Fungal influence on immune cells and inflammatory responses in the tumor microenvironment (Review)
- Authors:
- Published online on: November 11, 2024 https://doi.org/10.3892/ol.2024.14796
- Article Number: 50
Abstract
Introduction
Human microbiota
The human microbiota comprises diverse microbial communities that inhabit various regions inside and outside the human body. These communities include bacteria, fungi and viruses (1). These microbiota colonize distinct anatomical sites such as the oral cavity (2), skin (3), intestinal tract (4), reproductive tract (5) and even the brain (6). Each anatomical site provides a unique physiological environment, fostering the growth and proliferation of specific microorganisms tailored to those conditions (7,8). Consequently, every part of the body harbors its own distinct microbial community. Disruptions in the regulatory mechanisms that govern the microbiota of the host, whether caused by infections, dietary changes or lifestyle factors, can disturb the delicate balance between the microbiota and the host (9). These disturbances often result in dysbiosis, marked by an imbalance in the microbial composition within the body. Dysbiosis can manifest in various ways, including alterations in the abundance of specific microbial taxa, changes in microbial diversity or shifts in the metabolic activities of the microbiota (10). This dysbiotic state can have significant implications for human health, as it is increasingly recognized to contribute to the development or progression of various diseases, such as colorectal cancer, inflammatory bowel disease, and non-alcoholic fatty liver disease (11,12).
Tumor microenvironment (TME)
The concept of the TME was organically proposed by Stephen Paget, an assistant surgeon at the Royal London Hospital, over a century ago (13). Paget introduced the ‘seed and soil’ hypothesis of cancer, suggesting that the microenvironment surrounding tumors play a critical role in their growth and spread. TME refers to the intricate milieu in which tumor cells reside (14). It includes surrounding blood vessels, immune cells, fibroblasts, myeloid-derived inflammatory cells, various signaling molecules and extracellular matrix (ECM) (15). It serves as a dynamic ecosystem that influences the survival and progression of tumor cells. The interplay between tumor cells and immune cells, along with the complex signaling networks and molecular interactions, profoundly shape the fate of the tumor (16).
The present review focuses on the interaction between fungi and inflammatory response within the TME. Specifically, this study explores the potential role of fungi in tumor development through their interactions with immune cells. Understanding these interactions may offer novel insights into innovative therapeutic strategies that target TEM.
Relationship among human microbiota, TME and tumors
Recent advancements in molecular biology, genomics and high-throughput sequencing technology have shed light on the critical role of the microbiota in maintaining organismal homeostasis and influencing cancer development (17). The contribution of microorganisms and microbiota to carcinogenesis can be broadly categorized into altering the balance of host cell proliferation and death, modulating immune system function, and influencing host metabolism (11). Notably, certain microbial species, such as pks+ Escherichia coli and Clostridium nucleatum, have been implicated in colorectal carcinogenesis through modulating the TME and directly inducing gene mutations in epithelial cells (18).
Although bacteria and viruses have been extensively studied in relation to cancer (17), fungi have received less attention. Emerging evidence highlights the importance of fungi in cancer development by influencing the TME (19,20). A comprehensive study analyzing the mycobiomes of 17,401 tissue and blood samples across 35 cancer types has revealed a noteworthy positive correlation between fungal communities and cancer incidence (21). This finding emphasizes the potential role of fungi in tumorigenesis across diverse cancer types. Fungi exhibit heterogeneous distributions within different cancers, suggesting a nuanced relationship with the progression of specific tumor types. This variability in fungal presence and abundance highlights the complexity of their interactions within the TME and their potential impact on tumor behavior (22).
While the precise mechanisms by which fungi contribute to cancer development are still being elucidated, several fungi have been implicated in this process. Candida albicans, Malassezia spp., and Aspergillus spp. have garnered attention for their potential roles in oral squamous cell carcinoma, pancreatic ductal adenocarcinoma and lung adenocarcinoma. These fungi, along with others that yet to be fully characterized, represent intriguing targets for further investigation into their mechanisms of action and their implications for cancer development.
Candida albicans and cancer
The association between Candida spp. and gastrointestinal cancers, particularly gastric (23) and colon cancer (24), have been documented. Candida albicans, a common fungus in the human body, is typically found as a commensal organism, mainly in the colon and vagina. However, when the disturbance in the host-fungus balance can lead to excessive growth of C. albicans, leading to various health issues such as serious candidiasis (25) and current vulvovaginal candidiasis. The potential link between C. albicans and oral tumors was first observed in the 1960s (26). Subsequent reports have strengthened the evidence, suggesting a correlation between the presence of C. albicans in the oral mucosa and the development of oral squamous cell carcinoma (OSCC) (27–29). The association between C. albicans and OSCC is an example of explored fungus-cancer connections, the evidence for such associations (30–32). Abnormal colonization of the intestine by C. albicans and Malassezia furfur promotes the development and progression of HCC. In the integrated experiment, compared with the control group, the weight and volume of HCC tumors in the Candida albicans and Malassezia furfur groups significantly increased (33).
Malassezia and cancer
The connection between Malassezia and cancer, particularly pancreatic ductal adenocarcinoma (PDA), has been highlighted in a case report from 2022 (34) The report observed a significant increase of ~3,000-fold in the number of fungi in PDA compared with normal pancreatic tissue in both humans and mice subjects (34). To further investigate this relationship, researchers established models of slowly progressive and invasive PDA. The authors revealed that removing the mycobiome (the fungal community) protects against tumor growth, whereas re-introducing Malassezia accelerates oncogenesis, potentially through the glycans of the fungal wall binding to mannose-binding lectin, activating the complement cascade (34) and accelerating pancreatic cancer progression (35). Other experiments also confirmed that fungal pancreatic carcinogenesis occurred due to the activation of the complement cascade and the secretion of IL-33 and IL-6 (36,37).
In addition to PDA, elevated levels of Malassezia are also found in intestinal mucosa samples from patients with Crohn's disease compared with healthy individuals. Crohn's disease is closely associated with the development of colorectal cancer (CRC). This finding suggests the potential for early CRC diagnosis by assessing the presence of Malassezia in intestinal mucosa samples (38).
Aspergillus and cancer
The aflatoxin produced by Aspergillus flavus has confirmed to be one of the most potent natural carcinogens, