitors (ICIs). Drawing from both human studies and preclinical models, we discuss evidence linking fungal dysbiosis to altered immune responses and therapy outcomes in cancer patients. Specific fungal taxa, such as Candida and Malassezia, have been associated with changes in the tumor microenvironment and modulation of host immunity. We also review the proposed mechanisms through which fungi may impact antitumor immunity, including interactions with immune receptors and production of bioactive metabolites. Although research in this field is still in its early stages, emerging data suggest that the gut mycobiome may serve as a biomarker for immunotherapy response and a potential target for therapeutic intervention. A better understanding of host–fungus interactions in the gut could contribute to the development of personalized strategies to improve the effectiveness and safety of cancer immunotherapy.
Keywords: Gut mycobiota, gut fungi, mycobiome, gut dysbiosis, cancer, immunotherapy, ICI, ICB
Introduction
Immunotherapy has emerged as a transformative strategy in the treatment of cancer, fundamentally altering therapeutic landscapes and patient outcomes. The development and clinical deployment of immune checkpoint inhibitors (ICIs), particularly those targeting programmed death-1 receptor (PD-1), its ligand PD-L1, and cytotoxic T-lymphocyte antigen-4 (CTLA-4), have yielded unprecedented responses in a range of malignancies including but not limited to melanoma,1,2 non-small cell lung cancer (NSCLC),3,4 and renal cell carcinoma (RCC).5,6 In parallel, advances in adoptive cell therapy, such as chimeric antigen receptor (CAR)-T cells,7 and cytokine-based treatments8 have further expanded the arsenal of immuno-oncological tools.
Despite these advances, many patients do not respond to ICIs9 or develop immune-related adverse events (irAEs),10 prompting efforts to identify factors that influence treatment outcomes. These challenges have led to a growing body of research into the factors influencing immunotherapy efficacy, with the gut microbiota emerging as a critical modulator of treatment outcomes. Landmark studies have demonstrated that specific bacterial taxa within the gut can affect the activity of ICIs, modulate systemic cytokine profiles, and enhance tumor-specific immune responses.11-15 For example, Candida albicans overgrowth has been linked to colorectal and liver cancer progression, while Malassezia expansion contributes to pancreatic carcinogenesis through complement activation.16-20
While the bacteriome has been extensively characterized in this context, considerably less attention has been paid to the fungal component of the gut microbiota – the mycobiome. This underexplored microbial kingdom comprises a smaller, yet functionally potent, proportion of the gastrointestinal ecosystem. The mycobiome includes commensal, opportunistic, and transient fungal species belonging to Candida, Malassezia, Saccharomyces, and Cladosporium genera, among others, which interact with host immunity and microbial communities within a human body.21-24 Historically, the study of gut fungi has been hampered by technical challenges, including lower fungal biomass, difficulty in DNA extraction, and limited reference databases.25 However, recent advances in sequencing technologies and bioinformatics have begun to overcome these barriers, enabling more accurate profiling of the gut mycobiome and revealing its potential as an independent or synergistic modulator of immune responses.
Although fungi constitute only a minor fraction of the gut microbiome, they possess unique cellular components, including ergosterol-containing membranes, chitin-based cell walls, and large genomes capable of encoding a broad array of virulence factors. Fungal pathogen-associated molecular patterns (PAMPs) such as β-glucans, mannans, and chitin are recognized by innate immune receptors, particularly C-type lectin receptors (CLRs),26,27 the main class of fungal-sensing pattern recognition receptors, including Dectin-1, Toll-like receptor 2 (TLR2), and Mincle. Their engagement triggers inflammatory signaling cascades and modulates adaptive immunity, influencing T cell development and polarization, dendritic cells (DCs) function, and the production of key cytokines such as interleukin (IL)-17 and IL-22, central to mucosal immune responses.28,29
Furthermore, fungi contribute to the gut metabolome by producing a variety of substances, including ethanol, acetaldehyde, oxylipins, and other bioactive lipids that may modulate epithelial integrity, immune tolerance, and cross-talk with bacterial symbionts.30-32 When the balance of the mycobiome is disrupted, such as through overgrowth of particular species or loss of fungal diversity, these metabolic functions can shift toward proinflammatory or pathogenic outcomes. Such dysbiosis has been linked to gastrointestinal and systemic inflammatory diseases, and emerging evidence suggests it may also play a role in cancer development and treatment response.
In this review, we provide emerging evidence on the role of the gut mycobiome in cancer immunotherapy. We discuss its composition, functional capabilities, and interactions with the host immune system. We also explore its impact on tumor progression and immunotherapy efficacy.
The gut mycobiome in cancer
Fungal dysbiosis in cancer is not limited to changes in abundance or diversity; it also entails profound shifts in functional capacity, with direct consequences for host immunity and metabolism. In cancer patients, the gut mycobiome undergoes substantial remodeling, both in taxonomic composition and ecological structure. These alterations are increasingly recognized as important modulators of disease progression, immune dynamics, and responsiveness to therapy. A key aspect of this remodeling is the change in fungal diversity. For instance, colorectal cancer (CRC) and hepatocellular carcinoma (HCC) are both characterized by a markedly reduced richness of gut fungal communities.18-20 However, this pattern is not universal: fungal diversity may increase in certain extraintestinal tumors such as lung adenocarcinoma and melanoma, likely reflecting differences in the local conditions rather than gut-specific effects.16,17 These differences likely reflect distinct local conditions, such as tissue oxygenation, nutrient availability, immune surveillance, and epithelial barrier properties.
Regardless of directionality, fungal dysbiosis in cancer is typified by reproducible taxonomic shifts. Although the fungal composition varies by cancer type, specific signatures have been reported: Candida spp. enrichment in CRC and HCC, Malassezia expansion in pancreatic cancer (PDA), Aspergillus in CRC, and increased diversity with enrichment of Saccharomyces and Aspergillus in lung adenocarcinoma and melanoma.16-20 Overall, opportunistic and immunomodulatory genera, especially Candida, Malassezia, or Aspergillus, are frequently enriched across tumor types. Among them, Candida tropicalis and C. albicans have emerged as key contributors to the altered fungal landscape, with consistent overrepresentation reported in CRC, HCC, PDA and lung, and skin cancers.33-40 Both species contribute to disease pathology via secretion of secreted aspartyl proteases (SAPs), phospholipases, and candidalysin, which disrupt epithelial barriers, activate Th17 responses, and promote chronic inflammation, thereby reshaping the tumor microenvironment.41
In CRC, a significant increase in fungal burden, especially of C. tropicalis, is accompanied by a notable reduction in overall fungal diversity compared to healthy individuals.34-36 Furthermore, the Ascomycota/Basidiomycota ratio, a marker of ecological imbalance within the fungal community, is increased in CRC patients.42 Taxa such as Candida, Malassezia, and Aspergillus flavus (a known aflatoxin producer) are consistently enriched in this context.35,42 Candida spp. contribute to disease pathology through the production of virulence factors such as SAPs, phospholipases, and candidalysin, which collectively damage epithelial barriers and trigger pro-inflammatory cytokine responses.41
In HCC, intestinal overgrowth of C. albicans, often alongside Malassezia furfur, has been associated with microbial imbalance and enhanced gut inflammation.37 These alterations may influence both local mucosal immunity and systemic immune responses.
In PDA, gut enrichment of Malassezia is linked to complement activation via the mannose-binding lectin (MBL) pathway and to enhanced IL-33 secretion, both of which contribute to a pro-inflammatory milieu.43 Moreover, C. albicans plays a central role in shaping antifungal T helper (Th)17 immunity in humans, acting as the dominant antigen for Th17 cell activation. Intestinal inflammation can drive the expansion of both C. albicans-specific and cross-reactive Th17 cells, potentially impacting immune surveillance and tumor progression.38
Remarkably, the influence of the gut mycobiome extends beyond gastrointestinal malignancies to anatomically distant cancers, such as those of the lung and bladder. In lung adenocarcinoma, for example, fungal diversity is paradoxically increased. This shift is characterized by the enrichment of genera such as Saccharomyces, Aspergillus, and Apiotrichum, while Candida species appear to be diminished.44 These findings suggest that distinct fungal signatures may be linked to tumor-specific immune landscapes. For example, in melanoma, enrichment of C. albicans and Malassezia restricta was associated with increased PD-L2 expression and regulatory immune cell infiltration,39,45 while in CRC, fungal dysbiosis drives Th17 polarization and MDSC accumulation,34,41 both shaping tumor-specific immune landscapes.
In our study,39 we found that gut mycobiota composition differed significantly between melanoma patients and healthy individuals and underwent dynamic changes during anti-PD-1 immunotherapy. Melanoma patients exhibited an increased abundance of potentially pro-inflammatory fungi such as C. albicans, Candida dubliniensis, Neurospora crassa, and M. restricta, and decreased levels of potentially beneficial species like Saccharomyces cerevisiae and Debaryomyces hansenii. Notably, C. albicans and M. restricta levels increased during immunotherapy and were associated with poorer response to the therapy and overall worse clinical outcomes, including higher risk of disease progression and shorter progression-free survival. Conversely, Saccharomyces paradoxus was associated with treatment response. These changes correlated with immune profiles: responders showed lower fungal diversity and positive correlations between C. albicans and markers of adaptive immunity, while in non-responders, C. albicans correlated with regulatory and suppressive immune cells. Of note, M. restricta was linked to increased PD-L2 expression in non-responders, potentially contributing to immune evasion.45
Crucially, the gut mycobiome does not operate in isolation. Emerging evidence suggests that the mycobiome interacts with the bacteriome and virome to shape the overall immunological tone of the gut and distant tissues.46-48 Interkingdom interactions between fungi and bacteria, encompassing both competition and metabolic interplay, can significantly influence host immunity. For instance, Candida overgrowth in the gut has been associated with increased lactate-producing bacteria and a concurrent decrease in short-chain fatty acid (SCFA)-producing taxa. Recent findings by Seelbinder et al. demonstrated that in lung cancer patients, expansion of intestinal Candida was driven by an ecological shift characterized by increased lactate-producing bacteria and reduced SCFA producers. This niche favored fungal proliferation by enabling Candida to utilize lactate as a nutrient, giving it a competitive advantage over other fungi.49 This microbial shift alters the mucosal immune environment and may have downstream effects on systemic anti-tumor immune responses.17,50 Co-aggregation of fungi and bacteria in biofilms, as well as metabolic exchange between them, further exacerbate inflammation and immune evasion.51,52
These observations point to a dynamic role of the gut mycobiome in modulating host immunity during cancer. Depending on the fungal species present, their abundance, and their interactions with immune cells, the gut mycobiome can either promote tumor-associated inflammation or support anti-tumor immune surveillance, reflecting its dual and context-dependent role in cancer. Through systemic cytokine modulation, epithelial barrier regulation, and cross-talk with the bacterial microbiota, fungi contribute to shaping the immunological tone of the host and may influence the course of the disease. Consequently, the gut mycobiome is emerging not only as a potential biomarker of disease progression53 but also as a modifiable component of the tumor–host interface that may be harnessed therapeutically.54 Furthermore, fungal dysbiosis has been associated with increased intestinal permeability, systemic inflammation, and metabolic disturbances, all of which can compromise the patient’s resilience to treatment and alter drug pharmacokinetics.55 These findings underscore the potential of targeting the gut mycobiome as a complementary strategy to enhance the precision and efficacy of cancer therapies. Additionally, there is growing evidence of fungal translocation, particularly of Malassezia, from the gut to distant organs, including the pancreas and liver.56-58 This process appears to facilitate tumor-promoting immune signals and highlights the functional significance of gut fungi in systemic cancer biology.37,56,59
In summary, gut fungal dysbiosis in cancer is characterized by decreased diversity, overrepresentation of pathobionts, ecological imbalance, and disrupted microbial cooperation, all converging on immune modulation and cancer progression. These insights provide a foundation for developing diagnostic and therapeutic strategies targeting the mycobiome.
Gut mycobiome as a modulator and predictor of cancer immunotherapy response
In a recent study, Hu et al.60 demonstrated that specific characteristics of the gut mycobiome may serve as predictive biomarkers for the efficacy of ICIs, particularly anti-PD-1/PD-L1 therapies, highlighting a previously underappreciated role of fungi in shaping CRC treatment outcomes. By analyzing metagenomic data from nearly 600 cancer patients before treatment, the authors identified two distinct gut mycobiome-based enterotypes (distinct clusters or groups of microbial communities present in the human gut microbiome) strongly associated with clinical responses to immunotherapy. Favorable-type enterotype has been characterized by higher fungal and bacterial alpha diversity, a higher Basidiomycota/Ascomycota ratio, an increased presence of butyrate-producing bacteria, and metabolic pathways related to butyric acid and sugar/starch metabolism. Moreover, when externally validated on an additional cohort comprising 125 pan-cancer patients that received PD1 antibody treatment, identified fungal enterotypes were significantly associated with immunotherapy efficacy outcomes. Importantly, fecal microbiota transplantation (FMT) from favorable-type donors enhanced response to anti-PD-1/PD-L1 therapy. These findings are consistent with earlier research demonstrating that fungal overgrowth, particularly of C. albicans, can impair host immune modulation and diminish the success of FMT.61 Such evidence underscores how fungal dysbiosis may compromise microbiota-driven immune regulation and, by extension, influence the clinical effectiveness of cancer immunotherapies. Additionally, this microbiome configuration was correlated with enhanced cytotoxic CD8+ T cell infiltration in the tumor microenvironment (TME) and improved treatment outcomes.60 Multi-omics and single-cell RNA sequencing further confirmed stronger immune activation signatures in patients with the favorable fungal-bacterial profile.
The study of Huang et al. is another large-scale, multi-cohort investigation into the predictive power of the gut mycobiota in the context of immunotherapy.62 Using metagenomic data from 862 cancer patients across nine cohorts (treated with anti-PD-1, PD-L1, or CTLA-4 therapies), the authors identified distinct fungal signatures that distinguish responders from non-responders. Notably, fungal markers alone achieved higher predictive accuracy (AUC = 0.87) than bacterial markers (AUC = 0.83), and the combination of both kingdoms in a multi-kingdom model improved prediction further (AUC = 0.89). The fungal model also correlated with prolonged overall and progression-free survival in multiple cohorts. Specific fungal taxa, such as Schizosaccharomyces octosporus, were enriched in responders and were associated with beneficial metabolic functions, such as SCFA production, which is linked to enhanced anti-tumor immunity in some research.63,64 Additionally, responders showed distinct fungal–bacterial interaction networks.62 These interactions were largely absent or less coordinated in non-responders, suggesting that fungal–bacterial synergy may be important for shaping an immune environment conducive to ICI efficacy. Mechanistically, predicted responders exhibited a TME with increased expression of exhaustion markers (PD-1, PD-1L, CTLA-4, and TIM-3), indicating a pre-activated immune state more amenable to ICI response. Patients predicted to respond based on fungal markers are more likely to benefit from PD-1 and CTLA-4 blockades, as indicated by an enriched exhausted T cell signature marked by elevated expression of PD-1 and CTLA-4.62 These findings support the hypothesis that fungal dysbiosis in the gut may influence the efficacy of ICB and melanoma progression, highlighting the gut mycobiota as a potential modulator of immunotherapy outcomes and a source of therapeutic targets or prognostic biomarkers.
In line with these large-scale studies, more focused cohort analysis by Dora et al. revealed the prognostic relevance of specific gut fungal taxa in lung cancer patients undergoing ICI therapy.65 By integrating shotgun metagenomics and ITS sequencing with CT-based radiomics in advanced NSCLC, the authors demonstrated that intestinal overrepresentation of Cortinarius davemallochii, as well as fungi from the orders Helotiales and Chaetosphaeriales, and the class Tremellomycetes correlated with short overall survival (≤6 months). In contrast, Hymenoscyphus immutabilis and Clavulinopsis fusiformis were enriched in patients with high PD-L1–expressing tumors, indicating a potential link between specific fungal taxa and tumor immune checkpoint status. Members of the Thelephoraceae family were linked to immune-related toxicities, whereas Cutaneotrichosporon cutaneum and members of Rozellomycota were more prevalent in patients without iRAEs. Interestingly, more fungal than bacterial taxa were associated with poor outcomes, suggesting that fungal over-colonization may represent a negative modulator of ICI efficacy. These findings underscore the emerging importance of the gut mycobiome not only as a biomarker of immunotherapy response but also as a potential determinant of treatment-related toxicity.
Importantly, FMT experiments in murine models demonstrated that transfer from the donor with the favorable enterotype led to increased butyrate production, upregulation of anti-tumor immune pathways, and potentiation of anti-PD-1 efficacy.60 Work by Lam et al. emphasized that FMT efficacy is governed not only by bacterial composition but also by the fungal and viral constituents of the gut microbiota, which interact in complex and dynamic ways to modulate donor engraftment and therapeutic outcomes.66 Further supporting this multi-kingdom perspective, Zuo et al. reported that the gut virome and mucosal mycobiota undergo coordinated shifts in inflammatory diseases – alterations that may influence immune checkpoint pathways.67 These findings suggest that gut fungal states could indirectly modulate systemic immunity by priming mucosal and lymphoid tissues toward either pro-inflammatory or tolerogenic phenotypes. Key fungi, such as Malassezia pachydermatis, Kalmanozyma brasiliensis, Pseudogymnoascus sp. VKMF-4246, and Thermothielavioides terrestris, showed increased presence in recipients after FMT from favorable-type donors. Of note, fungal species found to be enriched in patients with favorable responses to immunotherapy, such as Penicillium antarcticum, Aspergillus terreus, and T. terrestris, are known to produce a range of glycoside hydrolase enzymes. These include β-galactosidases, α-L-arabinofuranosidases, and enzymes involved in the breakdown of plant-derived lignocellulose.68-70 Among them, β-galactosidases are particularly notable for their role in synthesizing lactose-derived compounds like galacto-oligosaccharides (GOSs).71 GOSs are recognized for their prebiotic effects, as they support the proliferation of beneficial gut bacteria, notably butyrate-producing species such as Faecalibacterium prausnitzii,72-74 which are themselves associated with improved responses to ICIs.75-77 Altogether, these findings suggest that the gut mycobiome serves not only as a biomarker of response but also as a functional modulator of cancer immunotherapy, likely through metabolic and immunological cross-talk with the bacterial microbiota.
Host immune modulation by fungi
Fungi modulate the host immune system predominantly via activation of innate immune pathways. Fungal PAMPs, including β-glucans, mannans, and chitin localized on the fungal wall, engage pattern recognition receptors (PRRs) such as Dectin-1, Dectin-2, Dectin-3, TLR2, NOD-like receptors (NLRs), retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), and the mannose receptor (MR).78-81 Distinct PRRs recognize specific PAMPs from different fungal species, e.g., Dectin-1 primarily binds β-glucans,82 chitin is bound by LysM domain 3,83 whereas mannans are recognized by Dectin-2, Dectin-3, Mincle, MR, and DCs-specific intercellular adhesion molecule 3 (ICAM3)-grabbing non-integrin (DC-SIGN).81,84-86 Binding PAMPs by PRRs triggers intracellular signaling cascades that lead to cytokine secretion and recruitment of immune effector cells. CLRs such as Dectin-1, Dectin-2, and Mincle activate the spleen tyrosine kinase - caspase recruitment domain-containing protein 9 (SYK-CARD9) signaling axis, which plays a central role in antifungal innate immunity by driving proinflammatory responses and promoting protective Th17 cell differentiation.41,87-90 Upon fungal recognition, SYK–CARD9 signaling induces NF-κB, extracellular signal-regulated kinase (ERK), p38 MAPK, and JNK activation, leading to the production of cytokines including IL-1β, IL-2, IL-6, IL-10, IL-17, IL-18, and tumor necrosis factor alpha (TNF-α), which collectively orchestrate downstream adaptive immune responses.88,91-94 Beyond this canonical SYK-dependent route, Dectin-1 can also signal through a SYK-independent pathway involving Raf-1 and NF-κB-inducing kinase (NIK), further modulating NF-κB activity.95 This parallel pathway is thought to integrate with SYK-mediated signaling to fine-tune DCs’ responses and facilitate Th cell differentiation. Through these combined mechanisms, fungal engagement of innate immunity can shape T cell priming and polarization, potentially modulating the effectiveness of immune checkpoint blockade (ICB) therapies. An overview of the immunomodulatory effects of fungi in cancer is presented in Table 1 and illustrated in Figure 1.
Table 1. Immunomodulatory effects of fungi in cancer.
| PRRs | Immune cells | Effect on cancer | Immunomodulatory effect | Cancer types | Model | References |
|---|---|---|---|---|---|---|
| Dectin -1 | macrophages | Pro-tumoral | differentiation of macrophages toward a tolerogenic phenotype; T cell dysfunction | PDA, gastric cancer | mouse PDA + human PDA samples | [96,97] |
| Dectin -1 | MDSCs | Pro-tumoral | promotion of PGE₂ production; suppression of IL-22 binding protein expression | CRC | mouse AOM/DSS CRC + human CRC samples | [98] |
| Dectin -1 | MDSCs | Pro-tumoral | MDSCs activation, reduction of T cell cytotoxicity, promotion of the accumulation of PD-1⁺ CD8⁺ T cells | lung cancer | mouse lung cancer + human samples | [99] |
| Dectin -1 | MDSCs | Pro-tumoral | increased infiltration of Tregs and MDSCs, enhanced IL-1β production | OSCC | mouse OSCC + human samples | [100] |
| Dectin -1 | macrophages | Pro-tumoral | M2 macrophage polarization | gliomas | human | [101] |
| Dectin -1 | macrophages | Anti-tumoral | M-CSF–mediated downregulation of TLR4 and CD14 | liver cancer | mouse + human | [102] |
| Dectin -1 | macrophages | Anti-tumoral | reprogramming macrophages, enhancement of T cell responses | PDA | mouse PDA | [103] |
| Dectin -1 | macrophages | Anti-tumoral | conversion of TAMs into M1-like phenotype via SYK–CARD9–Erk pathway | lung cancer, breast cancer, melanoma | mouse | [104] |
| Dectin -1 | macrophages/NK cells | Anti-tumoral | interaction with MS4A4A in lipid rafts enhances NK activation and anti-metastatic responses | melanoma, lung cancer, renal cancer | mouse + human | [105] |
| Dectin -1 | DCs | Anti-tumoral | upregulation of IL-33 via SYK/Raf-1/NF-κB, leading to Th9 differentiation and IL-9 production | unspecified | mouse, B16 and B16-OVA, MPC-11 cell lines, | [106–109] |
| Dectin -1 | DCs | Anti-tumoral | plasma B cell differentiation, upregulation of costimulatory molecules, increased antibody production; increased CD19+ B cell infiltration in the TME, expansion of germinal center B cells | lung cancer | mouse + tumor cell lines | [110] |
| Dectin -2 | Kupffer cells | Anti-tumoral | interaction with ERMAP and galectin-9 enhances phagocytosis of metastatic cells | liver cancer | mouse + human samples | [121] |
| Dectin -2 | macrophages | Anti-tumoral | activation by BDC-3042 reprograms TAMs to a pro-inflammatory phenotype, enhancement of T cell infiltration | NSCLC, solid tumors | [122,123] | |
| Dectin -3 | macrophages | Anti-tumoral | promotes fungal clearance and epithelial repair during colitis; Dectin-3 deficiency exacerbates inflammation and tumorigenesis | CRC (colitis-associated) | mouse + fungi | [124] |
| Dectin -3 | MDSCs | Pro-tumoral | activation of NLRP3 inflammasome via JAK–STAT1 and mitochondrial ROS pathways in response to C. tropicalis; promotes MDSC accumulation and suppresses T cell function | CRC | mouse + human + fungi | [33,34] |
| Dectin -3 | macrophages | Anti-tumoral | inhibits glycolysis in TAMs, enhances CD4⁺ and CD8⁺ T cell infiltration; limits tumor growth | HCC | mouse + human | [125] |
| Dectin -3 | macrophages | Pro-tumoral | deficiency promotes C. albicans overgrowth, increased IL-7 → IL-22 in ILC3s via AhR/STAT3 → tumor-promoting inflammation | CRC | mouse + human | [126] |
| Mincle | macrophages | Pro-tumoral | expressed on M2 TAMs; promotes immunosuppressive phenotype via SYK–NF-κB signaling; associated with reduced T cell infiltration and poor prognosis | lung cancer | mouse + human + LLC, B16F10 cell lines | [132] |
| Mincle | macrophages | Pro-tumoral | senses SAP130 from necrotic cells; activates RIP1/RIP3 signaling; suppresses recruitment of immunogenic myeloid cells; facilitates immune evasion | PDA | mouse + human PDA cell lines | [133] |
| Mincle | macrophages | Anti-tumoral | activation with synthetic ligand TDB induces GM-CSF–dependent TAM repolarization toward M1-like phenotype; enhances anti-tumor immunity | Preclinical model | mouse | [134] |
Figure 1.
Schematic representation of the immunomodulatory pathways triggered by fungal components in the tumor microenvironment. This diagram illustrates the dual role of fungal ligands in modulating anti- and pro-tumor immune responses via C-type lectin receptors (CLRs) such as Dectin-1, Dectin-2, Dectin-3, and Mincle. Recognition of fungal components (e.g., β-glucans, α-mannans, TDM, SAP130) by CLRs on immune cells activates downstream signaling cascades involving Syk, CARD9, PKC-δ, and the NLRP3 inflammasome, leading to the production of cytokines such as IL-1β, IL-6, IL-9, IL-10, IL-17, and TNF-α. These pathways may promote either tumor-supporting inflammation (protumor pathway) or anti-tumor immunity through mechanisms such as phagocytosis, Th9 differentiation, or M1-like macrophage polarization.
Dectin-1 – a key fungal-sensing receptor in cancer immunity
Dectin-1 (CLEC7A), a C-type lectin receptor, exhibits dual roles in cancer immunity, acting as either an immunosuppressive or immunostimulatory agent depending on the tumor context and ligand availability. Recent studies have elucidated its capacity to either enhance anti-tumor immune responses or diminish them, in both cases through various mechanisms. Given its impact on the immune system, including T cell function, myeloid cell behavior, and cytokine signaling, Dectin-1 represents a promising therapeutic target, either alone or in combination with ICIs.
Dectin-1 as an immunosuppressive agent
Multiple studies have highlighted the immunosuppressive role of Dectin-1 signaling in the TME, particularly in PDA and other malignancies. Daley et al. demonstrated that Dectin-1 is highly expressed on tumor-associated macrophages (TAMs) in PDA, where it acts as an immunosuppressive oncogene. This signaling promotes the differentiation of macrophages toward a tolerogenic phenotype and impairs both CD4⁺ and CD8⁺ T cell function, facilitating immune evasion.96 This pathway is activated by Galectin-9, a non-fungal ligand, suggesting that Dectin-1's immunomodulatory effects extend beyond fungal recognition. Blocking Galectin-9 led to marked tumor regression and synergized with anti-PD-1 therapy, suggesting that targeting Dectin-1 or its ligand may enhance immunotherapy efficacy in PDA.96 Similarly, in gastric cancer, Dectin-1-positive TAMs are associated with T cell dysfunction and poor patient survival.97 Blocking Dectin-1 in this context reprograms TAMs and enhances the efficacy of anti-PD-1 therapy.97
Beyond macrophages, Dectin-1 is also expressed on myeloid-derived suppressor cells (MDSCs), contributing to immunosuppression in the TME. In CRC, Dectin-1 signaling in MDSCs promotes prostaglandin E₂ (PGE₂) production, suppressing IL-22 binding protein expression, facilitating tumorigenesis. Blocking Dectin-1 reduces MDSC-mediated immunosuppression and tumor progression.98 In lung cancer, Dectin-1-mediated recognition of Aspergillus sydowii via β-glucan/Dectin-1/CARD9 signaling induces MDSCs activation, reduces T cell cytotoxicity, and promotes the accumulation of PD-1⁺ CD8⁺ T cells, further contributing to immune suppression.99 Similarly, in oral squamous cell carcinoma (OSCC), Dectin-1 expression was associated with increased infiltration of regulatory T cells (Tregs) and MDSCs and enhanced IL-1β production – an effect exacerbated with age.100 Genetic ablation of Dectin-1 in mice reversed these effects, reducing intratumoral immunosuppressive cell infiltration and cytokine expression. Gliomas studies further link high Dectin-1 expression to M2 macrophage polarization and poorer prognosis, suggesting that Dectin-1 inhibition could enhance anti-tumor immunity.101
Taken together, these findings highlight a unifying mechanism across multiple tumor types, where Dectin-1 signaling fosters an immunosuppressive TME by driving tolerogenic macrophage/MDSC polarization, promoting PGE₂ and IL-1β production, and increasing checkpoint expression, all of which converge on dampening T cells cytotoxicity.
Dectin-1 as an immunostimulatory agent
In contrast, Dectin-1 signaling can also promote anti-tumor immunity when activated in distinct cellular or therapeutic contexts. Dectin-1 expression is upregulated in hepatic fibrosis and liver cancer, and its deletion exacerbates disease progression. Mechanistically, Dectin-1 suppresses TLR4 signaling in hepatic inflammatory and stellate cells, partly via M-CSF–mediated downregulation of TLR4 and CD14. Dectin-1–deficient mice show heightened cytokine responses and reduced survival in LPS-induced sepsis, while Dectin-1 activation is protective.102 Moreover, combining Dectin-1 activation via systemic β-glucan therapy with CD40 agonists has shown synergistic effects in overcoming resistance to checkpoint inhibitors in PDA models. This coactivation reprograms macrophages and enhances T cell responses, leading to tumor regression and immunological memory.103
Additionally, β-glucan, a natural Dectin-1 ligand, can convert immunosuppressive TAMs into an M1-like phenotype, promoting anti-tumor immunity. This effect is mediated through the Dectin-1-SYK-CARD9–Erk signaling pathway.104 Research by Mattiola et al. demonstrated that Dectin-1 interacts with the tetraspan molecule MS4A4A within macrophage lipid rafts. This interaction is crucial for activating natural killer (NK) cells and the subsequent resistance to cancer metastasis. Without MS4A4A, Dectin-1 signaling is impaired, leading to reduced production of proinflammatory mediators and diminished NK cell-mediated anti-tumor activity.105 Dectin-1 activation in DCs has been shown to upregulate the expression of IL-33 via the SYK/Raf-1/NF-κB signaling pathway, which in turn enhances the expression of interferon regulatory factor 4 (IRF4).106 This cascade leads to increased IL-33 production, a cytokine known to play a significant role in anti-tumor immunity.106 Furthermore, IL-33 contributes to the induction of Th9 cells, a subset of Th cells that produce IL-9 and have potent anti-tumor effects.107,108 Blocking IL-33 or its receptor ST2 inhibits Th9 differentiation from CD4+ T cells, underscoring the importance of this pathway in anti-tumor responses.108 Dectin-1-activated DCs have been identified as powerful inducers of Th9 cells. Upon activation by β-glucan ligands, Dectin-1 stimulates DCs to upregulate costimulatory molecules such as OX40L and TNFSF15, essential for Th9 differentiation. These Th9 cells produce IL-9, contributing to robust anti-tumor immunity. In vivo studies have shown that immunization with Dectin-1-activated DCs leads to significant tumor regression, primarily mediated by Th9 cells and IL-9 production.107,109 A 2025 study by Bai et al. revealed that β-glucan-induced Dectin-1 activation promotes the differentiation of plasma B cells, co-stimulatory molecule expression, and Ig production, enhancing anti-tumor immune responses.110 In mouse models of lung cancer, β-glucan combined with anti-PD-1 therapy increased CD19+ B cell infiltration in the TME, expanded germinal center B cells, and improved anti-tumor efficacy. This finding extends the known immunostimulatory roles of Dectin-1 beyond T cell-mediated immunity, highlighting its potential in orchestrating comprehensive anti-tumor responses involving multiple immune cell types.110
In summary, Dectin-1 can amplify anti-tumor immunity through coordinated reprogramming of multiple immune compartments such as macrophages, DCs, NK cells, Th9 cells, and B cells, largely via SYK–CARD9–NF-κB–dependent pathways and ligand-driven activation by β-glucans.
Context-dependent role of Dectin-1 in cancer
The impact of Dectin-1 on tumor immunity is highly context-dependent. The same receptor can induce either tolerogenic pathways (via TAM/MDSC polarization and checkpoint induction) or immunostimulatory cascades (via DC/Th9 priming, NK and B cell activation), with the net outcome dictated by ligand type and cellular context. When engaged predominantly on TAMs and MDSCs, and triggered by ligands such as Galectin-9 or tumor-associated β-glucans, Dectin-1 signaling drives immunosuppressive reprogramming, leading to PGE₂ and IL-1β production, T cell dysfunction, and resistance to checkpoint blockade. Conversely, when activated on DCs, NK cells, or B cells by structured β-glucans, Dectin-1 initiates SYK–CARD9–NF-κB signaling cascades that promote M1 macrophage polarization, IL-33/Th9 and IL-9 axis activation, NK cell–mediated cytotoxicity, and B cell differentiation, thereby amplifying anti-tumor immunity. Thus, who senses (TAM/MDSC vs DC/NK/B), what is sensed (Gal-9 vs β-glucan and its form), and where (tumor type: PDA/GC/CRC/lung/glioma vs liver or β-glucan-primable settings) together seem to forecast whether Dectin-1 tilts toward tumor promotion or tumor control (Figure 2).
Figure 2.
Context-dependent roles of Dectin-1 in cancer. Schematic overview illustrating how Dectin-1 activation can skew either toward a pro-tumor (left, immunosuppressive) or anti-tumor (right, immunostimulatory) response depending on ligand type, dominant immune cell engagement, and tumor context. On the pro-tumor side, Dectin-1 signaling in TAMs/MDSCs promotes PGE₂, IL-1β, Treg infiltration, reduced IL-22, CD8⁺ T cell exhaustion, and M2 polarization, commonly observed in PDA, gastric cancer, CRC, lung cancer, OSCC, and glioma. On the anti-tumor side, particulate β-glucan or β-glucan + CD40 agonist promote activation of DCs, macrophages, NK cells, and B cells, leading to IL-33/Th9/IL-9 signaling, M1 reprogramming, NK cytotoxicity, and B cell infiltration with germinal center expansion and antibody production, observed in liver cancer and reprogrammable PDA. Actionable predictors include expression levels of Dectin-1, Galectin-9, MS4A4A, and IL-33/Th9 activity, which may guide therapeutic interventions and identify synergy with anti-PD-1 therapy.
Box.
Context matters for Dectin-1.
Pro-tumor (immunosuppressive):
|
Anti-tumor (immunostimulatory):
|
| Who senses what, in which tumor context (where), dictates whether Dectin-1 suppresses or amplifies anti-tumor immunity. | |
Clinical implications of targeting Dectin-1 in cancer
In acute myeloid leukemia (AML), Dectin-1 has emerged as a potential biomarker for therapeutic responsiveness. A 2024 study found that AML cells exhibiting high expression of Dectin-1 and CD14, indicative of monocytic differentiation, were more sensitive to the MEK inhibitor trametinib.111 Although authors did not explain the role of Dectin-1 in details, mechanistically, Dectin-1 engagement in monocyte-derived cells can modulate MAPK/ERK signaling through SYK–CARD9 pathways, potentially influencing responsiveness to MEK inhibition. This suggests that Dectin-1 expression could serve as a predictive marker for trametinib sensitivity in AML patients. Furthermore, genetic variations in Dectin-1 have been associated with increased susceptibility to invasive fungal diseases in AML patients undergoing induction chemotherapy.112 Specifically, the rs7309123 G/G genotype was linked to a higher risk of developing pulmonary infections, including invasive fungal diseases. Although this SNP is located in an intron and as such does not alter the protein sequence directly, it may affect Dectin-1 expression or splicing, potentially modulating antifungal immune responses and increasing vulnerability to infections during immunosuppressive treatment. These findings underscore the importance of Dectin-1 in host defense mechanisms and its potential as a target for therapeutic intervention.
In melanoma models, stimulating Dectin-1 with curdlan significantly reduced tumor progression.113 Dectin-1 expression was associated with an activated myeloid cell phenotype in the TME, characterized by expression of co-stimulatory molecules such as CD80 and CD86 and antigen presentation molecules (MHC-I and MHC-II). Overexpression of these molecules is likely to enhance anti-tumor immune responses. Mechanistically, Dectin-1 activation in myeloid cells leaded to increased production of pro-inflammatory cytokines such as IL-12 and TNF-α, which in turn promoted the recruitment and activation of effector T cells within the TME, thereby amplifying anti-tumor immunity. Consistently, RNAseq analyses of bone marrow–derived neutrophils indicated that curdlan treatment induced a shift toward an activated phenotype, characterized by upregulation of pro-inflammatory cytokines (IL-23a, IL-1β, IL-6, IL-12a, IL-1R2), chemokines (CXCL10, CCL3, CCL4), and co-stimulatory molecules (CD80, CD86, CD14), along with downregulation of suppressive markers (TGF-β2, CCL22, CD209, MRC1/CD206, CD163). These changes suggest that Dectin-1 activation enhances immune activation, pro-inflammatory signaling, and metabolic regulation, contributing to a protective anti-tumor mechanism in melanoma models. Notably, this Dectin-1–mediated mechanism appeared to be tumor type–specific, as similar effects were not observed in CRC models.
In a study by Wattenberg et al,103 systemic administration of β-glucan was combined with agonistic anti-CD40 antibody therapy in mouse models of PDA, a tumor type notably resistant to ICIs. This combinatorial treatment led to complete tumor eradication and the development of immunological memory. Mechanistically, Dectin-1 activation by β-glucan, together with CD40 signaling, reprogrammed intratumoral macrophages via interferon gamma (IFN-γ)–dependent pathways, promoting a tumoricidal microenvironment. Remarkably, this anti-tumor effect was independent of classical T cell–mediated cytotoxicity and did not rely on PD-1/PD-L1 or CTLA-4 blockade, underscoring an alternative route to effective immunosurveillance. The therapeutic synergy between fungal β-glucans and CD40 agonism demonstrates that components of the mycobiota, or their molecular analogs, can be leveraged to overcome tumor-induced immunosuppression, particularly in settings where conventional checkpoint inhibition fails. These results suggest that fungal ligands such as β-glucans may serve not only as immune adjuvants but also as central modulators of myeloid-driven anti-tumor responses. Collectively, these studies illuminate the critical role of Dectin-1 in immune regulation and its potential as a biomarker and therapeutic target in cancer.
Dectin-2 CLR family
Dectin-2, along with Dectin-3 and Mincle, belongs to the Dectin-2 family of CLRs. These receptors possess short cytoplasmic tails and a single extracellular carbohydrate recognition domain (CRD). They associate with the immunoreceptor tyrosine-based activation motif (ITAM)-containing Fc receptor γ-chain (FcRγ) to initiate intracellular signaling cascades.114-116 Upon ligand binding, these receptors activate SYK, leading to the formation of the CARD9-BCL10-MALT1 (CBM) complex. This complex subsequently activates the NF-κB pathway, resulting in the production of pro-inflammatory cytokines and chemokines.117,118
Dectin-2 – from α-mannan recognition to clinical applications in cancer
Dectin-2 primarily recognizes α-mannans in a calcium-dependent manner and can form heterodimers with Dectin-3.84,85 These heterodimers exhibit enhanced binding to α-mannans, triggering more potent inflammatory responses compared to their respective homodimers.85 Through activation of the SYK–CARD9 signaling cascade, Dectin-2 induces IL-17 production in response to C. albicans, bridging innate and adaptive antifungal immunity119). Beyond classical fungal defense, Dectin-2 plays an emerging role in cancer immunity. In the liver, Dectin-2 plays a crucial role in mediating the phagocytic activity of Kupffer cells, the resident macrophages of the liver.120 Kimura et al. demonstrated that Dectin-2 expression on Kupffer cells enhances their ability to phagocytose cancer cells, thereby suppressing liver metastasis. Further mechanistic insights revealed that this phagocytic capacity depends on the formation of a bridging complex involving erythroid membrane-associated protein (ERMAP) expressed on tumor cells and galectin-9 on Kupffer cells.121 This complex delivers an “eat me” signal to Kupffer cells, promoting the clearance of metastatic cancer cells. Notably, patients with tumors exhibiting low ERMAP expression showed a higher incidence of liver metastasis, underscoring the clinical relevance of this pathway.121 While these findings highlight the tumor-suppressive role of Dectin-2 in the liver, it remains unclear whether tumor cells can exploit this mechanism to evade immune surveillance. Moreover, the upstream activators that trigger the Dectin-2 complex in the liver microenvironment and the potential involvement of intra-tumoral fungi in this process warrant further investigation.
Despite these uncertainties, and considering the evidence for Dectin-2-mediated tumor control, Bolt Biotherapeutics has developed BDC-3042, a first-in-class agonist antibody targeting Dectin-2.122 Preclinical studies have shown that BDC-3042 can reprogram TAMs from an immunosuppressive to a pro-inflammatory phenotype, enhancing antigen presentation and promoting T-cell-mediated anti-tumor responses. Currently, BDC-3042 is undergoing a Phase 1/2 clinical trial to evaluate its safety and efficacy in patients with metastatic or unresectable solid tumors, including NSCLS. However, results from early-phase clinical trials showed that therapy was well-tolerated and exhibited immunostimulatory effects, including modulation of immune cell frequencies and activation markers, supporting further investigation into C-type lectin receptor-targeted therapies.123
Dectin-3 as a context-dependent regulator of fungal sensing and tumor immunity
Dectin-3, another C-type lectin receptor, is less studied compared to Dectin-1 and Dectin-2, but emerging research suggests its involvement in cancer immunity. Dectin-3 functions as a PRR in myeloid cells, recognizing C. albicans and Cryptococcus species.119 While specific clinical trials targeting Dectin-3 are limited, studies indicate its role in modulating immune responses within the TME. In CRC, Dectin-3 has been implicated in modulating immune responses via its interaction with fungal pathogens.124 Mice lacking Dectin-3 are more susceptible to DSS-induced colitis, associated with overgrowth of C. tropicalis.124 This phenotype results from impaired macrophage-mediated fungal clearance and defective cytokine responses, which in turn hinder epithelial repair. Antifungal treatment ameliorated colitis in Dectin-3–deficient mice, underscoring the importance of fungal sensing in gut immune regulation.
Dectin-3 also influences the function of MDSCs, which are known to suppress anti-term immunity. Studies have demonstrated that in CRC, overgrowth of C. tropicalis promotes the accumulation of MDSCs, fostering an immunosuppressive TME and accelerating tumor progression.34 Experimental models have shown that fungal overgrowth exacerbates CRC in CARD9-deficient mice, whereas antifungal treatment with fluconazole reduces MDSC infiltration and suppresses tumor development.34 Research indicates that C. tropicalis promotes CRC progression by activating NLRP3 inflammasome signaling in MDSCs via Dectin-3.33 This activation depends on enhanced glycolysis driven by glycogen metabolism and requires both priming and activation signals: transcriptional upregulation of inflammasome components via JAK–STAT1 signaling, and mitochondrial reactive oxygen species (ROS) as the second signal. The inhibition of NLRP3 signaling suppressed tumor growth and MDSC infiltration in an AOM/DSS-induced CRC model. Elevated STAT1 and NLRP3 expression in MDSCs from human CRC samples supports the clinical relevance of the STAT1–NLRP3 axis as a potential therapeutic target.
In the context of HCC, Dectin-3 appears to exert a tumor-suppressive effect. A study utilizing Dectin-3 knockout mice revealed that the absence of this receptor led to enhanced tumor growth, accompanied by increased macrophage glycolysis and reduced infiltration of CD4⁺ and CD8⁺ T cells.125 Mechanistically, Dectin-3 appears to inhibit tumor progression by regulating glycolytic metabolism in TAMs, thereby modulating the immune environment within the tumor. In vitro studies showed that Dectin-3-deficient macrophages promoted proliferation and inhibited apoptosis of H22 hepatoma cells.125 Moreover, mice lacking Dectin-3 exhibit increased tumorigenesis in CRC and a higher burden of C. albicans upon chemical induction.126 This elevated fungal load triggers glycolysis in macrophages, leading to the secretion of IL-7. IL-7, in turn, induces IL-22 production in group 3 innate lymphoid cells (ILC3s) via the aryl hydrocarbon receptor and STAT3 pathways.126 This cascade promotes tumorigenesis, highlighting the critical role of Dectin-3 in regulating immune responses to fungal components in the gut. C. albicans is also the primary driver of human antifungal Th17 responses, with Th17 cells targeting other fungi arising through cross-reactivity.38 Intestinal inflammation promotes expansion of both C. albicans-specific and cross-reactive Th17 cells.
The dualistic nature of Dectin-3's role in cancer underscores the importance of context when considering therapeutic strategies. Targeting Dectin-3 or its downstream signaling pathways could offer novel approaches to modulate the immune response in cancer. However, given its varying effects depending on the tumor type and microenvironment, personalized approaches would be essential.
Mincle – a checkpoint of myeloid-driven immunosuppression and a target for macrophage repolarization
Mincle (macrophage-inducible C-type lectin, CLEC4E) is a PRR that recognizes various PAMPs, including α-mannan, SAP130, and trehalose-6,6′-dimycolate (TDM).115,127,128 Mincle regulates cytokine production in response to Candida species by activating the SYK–CARD9 signaling pathway and protein kinase Cδ (PKC-δ), playing a key role in the clearance of C. tropicalis during systemic infection.129,130 While its expression is relatively low in myeloid cells, Mincle has been implicated in modulating the TME and influencing cancer progression.115 Moreover, its expression in myeloid cells is inducible and regulated by other CLRs, particularly Dectin-3.116,131 In lung cancer, Mincle is predominantly expressed on M2-polarized TAMs, where it promotes an immunosuppressive phenotype via the SYK–NF-κB signaling axis.132 Elevated Mincle expression correlates with poor prognosis, reduced T cell infiltration, and enhanced tumor progression.132 Mincle also plays a critical role in PDA, where its interaction with SAP130 released during necroptosis activates RIP1/RIP3-dependent signaling, suppresses immunogenic myeloid cell recruitment, and facilitates tumor immune evasion.133 Deletion of Mincle in murine PDA models leads to decreased tumor growth and a more pro-inflammatory TME, characterized by increased CD8⁺ T cell infiltration. Notably, Mincle signaling can have context-dependent effects. For example, Trehalose dibehenate (TDB), a synthetic Mincle ligand, has been shown to promote TAM repolarization toward a pro-inflammatory M1-like phenotype through GM-CSF–dependent pathways, enhancing anti-tumor immunity in preclinical models.134 These findings indicate that Mincle not only senses fungal and necrotic cell ligands but also shapes the immune landscape in tumors. Its dual role – as a promoter of immunosuppression or a potential target for macrophage reprogramming – makes it a promising candidate for novel immunotherapeutic strategies.135,136
Associations between immune checkpoint molecules and mycobiota
Increasing evidence suggests that the mycobiome plays a significant role in shaping immune responses to ICIs, although this area remains less explored compared to bacterial influences. The mycobiome may influence therapy outcomes both directly, through interactions with the host immune system, and indirectly by modulating bacterial community structure and function. The level of key immune checkpoint molecules, including PD-1, PD-L1, and CTLA-4, reflects the immunological landscape of the TME and is a central determinant of the efficacy of ICB therapies. Growing evidence suggests that tumor-associated fungi and antifungal immunity contribute to modulating these checkpoint pathways, particularly through CLRs. An overview of interactions between fungal components, CLR signaling, and immune checkpoint molecules is summarized in Table 2.
Table 2.
Interactions between fungal components, CLR signaling, and immune checkpoint molecules.
| Immune Checkpoint Molecule | Fungal Component/Organism/Ligand | CLR Involved | Immune Cell Type(s) | Mechanism of Action/Outcome | Implications for ICB Therapy | Model | References |
|---|---|---|---|---|---|---|---|
| PD-L1 | β-glucan (from C. albicans) | Dectin-1 | Neutrophils | nuclear translocation of PD-L1, secretion of CXCL1/2 → impaired migration on neutrophils, immunosuppression | PD-L1 blockade enhances neutrophils activity | mouse + human + fungi | [137] |
| PD-L1, others | Curdlan | Dectin-1 | Neutrophils, CD4⁺ T cells | ROS-dependent suppression of T cells; SYK-CARD9–mediated PD-L1 induction (autoimmune setting) | - | mouse | [138] |
| PD-L1 | Curdlan | Dectin-1 | MDSCs | ↓ PD-L1 in MDSCs (anti-tumor) | Modulation of Dectin-1 may enhance T cell responses while limiting suppressive MDSCs | human | [139] |
| PD-L1 | A. fumigatus | CLR (unspecified) | DCs | activation of Wnt/β-catenin pathway → ↑ PD-L1, ↑ Tregs → immunosuppression | Potential resistance mechanism; targetable via CLR pathway inhibitors | human + fungi | [140] |
| PD-L1 | β-glucan (WGP) | Dectin-1 | Macrophages, DCs, MDSCs, T cells (indirect) | ↑ infiltration of DCs and macrophages, ↓ frequency of MDSCs, ↓ proliferation of CD4+ T cells and Tregs in the tumor, ↓ PD-L1 in macrophages, ↑ PD-L1 in DCs, | Combination therapy (β-glucan + anti-PD-1/PD-L1) improves ICB responsiveness | mouse + tumor cell lines | [141] |
| PD-L1 | P. brasiliensis yeasts | Dectin-1, TLR2, TLR4 | M-MDSCs, PMN-MDSCs | Dectin-1 deficiency → ↓ PD-L1⁺ M-MDSCs accumulation in vivo → altered trafficking or survival of MDSCs + ↓ IL-10, ↓ nitrotyrosine + ↓ suppression of CD4+ and CD8+ T lymphocytes TLR2/TLR4 deficiency → ↑ PD-L1⁺ PMN-MDSCs + ↓ IL-10+ M-MDSCs + nitrotyrosine + ↓ CD4⁺ suppression |
PRRs contribute to the suppressive activity of MDSCs by inducing the expression of immunosuppressive molecules; Dectin-1 influences immune cell composition rather than direct PD-L1 expression | mouse + fungi | [142] |
| PD-L1 | S. cerevisiae, Candida spp. | Not CLR-dependent | Macrophages | PD-L1 binds fungal Rpl20b in phagosomes → IL-10 production → immune suppression | Highlights non-CLR mechanisms by which fungi modulate checkpoint activity | cell lines + primary cells | [143] |
| CTLA-4 | — | Dectin-1 | Tregs | Dectin-1 promotes Treg differentiation (αβ/γδ) even without TGF-β; deficiency worsens CTLA-4 haploinsufficiency phenotype | Dectin-1 acts as a modifier gene; CLR variants may influence ICB response or toxicity | mouse + human | [150] |
| CTLA-4/Costim. molecules | Curdlan | Dectin-1, TLR4 | DCs | co-stimulation → ↑ MHC-I/II, CD40, CD80, CD86 → improved antigen presentation and T cell priming; ↑ IL-12, IL-1β, TNF-α, IFN-β | Mycobiota may enhance efficacy of CTLA-4 blockade via costimulatory molecule induction | mouse | [151] |
Fungal recognition rewires PD-L1–mediated suppression
Recent studies have highlighted the nuanced role of Dectin-1 signaling in regulating PD-L1 expression across various myeloid cell types, with implications for immune modulation in both infectious and tumor contexts. Activation of Dectin-1 by β-glucans, especially those derived from C. albicans, has been shown to upregulate PD-L1 in neutrophils by promoting nuclear translocation of PD-L1 and the subsequent secretion of CXCL1 and CXCL2.137 This mechanism results in impaired neutrophil migration and enhanced accumulation in the bone marrow, contributing to a localized immunosuppressive phenotype.137 Neutrophil-specific PD-L1 deficiency or its pharmacological blockade enhances neutrophil release into circulation, improving antifungal immunity. Thus, the Dectin-1/PD-L1 axis acts as a negative regulator of antifungal responses and represents a potential therapeutic target. A similar immunoregulatory pathway has been observed in autoimmune settings, such as experimental autoimmune encephalomyelitis, where Dectin-1 signaling suppresses CD4⁺ T cell responses via a ROS-dependent manner, followed by PD-L1 and other immune checkpoint induction via SYK-CARD9 signaling.138
Mashhouri et al. reported co-localization of Dectin-1 and PD-L1 in tumor-infiltrating myeloid cells in melanoma.113 Stimulation with curdlan (a Dectin-1 ligand) increased the production of pro-inflammatory cytokines and immune cell infiltration, leading to tumor growth suppression. Notably, curdlan-mediated Dectin-1 activation reduced PD-L1 levels in MDSCs, indicating a potential anti-tumor role of Dectin-1.139 On the other hand, Karnam et al. demonstrated that A. fumigatus, via CLR signaling, activates the Wnt/β-catenin pathway in DCs, leading to PD-L1 upregulation and enhanced Treg-mediated immunosuppression.140 These findings expand the functional axis of fungal–CLR–PD-L1 regulation beyond neutrophils and macrophages to broader immune cell networks within the TME. The differential regulation of PD-L1 by Dectin-1 in MDSCs and DCs underscores the importance of context in immune modulation. Understanding these nuances is crucial for developing targeted therapies that can modulate the TME effectively. For example, strategies that inhibit Dectin-1 signaling in MDSCs may reduce their immunosuppressive functions, while promoting Dectin-1 activation in DCs could enhance their ability to stimulate anti-tumor T cell responses.
In line with this, a 2024 preclinical and clinical study demonstrated that whole glucan particle (WGP) β-glucan enhances the efficacy of PD-1/PD-L1 ICB therapy.141 In mouse tumor models, co-administration of β-glucan and anti-PD-1/PD-L1 antibodies resulted in increased recruitment of immune effector cells, improved regulation of T cell activation versus tolerance, and delayed tumor progression. Importantly, this combination also prolonged progression-free survival in cancer patients who had previously developed resistance to PD-1/PD-L1 therapy, suggesting that β-glucan can reverse immune escape by reprogramming innate and adaptive responses. These findings position β-glucan as a promising immune adjuvant that modulates checkpoint responsiveness, potentially through its impact on Dectin-1–mediated PD-L1 regulation.
A 2024 study by Kaminski et al. further investigated the role of Dectin-1, TLR2, and TLR4 in PD-L1 expression in regulating the production of immunosuppressive molecules by MDSCs during pulmonary fungal infection.142 Although Dectin-1 deficiency did not significantly change PD-L1 levels on MDSCs in vitro, it led to a reduction of PD-L1⁺ monocytic MDSCs (M-MDSCs) in vivo, suggesting Dectin-1 may influence the survival or trafficking of PD-L1-expressing M-MDSCs rather than PD-L1 direct expression. Dectin-1, TLR2, and TLR4 also reduced the expression of IL-10 and nitrotyrosine.142 Additionally, Li et al.143 revealed a novel mechanism whereby PD-L1 contributes to innate sensing of fungal pathogens: PD-L1 is selectively enriched in macrophage phagosomes containing S. cerevisiae and Candida spp. and directly binds the fungal ribosomal protein Rpl20b, leading to the anti-inflammatory cytokine IL-10 production independently of canonical PRRs signaling. Both genetic depletion of Rpl20b in yeast and functional blockade of PD-L1 on host cells resulted in diminished IL-10 responses, confirming the specificity and functional relevance of this interaction. This adds another layer to the immunoregulatory role of PD-L1 in the context of fungal exposure and raises the possibility that fungal components of the mycobiota could impact responsiveness to ICIs.
Altogether, these findings support the notion that β-glucans act as immune modulators that enhance PD-1/PD-L1 blockade primarily through trained immunity (TI)-driven myeloid reprogramming and context-dependent PD-L1 ‘rewiring’ on myeloid cells.
β-Glucans engaging Dectin-1 (SYK–CARD9 axis) induce epigenetic and metabolic reprogramming of monocytes, macrophages, and DCs, shifting them toward a pro-inflammatory, antigen-presenting phenotype that improves T cell priming and effector function.137,138,142 Clinically, this manifests as increased effector infiltration, higher inflammatory cytokine competence upon secondary challenge, and improved responsiveness to ICIs.141 TI also helps explain durable benefits observed after short β-glucan exposure. As for the ‘rewiring’ mechanism, CLR engagement also modulates PD-L1 at the level of expression, localization (including nuclear pools), and cell-subset distribution.137,139,142,143 In tumors, β-glucans can reduce PD-L1 on suppressive MDSCs while enhancing pro-inflammatory outputs from DCs;139,141 in other contexts (e.g., neutrophils or DCs under certain stimuli), PD-L1 can be transiently upregulated.137,138,143 The net tumor-context effect is predicted to lower myeloid-derived suppression and heighten ICI sensitivity.
It is worth to mention that CR3/complement–mediated mechanisms may also contribute to enhanced checkpoint therapy, although likely as a secondary effect. β-glucans can allosterically activate CR3 (CD11b/CD18) and shape complement biology, which can influence antitumor immunity144,145. However, given the limited Fc effector engagement of PD-1/PD-L1 antibodies compared to some other antagonistic monoclonal antibodies (mABs),146 these mechanisms are likely supportive rather than primary, potentially contributing through CR3-dependent cytotoxicity or iC3b-opsonization in specific contexts.
Future trials could evaluate these mechanisms using complementary approaches. For TI, ex vivo functional assays of patient monocytes, macrophages, and DCs, combined with epigenetic, transcriptional, and metabolic profiling, can reveal myeloid reprogramming. Context-dependent PD-L1 rewiring can be assessed by measuring PD-L1 expression and localization across myeloid subsets, functional co-cultures with T cells, and spatial mapping within tumors. Complement/CR3-mediated contributions can be monitored via CR3 activation and iC3b opsonization assays, together with serum complement profiling. Integrated, longitudinal biomarker analyses, combining single-cell multi-omics, functional readouts, and soluble mediators, will allow correlations between mechanistic changes and clinical outcomes such as immune response, tumor progression, and therapy durability.
CTLA-4 and other ICIs
In addition to PD-1/PD-L1, CTLA-4, a critical checkpoint molecule expressed primarily on Tregs, has also been implicated in fungal immune interactions.147 CTLA-4 competes with CD28 for binding to CD80/CD86, dampening T cell activation and promoting immune tolerance.148,149 Recent studies have highlighted the multifaceted role of Dectin-1 in immune regulation, particularly in the context of CTLA-4 haploinsufficiency and AML. Turnbull et al. identified a patient with immune dysregulation, autoimmunity, and lymphoproliferation who carried a maternally inherited pathogenic CTLA4 variant and a paternally inherited rare loss-of-function missense variant in CLEC7A.150 This CLEC7A variant led to impaired Dectin-1 dimerization and surface expression. Functional analyses demonstrated that Dectin-1 stimulation promotes Tregs differentiation from naïve αβ and γδ T cells, even in the absence of transforming growth factor beta (TGF-β). Partial Dectin-1 deficiency exacerbated the Treg defect conferred by CTLA-4 haploinsufficiency, suggesting that Dectin-1 acts as a modifier gene, influencing the expressivity of CTLA4 variants and playing a role in maintaining immune homeostasis and tolerance.150 Moreover, recent studies have revealed that fungal components can upregulate a broader array of checkpoint and costimulatory molecules, including MHC-I, MHC-II, CD40, CD80, and CD86, particularly via co-engagement of Dectin-1 and TLR4 signaling in DCs.151 This upregulation supports enhanced antigen presentation and T cell priming, potentially boosting the efficacy of ICB therapies. Altogether, these data point to a fungi–CLR–checkpoint axis as a critical regulatory pathway in cancer immunology. Through their interaction with innate immune receptors, tumor-associated fungi can either promote immune tolerance or stimulate anti-tumor immunity depending on the context and immune cell type. These findings open new avenues for integrating mycobiome profiling and fungal-targeted therapies with checkpoint inhibition strategies in cancer treatment.
The anti-tumor activity of YBG β-glucan
β-glucan-induced reprogramming of innate immunity
Yeast-derived beta-glucan (YBG) enhances anti-tumor immunity by training innate immune cells, including macrophages, neutrophils, and DCs. Particularly, whole β-glucan particles (WGPs) promote macrophage reprogramming and antimetastatic activity in murine models of cancer metastasis.152 In lung metastasis models, WGPs induced a TI phenotype in interstitial macrophages, reducing metastatic burden and prolonging survival. This effect is driven by the metabolite sphingosine-1-phosphate (S1P), with its inhibition abrogating protection, highlighting the importance of the sphingolipid–mitochondrial axis in sustaining TI.152 WGPs also induce TI in human monocytes by activating multiple innate immune receptors (Dectin-1/complement receptor 3 (CR3), TLR4, and macrophage MR) and downstream signaling pathways, including Raf-1, SYK, and PI3K.153 This synergistic activation enhanced the secondary immune response to unrelated stimuli. In murine melanoma and bladder carcinoma models, β-glucan pre-treatment significantly reduced tumor growth.153 In PDA, YBG accumulates in the pancreas, triggering a CCR2-dependent recruitment of monocytes and macrophages exhibiting TI features.154 These cells become highly cytotoxic upon encountering tumor cells or tumor-derived signals. In murine models of PDA, YBG treatment significantly reduces tumor burden and extends survival, with even greater efficacy observed when combined with immunotherapeutic agents. Beyond macrophages, fungal-derived β-glucans also induce durable anti-tumor effects through transcriptional and epigenetic reprogramming of granulopoiesis and neutrophil function. This trained state skews neutrophils toward a tumoricidal phenotype, contributing to improved cancer control.155 Moreover, β-glucan-trained neutrophils from donor animals can be adoptively transferred to naïve recipients, where they inhibit tumor growth via a ROS-dependent mechanism. Furthermore, the tumor-suppressive programming induced by β-glucans during granulopoiesis can be transferred through bone marrow transplantation (BMT), emphasizing the systemic and durable nature of this reprogramming.155 These findings underscore the broad potential of β-glucans as immune-training agents that rewire the innate immune system for enhanced anti-tumor responses. A mechanistic overview of β-glucan-mediated immune modulation in cancer models is presented in Table 3.
Table 3.
Mechanistic overview of β-glucan-mediated immune modulation in cancer models.
| Mechanism | Effect | Details | Models/Systems | Model | References |
|---|---|---|---|---|---|
| TI | Anti-tumoral | Promotion of macrophage reprogramming; reduced metastasis, prolonged survival, S1P required for TI | Lung metastasis models | mouse | [152] |
| TI | Anti-tumoral | Activation of Dectin-1/CR3, TLR4, macrophage MR; and Raf-1, SYK, PI3K pathways; reduced tumor growth | Melanoma and bladder cell carcinoma | mouse + human primary monocytes + fungi | [153] |
| TI | Anti-tumoral | Recruitment of CCR2-dependent monocytes/macrophages with trained features, reduced tumor growth, prolonged survival | PDA | mouse + cell lines | [154] |
| TI | Anti-tumoral | Transcriptional and epigenetic reprogramming of granulopoiesis and neutrophil function → skewing neutrophils to a tumoricidal phenotype; β-glucan-trained neutrophils suppress tumors in naïve mice via ROS; β-glucan-induced anti-tumor programming transferable via BMT | Melanoma | mouse + cell lines | [155] |
| Macrophage reprogramming | Anti-tumoral | Conversion of naïve M0 macrophages + reprogramming M2-polarized macrophages and TAMs → tumoricidal M1 phenotype | Melanoma, lymphoma | mouse + cell lines | [156] |
| Macrophage reprogramming | Anti-tumoral | YBG–Ferumoxytol hybrid activates macrophages via Dectin-1 → strong M1 polarization via SYK/MAPK → ↑ TNF-α, IL-6, ROS → tumor cell apoptosis and arrest | Melanoma | mouse | [157] |
| Initiation of adaptive immune responses | Anti-tumoral | YBG-induced autophagy as a key mechanism in β-glucan-induced DCs activation | Melanoma, bladder cell carcinoma | mouse + tumor cell lines | [141] |
| DCs modulation | Anti-tumoral | Reversion of DCs’ dysfunction induced by tumor-derived factors → activation of CTLs + promotion of Th1-mediated immune responses | Lung cancer | mouse + cell lines | [158] |
| MDSCs modulation | Anti-tumoral | Induction of PMN-MDSC apoptosis → promotion of M-MDSC maturation into APCs → restoration of CD4+/CD8+ T cell activation | Lung cancer, mammary-cell carcinoma | mouse + human | [159] |
| Activation of monocytes | Anti-tumoral | Activation of monocytes → direct cytotoxicity against tumor cells + ↑ TNF-α, M-CSF, CCL2 | lung metastatic melanoma | mouse + cell lines | [160] |
| Complement activation (CR3/iC3b axis) | Anti-tumoral | Oral YBG primes neutrophils via CR3 to kill iC3b-opsonized tumor cells; intravenous YBG forms immune complexes activating classical complement pathway→ augmentation of ADCP, ↑ ROS | Lymphoma, lung cancer | - | [144,145] |
| ROS-mediated cytotoxicity | Anti-tumoral | Increase of ROS in tumor cells → caspase 3/9 activation + cancer cell apoptosis | HeLa cervical cells | cell lines | [164] |
| ROS-mediated cytotoxicity | Anti-tumoral | Inhibition of autophagy by increasing lysosomal pH and blocking cathepsins B and D → mitochondrial dysfunction + ↑ ROS → caspase-8 activation + truncated BID translocation → cancer cells to apoptosis | mouse xenograft models, HCC | mouse + cell lines | [165] |
| Complement overactivation (MBL–C3a axis) | Pro-tumoral | Activation of lectin complement pathway; MBL/C3a signaling → PDA growth; deletion of MBL/C3/C3aR suppresses tumors | PDA models | mouse + cell lines | [56] |
Activation of immune signaling pathways to boost anti-tumor responses by YBG
YBG modulates both innate and adaptive immunity through diverse signaling cascades. One of the key mechanisms involves PRRs such as Dectin-1, initiating NF-κB, MAPK, and SYK pathways, leading to the production of pro-inflammatory cytokines and chemokines that amplify immune surveillance and cytotoxic responses. Oral administration of WGPs has been shown to delay tumor progression in various preclinical models by enhancing the functional activity of macrophages and NK cells.156 Notably, WGPs support macrophage polarization toward a tumoricidal M1 phenotype, not only by converting naïve M0 macrophages but also by reprogramming M2-polarized macrophages and TAMs, which are often co-opted by tumors to support immune evasion. In an innovative therapeutic approach, YBG has been conjugated with Ferumoxytol, an FDA-approved iron oxide nanoparticle, to form a nanocomposite with enhanced bioactivity.157 This hybrid material activates macrophages through Dectin-1 and induces robust M1 polarization via SYK and MAPK signaling, resulting in elevated TNF-α, IL-6, and ROS production. This pro-inflammatory shift facilitates tumor cell apoptosis and cell cycle arrest, as shown in models of melanoma and CRC.157 In addition to shaping macrophage and NK cell responses, YBG influences other critical aspects of tumor immunology. Ding et al showed that YBG-induced autophagy is a key mechanism in β-glucan-induced DCs activation, fostering their capacity to initiate effective adaptive immune responses.141 Moreover, YBG reverses DCs’ dysfunction induced by tumor-derived factors, restoring their ability to prime cytotoxic T lymphocytes (CTLs) and support Th1-mediated immune responses.158 This is particularly relevant given the role of Th1 cytokines like IFN-γ in promoting durable anti-tumor immunity. Another significant function of YBG is its impact on MDSCs, which play a pivotal role in tumor-induced immune suppression. YBG induces apoptosis in polymorphonuclear MDSCs (PMN-MDSCs), which suppress anti-tumor response and facilitates the differentiation of M-MDSCs into mature antigen-presenting cells (APCs) capable of activating CD4+ and CD8+ T cells.159 By disrupting MDSC-mediated immunosuppression, YBG restores the effectiveness of both innate and adaptive immune responses, further enhancing its therapeutic value. Alexander et al., using a lung metastatic melanoma model, demonstrated that the anti-tumor effect of YGP can occur independently of adaptive immunity, instead relying on inflammatory monocytes.160 These YGP-activated monocytes exert direct cytotoxicity against tumor cells in vitro, and systemic YGP treatment increases the levels of inflammatory mediators such as TNF-α, M-CSF, and CCL2 in the lungs. Importantly, combination strategies involving YBG and ICIs or other immunotherapies have shown synergistic effects in preclinical models, indicating the potential of YBG as an adjunct agent in cancer immunotherapy.161-163 Collectively, these findings underscore the multifaceted role of YBG as a powerful modulator of the tumor immune microenvironment and a promising candidate for integrative cancer treatment strategies.
YBG-induced complement activation and the role of ROS
Upon oral administration, fluorescein-labeled YBG is predominantly taken up by intestinal macrophages, which subsequently traffic the compound to central immune sites, including the spleen, lymph nodes, and bone marrow. Within the bone marrow microenvironment, degradation products of YBG interact specifically with CR3 expressed on neutrophils. This interaction initiates a signaling cascade that primes neutrophils to identify and eliminate tumor cells opsonized with the complement fragment iC3b, enhancing innate tumor surveillance and clearance.145 Conversely, intravenous delivery of YBG leads to its immediate association with circulating endogenous antibodies, forming immune complexes that activate the classical complement pathway. This activation results in the deposition of iC3b on tumor cells, which in turn facilitates engagement of immune effector cells via CR3 and the Fc region of the immunoglobulin G receptor IIa (FcγRIIA). The synergistic signaling through these receptors intensifies ROS production by neutrophils and augments macrophage-mediated antibody-dependent cellular phagocytosis (ADCP), collectively reinforcing anti-tumor immunity.144
In addition to its immunostimulatory effects, YBG-induced ROS play a critical role in mediating direct cytotoxicity toward tumor cells. Studies on β-glucan derived from Pleurotus ostreatus demonstrate that YBG elevates intracellular ROS levels, triggering apoptotic pathways in HeLa cervical cells.164 This oxidative stress is associated with marked nuclear condensation and fragmentation, hallmarks of apoptosis, and is accompanied by the activation of caspases 3 and 9 – key executioners of programmed cell death. In parallel, YBG exerts additional anti-tumor effects by interfering with cancer cell metabolic adaptation. Water-soluble yeast β-D-glucan (WSG) has been shown to function as a novel autophagy inhibitor, a process often hijacked by tumor cells to sustain growth under stress conditions.165 By increasing lysosomal pH and inhibiting the activity of cathepsins B and D, WSG disrupts autophagic degradation, leading to the accumulation of damaged mitochondria. This mitochondrial dysfunction results in a secondary surge in ROS, further amplifying oxidative stress. Elevated ROS levels not only directly impair tumor cell viability but also sensitize cancer cells to apoptosis by activating caspase-8 and mitochondrial translocation of truncated BH3 Interacting Domain Death Agonist (tBID), especially under nutrient-deprived conditions. In vivo experiments further support these observations as WSG administration significantly inhibited tumor growth without causing apparent toxicity in mouse xenograft models and chemically induced HCC (DEN/CCl₄) models.165 Thus, beyond enhancing immune-mediated tumor clearance, YBG exerts a dual anti-tumor effect by promoting ROS-dependent apoptosis in malignant cells, further underscoring its potential as a multifaceted therapeutic agent.
Beyond these mechanisms, recent studies have highlighted a pathogenic role of fungal-induced complement activation in PDA. Specifically, YBG can activate the lectin pathway of the complement system via binding to mannose-binding lectin (MBL). Ligation of MBL was shown to be essential for PDA progression: deletion of Mbl (encoding MBL) or C3 in the extratumoral compartment, as well as knockdown of the C3a receptor (C3aR) in tumor cells, significantly suppressed tumor growth. These findings suggest that while YBG may harness complement signaling for therapeutic benefit, uncontrolled or context-specific fungal complement activation, particularly via the MBL–C3a axis, can paradoxically promote tumor development.56
The fungal metabolome as a driver of tumor–immune interactions
Fungal metabolites are increasingly recognized as essential modulators of the TME, capable of influencing cancer progression and immune responses through diverse biochemical pathways. Among the most well-known of these metabolites are mycotoxins – toxic secondary metabolites produced by various fungal species – which have been extensively studied for their carcinogenic, genotoxic, and immunomodulatory properties. For instance, Candida species can produce acetaldehyde, a reactive metabolite implicated in DNA damage and local inflammation,166,167 as well as candidalysin, a cytolytic peptide toxin that disrupts epithelial barriers and activates immune signaling pathways.168,169 Other fungi, such as Aspergillus spp., may produce aflatoxins, well-known mycotoxins with genotoxic and carcinogenic potential.170-174 Apart from well known carcinogenic mycotoxins, such as candidalysin, aflatoxins, ochratoxins,175-177 fumonisins,178-180 and patulin181,182 that can damage DNA or induce chronic inflammation and immune dysregulation, fungi produce a variety of other bioactive metabolites that can modulate the TME and immune cell function, thereby influencing the efficacy of ICIs. These metabolic interactions may contribute to either immunosuppressive or immunostimulatory microenvironments, thereby affecting tumor immune surveillance and responsiveness to ICIs. For instance, fungi produce eicosanoids, including PGE2, prostaglandin D2 (PDE2), and F2α (PDF2α),183 which can influence both innate and adaptive immune responses.184-186 PGE2 plays a key role in tumor immune evasion by suppressing innate and adaptive immune responses, including T cell activity, thereby reducing the effectiveness of immunotherapies.185,187,188 Targeting the PGE2–EP4 signaling axis can help restore anti-tumor immunity. The selective EP4 antagonist HTL0039732 reverses PGE2-driven M2-like macrophage polarization and enhances immune responses, and preclinical studies demonstrate its synergy with PD-1/PD-L1 blockade.189 PGD2 creates an autocrine loop that maintains an immunosuppressive, protumoral phenotype. Through DP1/DP2 receptors, PGD2 inhibits CD8+ T-cell activity; blocking PGD2 activity enhances T-cell responses and sensitizes tumors to anti-PD-1 therapy.190
On the other hand, fungal-derived metabolites such as chromone,191-193 ketones (e.g., Brocaeloid D from Microdochium majus,194 depsidones and diaryl ethers from potato fungus Boeremia exigua195), sesquiterpenoids,196 and cytochalasins197 have demonstrated significant anticancer properties through mechanisms involving apoptosis induction, cell cycle arrest, and immune lmodulation. In addition to these compounds, several fungal species have been identified as sources of potent anti-tumor agents with potential therapeutic applications. He et al. reported that endophytic fungal secondary metabolites from Ginkgo biloba leaves significantly reduced tumor formation in HeLa-implanted mice by promoting apoptosis and inhibiting proliferation of HeLa cells.198 Similarly, extracellular polysaccharides from Schizophyllum radiatum showed anti-tumor activity.199 Schisandra polysaccharide, a product of Schisandra chinensis fermentation, exhibited strong immunomodulatory effects in vivo and enhanced anti-tumor properties.200,201 FIP-nha, a 114-amino-acid protein isolated from Nectria haematococca, suppressed lung cancer cell growth in vivo by inhibiting the PI3K/Akt pathway.202 Likewise, colletofragarone A2, derived from Colletotrichum spp., reduced mutant p53 levels and inhibited tumor progression.203 Furthermore, a novel L-asparaginase produced by Yarrowia lipolytica was shown to suppress the proliferation and migration of both lung and breast cancer cells.204
Oxylipins, a class of oxygenated fatty acid derivatives, another fungal metabolites, are known to influence macrophage polarization and PD‑L1 expression in the TME. For instance, in glioma, oxylipin 5-hydroxyeicosatetraenoic acid (5-HETE) promoted M2-like macrophage polarization and PD‑L1 upregulation via the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, leading to immunosuppressive TME.205 In fungal infection models, fungal oxylipins have been reported to modulate macrophage polarization and downstream immune checkpoint signaling.206,207 Other fungal metabolites, including fermentation products such as ethanol, organic acids, and fungal immunomodulatory proteins, profoundly affect APC phenotype and function. For example, fungal immunomodulatory proteins like FIP‑vvo enhance DCs maturation (elevated MHC II, IL‑12, IL‑2, TNF‑α), improving antigen presentation and T cell priming.208 These findings underscore the growing recognition of the fungal metabolome as a source of diverse compounds capable of modulating cancer biology and immune responses. As such, exploring the role of fungal metabolites in shaping the immunological landscape of cancer may reveal new avenues for enhancing the efficacy of immunotherapies. Immunomodulatory and oncogenic effects of fungal metabolites and mycotoxins are summarized in Table 4 and Figure 3.
Table 4.
Immunomodulatory and oncogenic effects of fungal metabolites and mycotoxins.
| Effect | Representative Metabolites/Compounds | Mechanism | Model | References |
|---|---|---|---|---|
| Carcinogenic metabolite | acetaldehyde | epithelial dysplasia, DNA adduct formation, inhibition of DNA repair, oxidative stress, local inflammation, promotion of mutagenesis and carcinogenesis | human + fungi | [166,167] |
| Carcinogenic mycotoxins | candidalysin | epithelial barrier disruption, induction of proinflammatory cytokines, activation of MAPK/AP-1 pathway, promotion of tissue damage and inflammation-mediated carcinogenesis | cell lines + organoid | [168,169] |
| Carcinogenic mycotoxins | aflatoxins | genotoxicity, immunosuppression, DNA adduct formation, oxidative stress, p53 mutation, general tumor promotion | - | [170–172] |
| Carcinogenic mycotoxins | ochratoxins | genotoxicity, nephrotoxicity, neurotoxicity, embryotoxicity, teratogenicity, carcinogenicity, inhibition of protein synthesis, oxidative damage, DNA strand breaks | - | [175–177] |
| Carcinogenic mycotoxins | fumonisins | hepatotoxicity, neurotoxicity, carcinogenicity, disruption of sphingolipid metabolism (inhibits ceramide synthase) | pig | [178–180] |
| Carcinogenic mycotoxins | patulin | immunotoxicity, genotoxicity, ROS generation, mitochondrial damage, DNA damage | mouse + cell lines | [181,182] |
| Immunosuppression | PGE2, PGD2, PDF2α | inhibits T cells, promotes M2 macrophages | mouse + human | [183–190] |
| Immunosuppression | PGD2 | inhibits CD8+ T-cell activity through DP1/DP2 receptors; blocking PGD2 activity enhances T-cell responses and synergizes with anti-PD-1 therapy | mouse + cell lines | [190] |
| Anti-cancer secondary metabolites | Chromones, ketones, sesquiterpenoids, cytochalasins | promotion of apoptosis, cell cycle arrest, immune modulation | fungi | [191–197] |
| Anti-cancer secondary metabolites | endophytic fungal secondary metabolites from G. biloba leaves | promotion of apoptosis and inhibition of the proliferation of HeLa cells | mouse | [198] |
| Anti-tumor polysaccharides | S. radiatum extracellular polysaccharides, Schisandra polysaccharide | immune activation, enhances immune responses | mouse + fungi | [199–201] |
| Anti-tumor fungal proteins | FIP-nha (from N. haematococca) | suppresses the PI3K/Akt pathway in lung cancer model | mouse | [202] |
| Anti-tumor | Colletofragarone A2 (from Colletotrichum spp.) | p53-dependent tumor suppression | fungi | [203] |
| Anti-tumor enzymes | L-asparaginase (from Y. lipolytica) | suppresses the proliferation of lung and breast cancer cells | cell lines | [204] |
| Immunosuppression | Oxylipins (e.g., 5-HETE) | promotes PD-L1 upregulation + M2-like macrophage polarization in glioma via the Nrf2 pathway | human + mouse | [205–207] |
| Anti-tumor protein | FIP-vvo | ↑ DC maturation (↑ MHC II, IL-12, TNF-α) → improved antigen presentation and T cell priming | mouse | [208] |
Figure 3.
Dual roles of fungal metabolites in cancer highlighting both pro-tumor (immunosuppressive) and anti-tumor (immunostimulatory) mechanisms. Fungi contribute to both pro-tumor (red, left) and anti-tumor (green, right) mechanisms. Pro-tumor effects include DNA damage, barrier disruption, immune suppression, resistance to ICIs, and tumor-promoting interactions with bacteria. In contrast, fungal-derived metabolites, proteins, and polysaccharides can exert anticancer activity by inducing apoptosis, inhibiting proliferation, modulating immunity, and enhancing anti-tumor responses, including through co-occurrence with SCFA-producing bacteria.
Conclusions
The gut mycobiome is increasingly recognized as an important, though often neglected, component of the tumor–microbiota–host axis. While bacterial communities have long been the focus of immuno-oncology research, recent findings suggest that fungi also play a critical role in shaping immune responses and influencing cancer progression and treatment efficacy. The mycobiome does not act in isolation; it interacts intricately with bacterial consortia and the host immune system, contributing to a dynamic ecological and immunological environment that can either support or hinder anti-tumor immunity. These interactions are not merely theoretical; they carry real clinical implications. Fungal genera such as Candida and Malassezia have been linked to key immunomodulatory pathways, potentially affecting ICB. Mechanistically, fungi engage the host immune system through conserved cell wall components such as β-glucans, mannans, and chitin, which are recognized by PRRs including Dectin-1, TLRs, and NLRs. These interactions can shape innate and adaptive responses, influencing T cell polarization and cytokine production. In addition, fungal metabolism generates a range of bioactive secondary metabolites, which can further modulate the TME and immune tone. Such metabolites may enhance inflammation, promote immune suppression, or interfere with antigen presentation. As such, comprehensive immunotherapeutic strategies must begin to account for the fungal dimension of the microbiome to improve patient stratification, predict outcomes, and guide interventions. Integrating fungal data into clinical oncology and microbiome-based therapeutics will require a paradigm shift. A mechanistic and translational understanding of fungi–host–tumor interactions is essential to fully exploit the therapeutic potential of the microbiome.
Funding Statement
This work was carried out under contract no. POIR.04.01.02-00-0025/17-00 to AP, as the ‘Polish Microbiome Map,’ co-financed by the European Regional Development Fund under the Smart Growth Operational Programme, Submeasure 4.1.2. Regional research agendas. (Narodowe Centrum Badań i Rozwoju)
Disclosure of potential conflicts of interest
No potential conflicts of interest were disclosed.
Author contributions
NS and AP were responsible for the conceptualization, literature search, and drafting and revising the manuscript. PK and TZ provided substantial intellectual input and critical contributions to the manuscript.
Abbreviations
- ADCP
antibody-dependent cellular phagocytosis
- AML
acute myeloid leukemia
- APCs
antigen-presenting cells
- AUC
area under the curve
- BMT
bone marrow transplantation
- CARD9
caspase recruitment domain-containing protein 9
- CAR
chimeric antigen receptor
- CBM complex
CARD9–BCL10–MALT1 signaling complex
- CLRs
C-type lectin receptors
- CRC
colorectal cancer
- CRD
carbohydrate recognition domain
- CR3
complement receptor 3
- CTLA-4
cytotoxic T-lymphocyte antigen-4
- CTLs
cytotoxic T lymphocytes
- DCs
dendritic cells
- DC-SIGN
dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin
- ERK
extracellular signal-regulated kinase
- ERMAP
erythroid membrane-associated protein
- FcRγ
Fc receptor γ-chain
- FcγRIIA
Fc region of the immunoglobulin G receptor IIa
- FMT
fecal microbiota transplantation
- GOSs
galacto-oligosaccharides
- HCC
hepatocellular carcinoma
- ICAM3
intercellular adhesion molecule 3
- ICB
immune checkpoint blockade
- ICI/ICIs
immune checkpoint inhibitor(s)
- IFN-γ
interferon gamma
- IL
interleukin
- ILC3s
group 3 innate lymphoid cells
- IRF4
interferon regulatory factor 4
- iRAEs
immune-related adverse events
- ITAM
immunoreceptor tyrosine-based activation motif
- JAK
Janus kinase
- mABs
antagonistic monoclonal antibodies
- MAPK
mitogen-activated protein kinase
- MALT1
mucosa-associated lymphoid tissue lymphoma translocation protein 1
- MBL
mannose-binding lectin
- MDSCs
myeloid-derived suppressor cells
- Mincle
macrophage-inducible C-type lectin (CLEC4E)
- MMR
macrophage mannose receptor
- MR
mannose receptor
- M-MDSCs
monocytic MDSCs
- NIK
NF-κB-inducing kinase
- NK cells
natural killer cells
- NLRs
NOD-like receptors
- NSCLC
non-small cell lung cancer
- OSCC
oral squamous cell carcinoma
- PAMPs
pathogen-associated molecular patterns
- PDA
pancreatic ductal adenocarcinoma
- PD
programmed death
- PD-L1/PD-L2
programmed death ligand-1/ligand-2
- PDE2
prostaglandin D2
- PDF2α
prostaglandin F2α
- PGE₂
prostaglandin E₂
- PGF₂
prostaglandin F₂
- PKC-δ
protein kinase Cδ
- PMN-MDSCs
polymorphonuclear MDSCs
- PRRs
pattern recognition receptors
- RCC
renal cell carcinoma
- RIG-I/RLRs
retinoic acid-inducible gene I (RIG-I)-like receptors
- ROS
reactive oxygen species
- SAPs
secreted aspartyl proteases
- SCFA
short-chain fatty acid
- SYK
spleen tyrosine kinase
- S1P
sphingosine-1-phosphate
- TAMs
tumor-associated macrophages
- tBID
truncated BH3 Interacting Domain Death Agonist
- TCR
T cell receptor
- TDB
Trehalose dibehenate
- TDM
trehalose-6,6′-dimycolate
- TGF-β
transforming growth factor beta
- Th
T helper
- TI
trained immunity
- TLR
Toll-like receptor
- TME
tumor microenvironment
- TNF-α
tumor necrosis factor alpha
- WGPs
whole β-glucan particles
- WSG
Water-soluble yeast β-D-glucan
- YBG
yeast-derived β-glucan
- 5-HETE
5-hydroxyeicosatetraenoic acid
References
- 1.Tawbi H, Schadendorf D, Lipson E, Ascierto P, Matamala L, Castillo Gutiérrez E, Rutkowski P, Gogas H, Lao C, De Menezes J, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 2022;386:24–34. doi: 10.1056/NEJMoa2109970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Wolchok J, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob J, Cowey C, Lao C, Wagstaff J, Schadendorf D, Ferrucci P, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377:1345–1356. doi: 10.1056/NEJMoa1709684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Reck M, Rodríguez-Abreu D, Robinson A, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al. Pembrolizumab versus chemotherapy for PD-l1-positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774. [DOI] [PubMed] [Google Scholar]
- 4.Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, Domine M, Clingan P, Hochmair M, Powell S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378:2078–2092. doi: 10.1056/NEJMoa1801005. [DOI] [PubMed] [Google Scholar]
- 5.Motzer R, Tannir N, McDermott D, Arén Frontera O, Melichar B, Choueiri T, Plimack E, Barthélémy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378:1277–1290. doi: 10.1056/NEJMoa1712126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Rini B, Plimack E, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380:1116–1127. doi: 10.1056/NEJMoa1816714. [DOI] [PubMed] [Google Scholar]
- 7.June C, O’Connor R, Kawalekar O, Ghassemi S, Milone M. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–1365. doi: 10.1126/science.aar6711. [DOI] [PubMed] [Google Scholar]
- 8.Berraondo P, Sanmamed M, Ochoa M, Etxeberria I, Aznar M, Pérez-Gracia J, Rodríguez-Ruiz M, Ponz-Sarvise M, Castañón E, Melero I. Cytokines in clinical cancer immunotherapy. Br J Cancer. 2019;120:6–15. doi: 10.1038/s41416-018-0328-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Piper M, Kluger H, Ruppin E, Hu-Lieskovan S. Immune resistance mechanisms and the road to personalized immunotherapy. Am Soc Clin Oncol Educ Book Am Soc Clin Oncol Annu Meet. 2023. (43):e390290. doi: 10.1200/EDBK_390290. [DOI] [PubMed] [Google Scholar]
- 10.Conroy M, Naidoo J. Immune-related adverse events and the balancing act of immunotherapy. Nat Commun. 2022;13:392. doi: 10.1038/s41467-022-27960-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Routy B, Le Chatelier E, Derosa L, Duong C, Alou M, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti M, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–97. doi: 10.1126/science.aan3706. [DOI] [PubMed] [Google Scholar]
- 12.Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M, Luke J, Gajewski T. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–108. doi: 10.1126/science.aao3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gopalakrishnan V, Spencer C, Nezi L, Reuben A, Andrews M, Karpinets T, Prieto P, Vicente D, Hoffman K, Wei S, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103. doi: 10.1126/science.aan4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Baruch E, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:602–609. doi: 10.1126/science.abb5920. [DOI] [PubMed] [Google Scholar]
- 15.McQuade J, Daniel C, Helmink B, Wargo J. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol. 2019;20:e77–e91. doi: 10.1016/S1470-2045(18)30952-5. [DOI] [PubMed] [Google Scholar]
- 16.Zhao Y, Yi J, Xiang J, Jia W, Chen A, Chen L, Zheng L, Zhou W, Wu M, Yu Z, et al. Exploration of lung mycobiome in the patients with non-small-cell lung cancer. BMC Microbiol. 2023;23:81. doi: 10.1186/s12866-023-02790-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Narunsky-Haziza L, Sepich-Poore G, Livyatan I, Asraf O, Martino C, Nejman D, Gavert N, Stajich J, Amit G, González A, et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185:3789–3806. doi: 10.1016/j.cell.2022.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.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. doi: 10.3389/fmicb.2022.812771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Coker O, Nakatsu G, Dai R, Wu W, Wong S, Ng S, Chan F, Sung J, Yu J. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 2019;68:654–662. doi: 10.1136/gutjnl-2018-317178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Szóstak N, Figlerowicz M, Philips A. The emerging role of the gut mycobiome in liver diseases. Gut Microbes. 2023;15:2211922. doi: 10.1080/19490976.2023.2211922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Underhill D, Iliev I. The mycobiota: interactions between commensal fungi and the host immune system. Nat Rev Immunol. 2014;14:405–416. doi: 10.1038/nri3684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sachdeva G, Das A. Communication between immune system and mycobiota impacts health and disease. Proc Indian Natl Sci Acad. 2022;88:250–262. doi: 10.1007/s43538-022-00082-5. [DOI] [Google Scholar]
- 23.Iliev I. Mycobiota–host immune interactions in IBD: coming out of the shadows. Nat Rev Gastroenterol Hepatol. 2022;19:91–92. doi: 10.1038/s41575-021-00541-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Szóstak N, Handschuh L, Samelak-Czajka A, Tomela K, Schmidt M, Pruss L, Milanowska-Zabel K, Kozlowski P, Philips A. Host factors associated with gut mycobiome structure. mSystems. 2023;8(2):e0098622. doi: 10.1128/msystems.00986-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Szóstak N, Szymanek A, Havránek J, Tomela K, Rakoczy M, Samelak-Czajka A, Schmidt M, Figlerowicz M, Majta J, Milanowska-Zabel K, et al. The standardisation of the approach to metagenomic human gut analysis: from sample collection to microbiome profiling. Sci Rep. 2022;12:8470. doi: 10.1038/s41598-022-12037-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Brown G, Willment J, Whitehead L. C-type lectins in immunity and homeostasis. Nat Rev Immunol. 2018;18:374–389. doi: 10.1038/s41577-018-0004-8. [DOI] [PubMed] [Google Scholar]
- 27.Hardison S, Brown G. C-type lectin receptors orchestrate antifungal immunity. Nat Immunol. 2012;13:817–822. doi: 10.1038/ni.2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lionakis M, Drummond R, Hohl T. Immune responses to human fungal pathogens and therapeutic prospects. Nat Rev Immunol. 2023;23:433–452. doi: 10.1038/s41577-022-00826-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Valeri M, Raffatellu M. Cytokines IL-17 and IL-22 in the host response to infection. Pathog Dis 2016;74:ftw111. doi: 10.1093/femspd/ftw111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tsitsigiannis D, Keller N. Oxylipins as developmental and host-fungal communication signals. Trends Microbiol. 2007;15:109–118. doi: 10.1016/j.tim.2007.01.005. [DOI] [PubMed] [Google Scholar]
- 31.Neff R, Radka C. Exploring oxylipins in host–microbe interactions and their impact on infection and immunity. Curr Issues Mol Biol. 2025;47:190. doi: 10.3390/cimb47030190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Begum N, Harzandi A, Lee S, Uhlen M, Moyes D, Shoaie S. Host-mycobiome metabolic interactions in health and disease. Gut Microbes. 2022;14:2121576. doi: 10.1080/19490976.2022.2121576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.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. [DOI] [PubMed] [Google Scholar]
- 34.Wang T, Fan C, Yao A, Xu X, Zheng G, You Y, Jiang C, Zhao X, Hou Y, Hung M, et al. The Adaptor Protein CARD9 protects against colon cancer by restricting mycobiota-mediated expansion of myeloid-derived suppressor cells. Immunity. 2018;49:504–514. doi: 10.1016/j.immuni.2018.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Liu N, Jiao N, Tan J, Wang Z, Wu D, Wang A, 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;7:238–250. doi: 10.1038/s41564-021-01030-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lin Y, Lau H, Liu Y, Kang X, Wang Y, Ting N, Kwong T, 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;163:908–921. doi: 10.1053/j.gastro.2022.06.038. [DOI] [PubMed] [Google Scholar]
- 37.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;21:126. doi: 10.1186/s12967-023-03940-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bacher P, Hohnstein T, Beerbaum E, Röcker M, Blango M, Kaufmann S, Röhmel 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;176:1340–1355. doi: 10.1016/j.cell.2019.01.041. [DOI] [PubMed] [Google Scholar]
- 39.Szóstak N, Handschuh L, Samelak-Czajka A, Tomela K, Pietrzak B, Schmidt M, Galus Ł, Mackiewicz J, Mackiewicz A, Kozlowski P, Philips A, et al. Gut mycobiota dysbiosis is associated with melanoma and response to anti-pd-1 therapy. Cancer Immunol Res. 2024;12(4):427–439. doi: 10.1158/2326-6066.CIR-23-0592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Laroumagne S, Lepage B, Hermant C, Plat G, Phelippeau M, Bigay-Game L, Lozano S, Guibert N, Segonds C, Mallard V, et al. Bronchial colonisation in patients with lung cancer: a prospective study. Eur Respir J. 2013;42:220–229. doi: 10.1183/09031936.00062212. [DOI] [PubMed] [Google Scholar]
- 41.Malik A, Sharma D, Malireddi R, Guy C, Chang T, Olsen S, Neale G, Vogel P, Kanneganti T. SYK-CARD9 signaling axis promotes gut fungi-mediated inflammasome activation to restrict colitis and colon cancer. Immunity. 2018;49:515–530. doi: 10.1016/j.immuni.2018.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang Z, Guo K, Liu Y, Huang C, Wu M. Dynamic impact of virome on colitis and colorectal cancer: immunity, inflammation, prevention and treatment. Semin Cancer Biol. 2022;86:943–954. doi: 10.1016/j.semcancer.2021.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Alam A, Levanduski E, Denz P, Villavicencio H, Bhatta M, Alhorebi L, Zhang Y, Gomez E, Morreale B, Senchanthisai S, et al. Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell. 2022;40:153–167. doi: 10.1016/j.ccell.2022.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu Q, Zhang W, Pei Y, Tao H, Ma J, Li R, Zhang F, Wang L, Shen L, Liu Y, et al. Gut mycobiome as a potential non-invasive tool in early detection of lung adenocarcinoma: a cross-sectional study. BMC Med. 2023;21:409. doi: 10.1186/s12916-023-03095-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Szóstak N, Budnik M, Tomela K, Handschuh L, Samelak-Czajka A, Pietrzak B, Schmidt M, Kaczmarek M, Galus Ł, Mackiewicz J, et al. Exploring correlations between gut mycobiome and lymphocytes in melanoma patients undergoing anti-PD-1 therapy. Cancer Immunol Immunother. 2025;74(4):110. doi: 10.1007/s00262-024-03918-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhang F, Aschenbrenner D, Yoo J, Zuo T. The gut mycobiome in health, disease, and clinical applications in association with the gut bacterial microbiome assembly. Lancet Microbe. 2022;3:e969–e983. doi: 10.1016/S2666-5247(22)00203-8. [DOI] [PubMed] [Google Scholar]
- 47.Feng Z, Burgermeister E, Philips A, Zuo T, Wen W. The gut virome in association with the bacteriome in gastrointestinal diseases and beyond: roles, mechanisms, and clinical applications. Precis Clin Med. 2025;8:pbaf010. doi: 10.1093/pcmedi/pbaf010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wu Y, Cheng R, Lin H, Li L, Jia Y, Philips A, Zuo T, Zhang H. Gut virome and its implications in the pathogenesis and therapeutics of inflammatory bowel disease. BMC Med. 2025;23:183. doi: 10.1186/s12916-025-04016-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Seelbinder B, Lohinai Z, Vazquez-Uribe R, Brunke S, Chen X, Mirhakkak M, Lopez-Escalera S, Dome B, Megyesfalvi Z, Berta J, et al. Candida expansion in the gut of lung cancer patients associates with an ecological signature that supports growth under dysbiotic conditions. Nat Commun. 2023;14:2673. doi: 10.1038/s41467-023-38058-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Huang J, Roosaar A, Axéll T, Ye W. A prospective cohort study on poor oral hygiene and pancreatic cancer risk. Int J Cancer. 2016;138:340–347. doi: 10.1002/ijc.29710. [DOI] [PubMed] [Google Scholar]
- 51.Hold G, Allen-Vercoe E. Gut microbial biofilm composition and organisation holds the key to CRC. Nat Rev Gastroenterol Hepatol. 2019;16:329–330. doi: 10.1038/s41575-019-0148-4. [DOI] [PubMed] [Google Scholar]
- 52.Tomkovich S, Dejea C, Winglee K, Drewes J, Chung L, Housseau F, Pope J, Gauthier J, Sun X, Mühlbauer M, et al. Human colon mucosal biofilms from healthy or colon cancer hosts are carcinogenic. J Clin Invest. 2019;129:1699–1712. doi: 10.1172/JCI124196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jiang S, Xu L, Chen Y, Shu Z, Lv L, Zhao Y, Bi K, Yang S, Wang Q, Li L. Longitudinal gut fungal alterations and potential fungal biomarkers for the progression of primary liver disease. Sci China Life Sci. 2024;67(6):1183–1198. doi: 10.1007/s11427-023-2458-1. [DOI] [PubMed] [Google Scholar]
- 54.Wei Y, Song D, Wang R, Li T, Wang H, Li X. Dietary fungi in cancer immunotherapy: from the perspective of gut microbiota. Front Oncol. 2023;13. doi: 10.3389/fonc.2023.1038710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Iliev I, Leonardi I. Fungal dysbiosis: immunity and interactions at mucosal barriers. Nat Rev Immunol. 2017;17:635–646. doi: 10.1038/nri.2017.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim J, Shadaloey S, Wu D, Preiss P, Verma N, et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 2019;574:264–267. doi: 10.1038/s41586-019-1608-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zeng S, Hartmann P, Park M, Duan Y, Lang S, Llorente C, Wang Y, Cabré N, Fouts D, Bacher P, et al. Malassezia restricta promotes alcohol-induced liver injury. Hepatol Commun. 2023;7:e0029. doi: 10.1097/HC9.0000000000000029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Spatz M, Richard M. Overview of the potential role of malassezia in gut health and disease. Front Cell Infect Microbiol. 2020;10:201. doi: 10.3389/fcimb.2020.00201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vitali F, Colucci R, Di Paola M, Pindo M, De Filippo C, Moretti S, Cavalieri D. Early melanoma invasivity correlates with gut fungal and bacterial profiles. Br J Dermatol. 2022;186:106–116. doi: 10.1111/bjd.20626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hu M, Zhu X, Huang X, Hua L, Lin X, Zhang H, Hu Y, Tong T, Li L, Xuan B, et al. Optimizing anti-PD-1/PD-L1 therapy efficacy and fecal microbiota transplantation donor selection through gut mycobiome-based enterotype. Cell Rep. 2025;44:115589. doi: 10.1016/j.celrep.2025.115589. [DOI] [PubMed] [Google Scholar]
- 61.Zuo T, Wong S, Cheung C, Lam K, Lui R, Cheung K, Zhang F, Tang W, Ching J, Wu J, et al. Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat Commun. 2018;9:3663. doi: 10.1038/s41467-018-06103-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.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;31:1930–1943. doi: 10.1016/j.chom.2023.10.005. [DOI] [PubMed] [Google Scholar]
- 63.Mowat C, Dhatt J, Bhatti I, Hamie A, Baker K. Short chain fatty acids prime colorectal cancer cells to activate antitumor immunity. Front Immunol. 2023;14:1190810. doi: 10.3389/fimmu.2023.1190810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Muradás T, Freitas R, Gonçalves J, Xavier F, Marinowic D. Potential antitumor effects of short-chain fatty acids in breast cancer models. Am J Cancer Res. 2024;14:1999–2019. doi: 10.62347/ETUQ6763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Dora D, Weiss G, Megyesfalvi Z, Gállfy G, Dulka E, Kerpel-Fronius A, Berta J, Moldvay J, Dome B, Lohinai Z. Computed tomography-based quantitative texture analysis and gut microbial community signatures predict survival in non-small cell lung cancer. Cancers. 2023;15:5091. doi: 10.3390/cancers15205091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lam S, Bai X, Shkoporov A, Park H, Wu X, Lan P, Zuo T. Roles of the gut virome and mycobiome in faecal microbiota transplantation. Lancet Gastroenterol Hepatol. 2022;7:472–484. doi: 10.1016/S2468-1253(21)00303-4. [DOI] [PubMed] [Google Scholar]
- 67.Zuo T, Lu X, Zhang Y, Cheung C, Lam S, Zhang F, Tang W, Ching J, Zhao R, Chan P, et al. Gut mucosal virome alterations in ulcerative colitis. Gut. 2019;68:1169–1179. doi: 10.1136/gutjnl-2018-318131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yan F, Xia W, Zhang X, Chen S, Nie X, Qian L. Characterization of β-glucosidase from Aspergillus terreus and its application in the hydrolysis of soybean isoflavones. J Zhejiang Univ Sci B. 2016;17:455–464. doi: 10.1631/jzus.B1500317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Velasco J, Oliva B, Gonçalves A, Lima A, Ferreira G, França B, Mulinari E, Gonçalves T, Squina F, Kadowaki M, et al. Functional characterization of a novel thermophilic exo-arabinanase from Thermothielavioides terrestris. Appl Microbiol Biotechnol. 2020;104:8309–8326. doi: 10.1007/s00253-020-10806-6. [DOI] [PubMed] [Google Scholar]
- 70.Liu N, Yu W, Guo X, Chen J, Xia D, Yu J, Li D. Oxidative cleavage of cellulose in the horse gut. Microb Cell Factories. 2022;21:38. doi: 10.1186/s12934-022-01767-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ambrogi V, Bottacini F, Cao L, Kuipers B, Schoterman M, van Sinderen D. Galacto-oligosaccharides as infant prebiotics: production, application, bioactive activities and future perspectives. Crit Rev Food Sci Nutr. 2023;63:753–766. doi: 10.1080/10408398.2021.1953437. [DOI] [PubMed] [Google Scholar]
- 72.Yin H, Bultema J, Dijkhuizen L, van Leeuwen S. Reaction kinetics and galactooligosaccharide product profiles of the β-galactosidases from Bacillus circulans, Kluyveromyces lactis and Aspergillus oryzae. Food Chem. 2017;225:230–238. doi: 10.1016/j.foodchem.2017.01.030. [DOI] [PubMed] [Google Scholar]
- 73.Godínez-Méndez L, Gurrola-Díaz C, Zepeda-Nuño J, Vega-Magaña N, Lopez-Roa R, Íñiguez-Gutiérrez L, García-López P, Fafutis-Morris M, Delgado-Rizo V. In vivo healthy benefits of galacto-oligosaccharides from lupinus albus (LA-GOS) in butyrate production through intestinal microbiota. Biomolecules. 2021;11:1658. doi: 10.3390/biom11111658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Harris H, Morrison D, Edwards C. Impact of the source of fermentable carbohydrate on SCFA production by human gut microbiota in vitro - a systematic scoping review and secondary analysis. Crit Rev Food Sci Nutr. 2021;61:3892–3903. doi: 10.1080/10408398.2020.1809991. [DOI] [PubMed] [Google Scholar]
- 75.Gao Y, Xu P, Sun D, Jiang Y, Lin X, Han T, Yu J, Sheng C, Chen H, Hong J, et al. Faecalibacterium prausnitzii abrogates intestinal toxicity and promotes tumor immunity to increase the efficacy of dual CTLA4 and PD-1 checkpoint blockade. Cancer Res. 2023;83:3710–3725. doi: 10.1158/0008-5472.CAN-23-0605. [DOI] [PubMed] [Google Scholar]
- 76.Bredon M, Danne C, Pham H, Ruffié P, Bessede A, Rolhion N, Creusot L, Brot L, Alonso I, Langella P, et al. Faecalibaterium prausnitzii strain EXL01 boosts efficacy of immune checkpoint inhibitors. Oncoimmunology. 2024;13:2374954. doi: 10.1080/2162402X.2024.2374954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bredon M, Malicot K, le, Louvet C, Evesque L, Gonzalez D, Tougeron D, Sokol H. Faecalibacterium prausnitzii Is associated with clinical response to immune checkpoint inhibitors in patients with advanced gastric adenocarcinoma: results of microbiota analysis of PRODIGE 59-FFCD 1707-DURIGAST Trial. Gastroenterology. 2025;168:601–603. doi: 10.1053/j.gastro.2024.10.020. [DOI] [PubMed] [Google Scholar]
- 78.Malamud M, Brown G. The Dectin-1 and Dectin-2 clusters: c-type lectin receptors with fundamental roles in immunity. EMBO Rep. 2024;25:5239–5264. doi: 10.1038/s44319-024-00296-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dambuza I, Brown G. C-type lectins in immunity: recent developments. Curr Opin Immunol. 2015;32:21–27. doi: 10.1016/j.coi.2014.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hatinguais R, Willment J, Brown G. C-type lectin receptors in antifungal immunity: current knowledge and future developments. Parasite Immunol. 2023;45:e12951. doi: 10.1111/pim.12951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tang J, Lin G, Langdon W, Tao L, Zhang J. Regulation of C-type lectin receptor-mediated antifungal immunity. Front Immunol. 2018;9:123. doi: 10.3389/fimmu.2018.00123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Brown G, Gordon S. Immune recognition. A new receptor for beta-glucans. Nature. 2001;413:36–37. doi: 10.1038/35092620. [DOI] [PubMed] [Google Scholar]
- 83.He X, Howard B, Liu Y, Neumann A, Li L, Menon N, Roach T, Kale S, Samuels D, Li H, et al. LYSMD3:a mammalian pattern recognition receptor for chitin. Cell Rep. 2021;36:109392. doi: 10.1016/j.celrep.2021.109392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Saijo S, Ikeda S, Yamabe K, Kakuta S, Ishigame H, Akitsu A, Fujikado N, Kusaka T, Kubo S, Chung S, et al. Dectin-2 recognition of alpha-mannans and induction of Th17 cell differentiation is essential for host defense against Candida albicans. Immunity. 2010;32:681–691. doi: 10.1016/j.immuni.2010.05.001. [DOI] [PubMed] [Google Scholar]
- 85.Zhu L, Zhao X, Jiang C, You Y, Chen X, Jiang Y, Jia X, 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;39:324–334. doi: 10.1016/j.immuni.2013.05.017. [DOI] [PubMed] [Google Scholar]
- 86.van de Veerdonk F, Marijnissen R, Kullberg B, Koenen H, Cheng S, Joosten I, van den Berg W, Williams D, van der Meer J, Joosten L, et al. The macrophage mannose receptor induces IL-17 in response to Candida albicans. Cell Host Microbe. 2009;5:329–340. doi: 10.1016/j.chom.2009.02.006. [DOI] [PubMed] [Google Scholar]
- 87.Yasukawa S, Miyazaki Y, Yoshii C, Nakaya M, Ozaki N, Toda S, Kuroda E, Ishibashi K, Yasuda T, Natsuaki Y, et al. An ITAM-Syk-CARD9 signalling axis triggers contact hypersensitivity by stimulating IL-1 production in dendritic cells. Nat Commun. 2014;5:3755. doi: 10.1038/ncomms4755. [DOI] [PubMed] [Google Scholar]
- 88.Gross O, Gewies A, Finger K, Schäfer M, Sparwasser T, Peschel C, Förster I, Ruland J. Card9 controls a non-TLR signalling pathway for innate anti-fungal immunity. Nature. 2006;442:651–656. doi: 10.1038/nature04926. [DOI] [PubMed] [Google Scholar]
- 89.Gross O, Poeck H, Bscheider M, Dostert C, Hannesschläger N, Endres S, Hartmann G, Tardivel A, Schweighoffer E, Tybulewicz V, et al. Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature. 2009;459:433–436. doi: 10.1038/nature07965. [DOI] [PubMed] [Google Scholar]
- 90.Saijo S, Ikeda S, Yamabe K, Kakuta S, Ishigame H, Akitsu A, Fujikado N, Kusaka T, Kubo S, Chung S, et al. Dectin-2 recognition of α-mannans and induction of Th17 cell differentiation is essential for host defense against candida albicans. Immunity. 2010;32:681–691. doi: 10.1016/j.immuni.2010.05.001. [DOI] [PubMed] [Google Scholar]
- 91.Ruland J. CARD9 signaling in the innate immune response. Ann N Y Acad Sci. 2008;1143:35–44. doi: 10.1196/annals.1443.024. [DOI] [PubMed] [Google Scholar]
- 92.LeibundGut-Landmann S, Gross O, Robinson M, Osorio F, Slack E, Tsoni S, Schweighoffer E, Tybulewicz V, Brown G, 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;8:630–638. doi: 10.1038/ni1460. [DOI] [PubMed] [Google Scholar]
- 93.Gringhuis S, Wevers B, Kaptein T, van Capel T, Theelen B, Boekhout T, de Jong E, Geijtenbeek T. Selective C-Rel activation via Malt1 controls anti-fungal T(H)-17 immunity by dectin-1 and dectin-2. PLoS Pathog. 2011;7:e1001259. doi: 10.1371/journal.ppat.1001259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Jia X, Tang B, Zhu L, Liu Y, Zhao X, Gorjestani S, Hsu Y, Yang L, Guan J, Xu G, et al. CARD9 mediates Dectin-1-induced ERK activation by linking Ras-GRF1 to H-Ras for antifungal immunity. J Exp Med. 2014;211:2307–2321. doi: 10.1084/jem.20132349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gringhuis S, den Dunnen J, Litjens M, van der Vlist M, Wevers B, Bruijns S, Geijtenbeek T. Dectin-1 directs T helper cell differentiation by controlling noncanonical NF-kappaB activation through Raf-1 and Syk. Nat Immunol. 2009;10:203–213. doi: 10.1038/ni.1692. [DOI] [PubMed] [Google Scholar]
- 96.Daley D, Mani V, Mohan N, Akkad N, Ochi A, Heindel D, Lee K, Zambirinis C, Pandian G, Savadkar S, et al. Dectin 1 activation on macrophages by galectin 9 promotes pancreatic carcinoma and peritumoral immune tolerance. Nat Med. 2017;23:556–567. doi: 10.1038/nm.4314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Liu X, Lv K, Wang J, Lin C, Liu H, Zhang H, Li H, Gu Y, Li R, He H, et al. C-type lectin receptor Dectin-1 blockade on tumour-associated macrophages improves anti-PD-1 efficacy in gastric cancer. Br J Cancer. 2023;129:721–732. doi: 10.1038/s41416-023-02336-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tang C, Sun H, Kadoki M, Han W, Ye X, Makusheva Y, Deng J, Feng B, Qiu D, Tan Y, et al. Blocking Dectin-1 prevents colorectal tumorigenesis by suppressing prostaglandin E2 production in myeloid-derived suppressor cells and enhancing IL-22 binding protein expression. Nat Commun. 2023;14:1493. doi: 10.1038/s41467-023-37229-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Liu N, Yi C, Wei L, Zhou J, Jiang T, Hu C, Wang L, Wang Y, Zou Y, Zhao Y, et al. The intratumor mycobiome promotes lung cancer progression via myeloid-derived suppressor cells. Cancer Cell. 2023;41:1927–1944. doi: 10.1016/j.ccell.2023.08.012. [DOI] [PubMed] [Google Scholar]
- 100.Bhaskaran N, Jayaraman S, Quigley C, Mamileti P, Ghannoum M, Weinberg A, Thuener J, Pan Q, Pandiyan P. The role of dectin-1 signaling in altering tumor immune microenvironment in the context of aging. Front Oncol. 2021;11:669066. doi: 10.3389/fonc.2021.669066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Wang J, Li X, Wang K, Li K, Gao Y, Xu J, Peng R, Zhang X, Zhang S, Zhou Y, et al. CLEC7A regulates M2 macrophages to suppress the immune microenvironment and implies poorer prognosis of glioma. Front Immunol. 2024;15. doi: 10.3389/fimmu.2024.1361351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Seifert L, Deutsch M, Alothman S, Alqunaibit D, Werba G, Pansari M, Pergamo M, Ochi A, Torres-Hernandez A, Levie E, et al. Dectin-1 regulates hepatic fibrosis and hepatocarcinogenesis by suppressing TLR4 signaling pathways. Cell Rep. 2015;13:1909–1921. doi: 10.1016/j.celrep.2015.10.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wattenberg M, Coho H, Herrera V, Graham K, Stone M, Xue Y, Chang R, Cassella C, Liu M, Choi-Bose S, et al. Cancer immunotherapy via synergistic coactivation of myeloid receptors CD40 and Dectin-1. Sci Immunol. 2023;8:eadj5097. doi: 10.1126/sciimmunol.adj5097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Liu M, Luo F, Ding C, Albeituni S, Hu X, Ma Y, Cai Y, McNally L, Sanders M, Jain D, et al. Dectin-1 activation by a natural product β-glucan converts immunosuppressive macrophages into an M1-like phenotype. J Immunol. 195:5055–5065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mattiola I, Tomay F, De Pizzol M, Silva-Gomes R, Savino B, Gulic T, Doni A, Lonardi S, Astrid Boutet M, Nerviani A, et al. The macrophage tetraspan MS4A4A enhances dectin-1-dependent NK cell-mediated resistance to metastasis. Nat Immunol. 2019;20:1012–1022. doi: 10.1038/s41590-019-0417-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Wang D, Gao S, Chen J, Zhao Y, Jiang Y, Chu X, Wang X, Liu N, Qin T, Yi Q, et al. Dectin-1 stimulates IL-33 expression in dendritic cells via upregulation of IRF4. Lab Invest. 2018;98:708–714. doi: 10.1038/s41374-018-0047-2. [DOI] [PubMed] [Google Scholar]
- 107.Zhao Y, Chu X, Chen J, Wang Y, Gao S, Jiang Y, Zhu X, Tan G, Zhao W, Yi H, et al. Dectin-1-activated dendritic cells trigger potent antitumour immunity through the induction of Th9 cells. Nat Commun. 2016;7:12368. doi: 10.1038/ncomms12368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Chen J, Zhao Y, Jiang Y, Gao S, Wang Y, Wang D, Wang A, Yi H, Gu R, Yi Q, et al. Interleukin-33 contributes to the induction of th9 cells and antitumor efficacy by dectin-1-activated dendritic cells. Front Immunol. 2018;9:1787. doi: 10.3389/fimmu.2018.01787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chen J, Zhao Y, Chu X, Lu Y, Wang S, Yi Q. Dectin-1-activated dendritic cells: a potent Th9 cell inducer for tumor immunotherapy. Oncoimmunology. 2016;5:e1238558. doi: 10.1080/2162402X.2016.1238558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Bai Y, Ding J, He L, Zhu Z, Pan J, Qi C. β-Glucan induced plasma B cells differentiation to enhance antitumor immune responses by Dectin-1. BMC Immunol. 2025;26:2. doi: 10.1186/s12865-025-00681-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Quesnel-Vallières M, Schultz D, Orlenko A, Lo Y, Moore J, Ritchie M, Roth D, Carroll M, Barash Y, Lynch K, et al. Trametinib sensitivity is defined by a myeloid differentiation profile in acute myeloid leukemia. Drugs RD. 2024;24:489–499. doi: 10.1007/s40268-024-00491-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Fischer M, Spies-Weisshart B, Schrenk K, Gruhn B, Wittig S, Glaser A, Hochhaus A, Scholl S, Schnetzke U. Polymorphisms of dectin-1 and TLR2 predispose to invasive fungal disease in patients with acute myeloid leukemia. PLoS One. 2016;11:e0150632. doi: 10.1371/journal.pone.0150632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Mashhouri S, Rahmati A, Azimi A, Fava R.A, Ismail I.H, Walker J, Elahi S. Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model. Cell Oncol Dordr Neth. 2024;47:1735–1756. doi: 10.1007/s13402-024-00950-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Robinson M, Osorio F, Rosas M, Freitas R, Schweighoffer E, Gross O, Verbeek J, Ruland J, Tybulewicz V, Brown G, et al. Dectin-2 is a Syk-coupled pattern recognition receptor crucial for Th17 responses to fungal infection. J Exp Med. 2009;206:2037–2051. doi: 10.1084/jem.20082818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yamasaki S, Ishikawa E, Sakuma M, Hara H, Ogata K, Saito T. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol. 2008;9:1179–1188. doi: 10.1038/ni.1651. [DOI] [PubMed] [Google Scholar]
- 116.Miyake Y, Toyonaga K, Mori D, Kakuta S, Hoshino Y, Oyamada A, Yamada H, Ono K, Suyama M, Iwakura Y, et al. C-type lectin MCL is an FcRγ-coupled receptor that mediates the adjuvanticity of mycobacterial cord factor. Immunity. 2013;38:1050–1062. doi: 10.1016/j.immuni.2013.03.010. [DOI] [PubMed] [Google Scholar]
- 117.Sato K, Yang X, Yudate T, Chung J, Wu J, Luby-Phelps K, Kimberly R, Underhill D, Cruz P, Ariizumi K. Dectin-2 is a pattern recognition receptor for fungi that couples with the Fc receptor gamma chain to induce innate immune responses. J Biol Chem. 2006;281:38854–38866. doi: 10.1074/jbc.M606542200. [DOI] [PubMed] [Google Scholar]
- 118.Kerscher B, Willment J, Brown G. The Dectin-2 family of C-type lectin-like receptors: an update. Int Immunol. 2013;25:271–277. doi: 10.1093/intimm/dxt006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Li L, Huang X, Chen H. Unveiling the hidden players: exploring the role of gut mycobiome in cancer development and treatment dynamics. Gut Microbes. 2024;16:2328868. doi: 10.1080/19490976.2024.2328868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kimura Y, Inoue A, Hangai S, Saijo S, Negishi H, Nishio J, Yamasaki S, Iwakura Y, Yanai H, Taniguchi T. The innate immune receptor dectin-2 mediates the phagocytosis of cancer cells by Kupffer cells for the suppression of liver metastasis. Proc Natl Acad Sci U S A. 2016;113:14097–14102. doi: 10.1073/pnas.1617903113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Li J, Liu X, Ge R, Yin Y, Liu Y, Lu W, Huang M, He X, Wang J, Cai G, et al. The ligation between ERMAP, galectin-9 and dectin-2 promotes Kupffer cell phagocytosis and antitumor immunity. Nat Immunol. 2023;24:1813–1824. doi: 10.1038/s41590-023-01634-7. [DOI] [PubMed] [Google Scholar]
- 122.Bolt Biotherapeutics Inc. Bolt Biotherapeutics Reports First Quarter 2025 Financial Results and Provides Business Update. 2025. GlobeNewswire News Room. [Google Scholar]
- 123.Dumbrava E, Rodriguez I, Salkeni M, Giordano A, Chen C, Sahota S, Perez E, Yu T, Rosenthal K, Balacy K, et al. Abstract CT154: BDC-3042, a first-in-class Dectin-2 agonist, in patients with advanced malignancies: results from the first-in-human dose-escalation study. Cancer Res. 2025;85:CT154. doi: 10.1158/1538-7445.AM2025-CT154. [DOI] [Google Scholar]
- 124.Wang T, Pan D, Zhou Z, You Y, Jiang C, Zhao X, Lin X. Dectin-3 deficiency promotes colitis development due to impaired antifungal innate immune responses in the gut. PLoS Pathog. 2016;12:e1005662. doi: 10.1371/journal.ppat.1005662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Qu W, Qiao S, Liu L, Chen Y, Peng C, Hou Y, Xu Z, Lv M, Wang T. Dectin3 protects against hepatocellular carcinoma by regulating glycolysis of macrophages. Int Immunopharmacol. 2022;113:109384. doi: 10.1016/j.intimp.2022.109384. [DOI] [PubMed] [Google Scholar]
- 126.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:e105320. doi: 10.15252/embj.2020105320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Bugarcic A, Hitchens K, Beckhouse A, Wells C, Ashman R, Blanchard H. Human and mouse macrophage-inducible C-type lectin (Mincle) bind Candida albicans. Glycobiology. 2008;18:679–685. doi: 10.1093/glycob/cwn046. [DOI] [PubMed] [Google Scholar]
- 128.Ishikawa E, Ishikawa T, Morita Y, Toyonaga K, Yamada H, Takeuchi O, Kinoshita T, Akira S, Yoshikai Y, Yamasaki S. Direct recognition of the mycobacterial glycolipid, trehalose dimycolate, by C-type lectin Mincle. J Exp Med. 2009;206:2879–2888. doi: 10.1084/jem.20091750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Höft M, Hoving J, Brown G. Signaling C-type lectin receptors in antifungal immunity. Curr Top Microbiol Immunol. 2020;429:63–101. [DOI] [PubMed] [Google Scholar]
- 130.Thompson A, da Fonseca D, Walker L, Griffiths J, Taylor P, Gow N, Orr S. Dependence on mincle and dectin-2 varies with multiple candida species during systemic infection. Front Microbiol. 2021;12 633229. doi: 10.3389/fmicb.2021.633229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhao X, Zhu L, Chang Q, Jiang C, You Y, Luo T, Jia X, Lin X. C-type lectin receptor dectin-3 mediates trehalose 6,6′-dimycolate (TDM)-induced mincle expression through CARD9/Bcl10/MALT1-dependent nuclear factor (NF)-κB activation. J Biol Chem. 2014;289:30052–30062. doi: 10.1074/jbc.M114.588574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Li C, Xue V, Wang Q, Lian G, Huang X, Lee T, To K, Tang P, Lan H. The Mincle/Syk/NF-κB signaling circuit is essential for maintaining the protumoral activities of tumor-associated macrophages. Cancer Immunol Res. 2020;8:1004–1017. doi: 10.1158/2326-6066.CIR-19-0782. [DOI] [PubMed] [Google Scholar]
- 133.Seifert L, Werba G, Tiwari S, Giao Ly N, Alothman S, Alqunaibit D, Avanzi A, Barilla R, Daley D, Greco S, et al. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature. 2016;532:245–249. doi: 10.1038/nature17403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kodar K, Harper J, McConnell M, Timmer M, Stocker B. The mincle ligand trehalose dibehenate differentially modulates M1-like and M2-like macrophage phenotype and function via Syk signaling. Immun Inflamm Dis. 2017;5:503–514. doi: 10.1002/iid3.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Weth A, Dangerfield E, Timmer M, Stocker B. Recent advances in the development of mincle-targeting vaccine adjuvants. Vaccines. 2024;12:1320. doi: 10.3390/vaccines12121320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Woodworth J, Contreras V, Christensen D, Naninck T, Kahlaoui N, Gallouët A, Langlois S, Burban E, Joly C, Gros W, et al. MINCLE and TLR9 agonists synergize to induce Th1/Th17 vaccine memory and mucosal recall in mice and non-human primates. Nat Commun. 2024;15:8959. doi: 10.1038/s41467-024-52863-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yu Y, Wang R, Miao N, Tang J, Zhang Y, Lu X, Yan P, Wang J, Jia X. PD-L1 negatively regulates antifungal immunity by inhibiting neutrophil release from bone marrow. Nat Commun. 2022;13. doi: 6857. 10.1038/s41467-022-34722-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Deerhake M, Cardakli E, Shinohara M. Dectin-1 signaling in neutrophils up-regulates PD-L1 and triggers ROS-mediated suppression of CD4+ T cells. J Leukoc Biol. 2022;112:1413–1425. doi: 10.1002/JLB.3A0322-152RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Heine A, Held S, Schulte-Schrepping J, Wolff J, Klee K, Ulas T, Schmacke N, Daecke S, Riethausen K, Schultze J, et al. Generation and functional characterization of MDSC-like cells. Oncoimmunology. 2017;6:e1295203. doi: 10.1080/2162402X.2017.1295203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Karnam A, Bonam S, Rambabu N, Wong S, Aimanianda V, Bayry J. Wnt-β-catenin signaling in human dendritic cells mediates regulatory t-cell responses to fungi via the pd-l1 pathway. mBio. 2021;12:e02824-21. doi: 10.1128/mBio.02824-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wang M, Bai Y, Pei J, Li D, Pu X, Zhu W, Xia L, Qi C, Jiang H, Ning Y. β-Glucan combined with pd-1/pd-l1 checkpoint blockade for immunotherapy in patients with advanced cancer. Front Pharmacol. 2022;13:887457. doi: 10.3389/fphar.2022.887457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Kaminski V, Borges B, Santos B, Preite N, Calich V, Loures F. MDSCs use a complex molecular network to suppress T-cell immunity in a pulmonary model of fungal infection. Front Cell Infect Microbiol. 2024;14:1392744. doi: 10.3389/fcimb.2024.1392744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Li K, Chatterjee A, Qian C, Lagree K, Wang Y, Becker C, Freeman M, Murali R, Yang W, Underhill D. Profiling phagosome proteins identifies PD-L1 as a fungal-binding receptor. Nature. 2024;630:736–743. doi: 10.1038/s41586-024-07499-6. [DOI] [PubMed] [Google Scholar]
- 144.Chan A, Jonas A, Qiu X, Ottoson N, Walsh R, Gorden K, Harrison B, Maimonis P, Leonardo S, Ertelt K, et al. Imprime PGG-mediated anti-cancer immune activation requires immune complex formation. PLoS One. 2016;11:e0165909. doi: 10.1371/journal.pone.0165909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Hong F, Yan J, Baran J, Allendorf D, Hansen R, Ostroff G, Xing P, Cheung N, Ross G. Mechanism by which orally administered β-1,3-glucans enhance the tumoricidal activity of antitumor monoclonal antibodies in murine tumor models. J Immunol. 2004;173:797–806. doi: 10.4049/jimmunol.173.2.797. [DOI] [PubMed] [Google Scholar]
- 146.Martineau R, Susini S, Marabelle A. Fc effector function of immune checkpoint blocking antibodies in oncology. Immunol Rev. 2024;328:334–349. doi: 10.1111/imr.13427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Walker L, Sansom D. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol. 2011;11:852–863. doi: 10.1038/nri3108. [DOI] [PubMed] [Google Scholar]
- 148.Sansom D, Walker L. The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev. 2006;212:131–148. doi: 10.1111/j.0105-2896.2006.00419.x. [DOI] [PubMed] [Google Scholar]
- 149.Ovcinnikovs V, Ross E, Petersone L, Edner N, Heuts F, Ntavli E, Kogimtzis A, Kennedy A, Wang C, Bennett C, et al. CTLA-4–mediated transendocytosis of costimulatory molecules primarily targets migratory dendritic cells. Sci Immunol. 2019;4:eaaw0902. doi: 10.1126/sciimmunol.aaw0902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Turnbull C, Bones J, Stanley M, Medhavy A, Wang H, Lorenzo A, Cappello J, Shanmuganandam S, Pandey A, Seneviratne S, et al. DECTIN-1: amodifier protein in CTLA-4 haploinsufficiency. Sci Adv. 2023;9:eadi9566. doi: 10.1126/sciadv.adi9566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Kim H, Park K, Lee H, Kim J, Kim Y, Lee J, Kim K, Yun J, Hwang B, Hong J, et al. Curdlan activates dendritic cells through dectin-1 and toll-like receptor 4 signaling. Int Immunopharmacol. 2016;39:71–78. doi: 10.1016/j.intimp.2016.07.013. [DOI] [PubMed] [Google Scholar]
- 152.Ding C, Shrestha R, Zhu X, Geller A, Wu S, Woeste M, Li W, Wang H, Yuan F, Xu R, et al. Inducing trained immunity in pro-metastatic macrophages to control tumor metastasis. Nat Immunol. 2023;24:239–254. doi: 10.1038/s41590-022-01388-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Vuscan P, Kischkel B, Hatzioannou A, Markaki E, Sarlea A, Tintoré M, Cuñé J, Verginis P, de Lecea C, Chavakis T, et al. Potent induction of trained immunity by Saccharomyces cerevisiae β-glucans. Front Immunol. 2024;15:1323333. doi: 10.3389/fimmu.2024.1323333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Geller A, Shrestha R, Woeste M, Guo H, Hu X, Ding C, Andreeva K, Chariker J, Zhou M, Tieri D, et al. The induction of peripheral trained immunity in the pancreas incites anti-tumor activity to control pancreatic cancer progression. Nat Commun. 2022;13:759. doi: 10.1038/s41467-022-28407-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kalafati L, Kourtzelis I, Schulte-Schrepping J, Li X, Hatzioannou A, Grinenko T, Hagag E, Sinha A, Has C, Dietz S, et al. Innate immune training of granulopoiesis promotes anti-tumor activity. Cell. 2020;183:771–785. doi: 10.1016/j.cell.2020.09.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Zhu Z, He L, Bai Y, Xia L, Sun X, Qi C. Yeast β-glucan modulates macrophages and improves antitumor NK-cell responses in cancer. Clin Exp Immunol. 2023;214:50–60. doi: 10.1093/cei/uxad080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Liu X, Xu Y, Li Y, Pan Y, Zhao S, Hou Y. Ferumoxytol-β-glucan Inhibits melanoma growth via interacting with dectin-1 to polarize macrophages into m1 phenotype. Int J Med Sci. 2021;18:3125–3139. doi: 10.7150/ijms.61525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ning Y, Xu D, Zhang X, Bai Y, Ding J, Feng T, Wang S, Xu N, Qian K, Wang Y, et al. β-Glucan restores tumor-educated dendritic cell maturation to enhance antitumor immune responses. Int J Cancer. 2016;138:2713–2723. doi: 10.1002/ijc.30002. [DOI] [PubMed] [Google Scholar]
- 159.Albeituni S, Ding C, Liu M, Hu X, Luo F, Kloecker G, Bousamra M, Zhang I, Yan H. Yeast-Derived particulate β-glucan treatment subverts the suppression of myeloid-derived suppressor cells (mdsc) by inducing polymorphonuclear mdsc apoptosis and monocytic mdsc differentiation to apc in cancer. J Immunol. 2016;196:2167–2180. doi: 10.4049/jimmunol.1501853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Alexander M, Fiering S, Ostroff G, Cramer R, Mullins D. Beta-glucan-induced inflammatory monocytes mediate antitumor efficacy in the murine lung. Cancer Immunol Immunother CII. 2018;67:1731–1742. doi: 10.1007/s00262-018-2234-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Liu J, Gunn L, Hansen R, Yan J. Combined yeast-derived beta-glucan with anti-tumor monoclonal antibody for cancer immunotherapy. Exp Mol Pathol. 2009;86:208–214. doi: 10.1016/j.yexmp.2009.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Noorbakhsh Varnosfaderani S, Ebrahimzadeh F, Akbari Oryani M, Khalili S, Almasi F, Mosaddeghi Heris R, Payandeh Z, Li C, Nabi Afjadi M, Alagheband Bahrami A. Potential promising anticancer applications of β-glucans: a review. Biosci Rep. 2024;44(1), doi: 10.1042/BSR20231686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Steimbach L, Borgmann A, Gomar G, Hoffmann L, Rutckeviski R, de Andrade D, Smiderle F. Fungal beta-glucans as adjuvants for treating cancer patients – A systematic review of clinical trials. Clin Nutr. 2021;40:3104–3113. doi: 10.1016/j.clnu.2020.11.029. [DOI] [PubMed] [Google Scholar]
- 164.Seifeldin S, Upadhyay T, Trivedi R, Rezgui R, Saeed A. Detection of antimicrobial, antioxidant and reactive oxygen species and caspases 3/9 mediated anticancerous activity of β-Glucan particles derived from Pleurotus ostreatus against cervical cancer cells HeLa. J King Saud Univ - Sci. 2024;36:103577. doi: 10.1016/j.jksus.2024.103577. [DOI] [Google Scholar]
- 165.Wang N, Liu H, Liu G, Li M, He X, Yin C, Tu Q, Shen X, Bai W, Wang Q, et al. Yeast β-D-glucan exerts antitumour activity in liver cancer through impairing autophagy and lysosomal function, promoting reactive oxygen species production and apoptosis. Redox Biol. 2020;32:101495. doi: 10.1016/j.redox.2020.101495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Gainza-Cirauqui M, Nieminen M, Novak Frazer L, Aguirre-Urizar J, Moragues M, Rautemaa R. Production of carcinogenic acetaldehyde by Candida albicans from patients with potentially malignant oral mucosal disorders. J Oral Pathol Med. 2013;42:243–249. doi: 10.1111/j.1600-0714.2012.01203.x. [DOI] [PubMed] [Google Scholar]
- 167.Seitz H, Stickel F. Acetaldehyde as an underestimated risk factor for cancer development: role of genetics in ethanol metabolism. Genes Nutr. 2010;5:121–128. doi: 10.1007/s12263-009-0154-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Morelli M, Queiroz K. Breaking barriers: candidalysin disrupts epithelial integrity and induces inflammation in a gut-on-chip model. Toxins. 2025;17:89. doi: 10.3390/toxins17020089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Ho J, Camilli G, Griffiths J, Richardson J, Kichik N, Naglik J. Candida albicans and candidalysin in inflammatory disorders and cancer. Immunology. 2021;162:11–16. doi: 10.1111/imm.13255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Pickova D, Ostry V, Toman J, Malir F. Aflatoxins: history, significant milestones, recent data on their toxicity and ways to mitigation. Toxins. 2021;13:399. doi: 10.3390/toxins13060399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Kumar P, Mahato D, Kamle M, Mohanta T, Kang S. Aflatoxins: a global concern for food safety, human health and their management. Front Microbiol. 2016;7:2170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Marchese S, Polo A, Ariano A, Velotto S, Costantini S, Severino L. Aflatoxin B1 and M1: biological properties and their involvement in cancer development. Toxins. 2018;10:214. doi: 10.3390/toxins10060214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Magnussen A, Parsi M. Aflatoxins, hepatocellular carcinoma and public health. World J Gastroenterol. 2013;19:1508–1512. doi: 10.3748/wjg.v19.i10.1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.National Toxicology Program . 2021. Aflatoxins. In 15th Report on Carcinogens [Internet]. National Toxicology Program.
- 175.Wang Y, Wang L, Liu F, Wang Q, Selvaraj J, Xing F, Zhao Y, Liu Y. Ochratoxin a producing fungi, biosynthetic pathway and regulatory mechanisms. Toxins. 2016;8:83. doi: 10.3390/toxins8030083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Malir F, Pickova D, Toman J, Grosse Y, Ostry V. Hazard characterisation for significant mycotoxins in food. Mycotoxin Res. 2023;39:81–93. doi: 10.1007/s12550-023-00478-2. [DOI] [PubMed] [Google Scholar]
- 177.Obafemi B, Adedara I, Rocha J. Neurotoxicity of ochratoxin a: molecular mechanisms and neurotherapeutic strategies. Toxicology. 2023;497-498:153630. doi: 10.1016/j.tox.2023.153630. [DOI] [PubMed] [Google Scholar]
- 178.Chen X, F Abdallah M, Chen X, Rajkovic A. Current knowledge of individual and combined toxicities of aflatoxin b1 and fumonisin b1 in vitro. Toxins. 2023;15:653. doi: 10.3390/toxins15110653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Zeebone Y, Bóta B, Halas V, Libisch B, Olasz F, Papp P, Keresztény T, Gerőcs A, Ali O, Kovács M, et al. Gut-faecal microbial and health-marker response to dietary fumonisins in weaned pigs. Toxins. 2023;15:328. doi: 10.3390/toxins15050328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Gao Z, Luo K, Zhu Q, Peng J, Liu C, Wang X, Li S, Zhang H. The natural occurrence, toxicity mechanisms and management strategies of Fumonisin B1: a review. Environ Pollut Barking Essex 1987. 2023;320:121065. doi: 10.1016/j.envpol.2023.121065. [DOI] [PubMed] [Google Scholar]
- 181.Katsuyama A, Konno T, Shimoyama S, Kikuchi H. The mycotoxin patulin decreases expression of density-enhanced phosphatase-1 by down-regulating PPARγ in human colon cancer cells. Tohoku J Exp Med. 2014;233:265–274. doi: 10.1620/tjem.233.265. [DOI] [PubMed] [Google Scholar]
- 182.Singh N, Dev I, Pal S, Yadav S, Idris M, Ansari K. Transcriptomic and proteomic insights into patulin mycotoxin-induced cancer-like phenotypes in normal intestinal epithelial cells. Mol Cell Biochem. 2022;477:1405–1416. doi: 10.1007/s11010-022-04387-3. [DOI] [PubMed] [Google Scholar]
- 183.Noverr M, Toews G, Huffnagle G. Production of prostaglandins and leukotrienes by pathogenic fungi. Infect Immun. 2002;70:400–402. doi: 10.1128/IAI.70.1.400-402.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Martínez-Colón G, Moore B. Prostaglandin E2 as a regulator of immunity to pathogens. Pharmacol Ther. 2018;185:135–146. doi: 10.1016/j.pharmthera.2017.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Santiso A, Heinemann A, Kargl J. Prostaglandin E2 in the tumor microenvironment, a convoluted affair mediated by ep receptors 2 and 4. Pharmacol Rev. 2024;76:388–413. doi: 10.1124/pharmrev.123.000901. [DOI] [PubMed] [Google Scholar]
- 186.Luo M, He N, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Roles of prostaglandins in immunosuppression. Clin Immunol Orlando Fla. 2024;265:110298. doi: 10.1016/j.clim.2024.110298. [DOI] [PubMed] [Google Scholar]
- 187.Elewaut A, Estivill G, Novatchkova M, Bayerl F, Hoffmann-Haas L, Andreatta F, Nguyen T, Vulin M, Boettcher J, Obenauf A. 1473 Prostaglandin E2 and the tumor microenvironment: unleashing the potential of cancer immunotherapy through rational combination therapies. J Immunother Cancer. 2023;11. [Google Scholar]
- 188.Take Y, Koizumi S, Nagahisa A. Prostaglandin E receptor 4 antagonist in cancer immunotherapy: mechanisms of action. Front Immunol. 2020;11:324. doi: 10.3389/fimmu.2020.00324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Sarker D, Cook N, Leung C, Aktas B, Dawson R, Demeris N, Mozgunov P, Howe D, Austin N, Swain N, et al. 679TiP Cancer Research UK phase I/IIa trial of the prostaglandin E2 (PGE2) receptor 4 (EP4) antagonist HTL0039732 as monotherapy and in combination with immunotherapy in patients (pts) with advanced solid tumors. Ann Oncol. 2024;35:S529–S530. doi: 10.1016/j.annonc.2024.08.745. [DOI] [Google Scholar]
- 190.Trotta R, Rivis S, Zhao S, Orban M, Trusso Cafarello S, Charatsidou I, Pozniak J, Dehairs J, Vanheer L, Pulido Vicuna C, et al. Activated T cells break tumor immunosuppression by macrophage reeducation. Cancer Discov. 2025;15:1410–1436. doi: 10.1158/2159-8290.CD-24-0415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Silva C, Pinto D, Silva A. Chromones: a promising ring system for new anti-inflammatory drugs. ChemMedChem. 2016;11:2252–2260. doi: 10.1002/cmdc.201600359. [DOI] [PubMed] [Google Scholar]
- 192.Fan Y, Tian C, Tong S, Liu Q, Xu F, Shi B, Ai H, Liu J. Chromones from the endophytic fungus Bipolaris eleusines. Phytochemistry. 2024;221:114046. doi: 10.1016/j.phytochem.2024.114046. [DOI] [PubMed] [Google Scholar]
- 193.Patil V, Masand N, Verma S, Masand V. Chromones: privileged scaffold in anticancer drug discovery. Chem Biol Drug Des. 2021;98:943–953. doi: 10.1111/cbdd.13951. [DOI] [PubMed] [Google Scholar]
- 194.Zhang J, Wang Z, Song Z, Karthik L, Hou C, Zhu G, Jiang L, Han J, Ma R, Li L, et al. Brocaeloid D, a novel compound isolated from a wheat pathogenic fungus, microdochium majus 99049. Synth Syst Biotechnol. 2019;4:173–179. doi: 10.1016/j.synbio.2019.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Chen Y, Sun L, Yang H, Li Z, Liu J, Ai H, Wang G, Feng T. Depsidones and diaryl ethers from potato endophytic fungus Boeremia exigua. Fitoterapia. 2020;141:104483. doi: 10.1016/j.fitote.2020.104483. [DOI] [PubMed] [Google Scholar]
- 196.Yang H, Ai H, Feng T, Wang W, Wu B, Zheng Y, Sun H, He J, Li Z, Liu J. Trichothecrotocins A-C, antiphytopathogenic agents from potato endophytic fungus trichothecium crotocinigenum. Org Lett. 2018;20:8069–8072. doi: 10.1021/acs.orglett.8b03735. [DOI] [PubMed] [Google Scholar]
- 197.Teng S, Zhang X, Li H, Luo X, Zhou Y, Liu H, Liu J, Feng T. Boerechalasins A-G, cytochalasans from the fungus Boeremia exigua with anti-inflammatory and cytotoxic activities. Phytochemistry. 2023;215:113861. doi: 10.1016/j.phytochem.2023.113861. [DOI] [PubMed] [Google Scholar]
- 198.He Q, Zeng Q, Shao Y, Zhou H, Li T, Song F, Liu W. Anti-cervical cancer activity of secondary metabolites of endophytic fungi from Ginkgo biloba. Cancer Biomark. 2020;28:371–379. doi: 10.3233/CBM-190462. [DOI] [PubMed] [Google Scholar]
- 199.López-Legarda X, Rostro-Alanis M, Parra-Saldivar R, Villa-Pulgarín J, Segura-Sánchez F. Submerged cultivation, characterization and in vitro antitumor activity of polysaccharides from Schizophyllum radiatum. Int J Biol Macromol. 2021;186:919–932. doi: 10.1016/j.ijbiomac.2021.07.084. [DOI] [PubMed] [Google Scholar]
- 200.Ikeda Y, Adachi Y, Ishii T, Tamura H, Aketagawa J, Tanaka S, Ohno N. Blocking effect of anti-dectin-1 antibodies on the anti-tumor activity Of 1,3-β-glucan And the binding of dectin-1 to 1,3-β-Glucan. Biol Pharm Bull. 2007;30:1384–1389. doi: 10.1248/bpb.30.1384. [DOI] [PubMed] [Google Scholar]
- 201.Zhong K, Tong L, Liu L, Zhou X, Liu X, Zhang Q, Zhou S. Immunoregulatory and antitumor activity of schizophyllan under ultrasonic treatment. Int J Biol Macromol. 2015;80:302–308. doi: 10.1016/j.ijbiomac.2015.06.052. [DOI] [PubMed] [Google Scholar]
- 202.Xie Y, Li S, Sun L, Liu S, Wang F, Wen B, Sun L, Fang X, Chai Y, Cao H, et al. Fungal Immunomodulatory protein from nectria haematococca suppresses growth of human lung adenocarcinoma by inhibiting the PI3K/Akt pathway. Int J Mol Sci. 2018;19:3429. doi: 10.3390/ijms19113429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Sadahiro Y, Hitora Y, Kimura I, Hitora-Imamura N, Onodera R, Motoyama K, Tsukamoto S. Colletofragarone A2 inhibits cancer cell growth in vivo and leads to the degradation and aggregation of mutant p53. Chem Res Toxicol. 2022;35:1598–1603. doi: 10.1021/acs.chemrestox.2c00202. [DOI] [PubMed] [Google Scholar]
- 204.Mazloum-Ravasan S, Madadi E, Mohammadi A, Mansoori B, Amini M, Mokhtarzadeh A, Baradaran B, Darvishi F. Yarrowia lipolytica L-asparaginase inhibits the growth and migration of lung (A549) and breast (MCF7) cancer cells. Int J Biol Macromol. 2021;170:406–414. doi: 10.1016/j.ijbiomac.2020.12.141. [DOI] [PubMed] [Google Scholar]
- 205.Chen T, Liu J, Wang C, Wang Z, Zhou J, Lin J, Mao J, Pan T, Wang J, Xu H, et al. ALOX5 contributes to glioma progression by promoting 5-HETE-mediated immunosuppressive M2 polarization and PD-L1 expression of glioma-associated microglia/macrophages. J Immunother Cancer. 2024;12:e009492. doi: 10.1136/jitc-2024-009492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Rodríguez J, Casas J, Balboa M, Balsinde J. Bioactive lipid signaling and lipidomics in macrophage polarization: impact on inflammation and immune regulation. Front Immunol. 2025;16:1550500. doi: 10.3389/fimmu.2025.1550500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Yang C, Chang M, Chang C, Shyur L. Phytogalactolipid dLGG inhibits mouse melanoma brain metastasis through regulating oxylipin activity and re-programming macrophage polarity in the tumor microenvironment. Cancers. 2021;13:4120. doi: 10.3390/cancers13164120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Li J, Lee Y, Ma J, Liu B, Hsieh N, Chen D, Chu C, You R. The enhancing effect of fungal immunomodulatory protein-volvariella volvacea (Fip-vvo) on maturation and function of mouse dendritic cells. Life Basel Switz. 2021;11:471. [DOI] [PMC free article] [PubMed] [Google Scholar]



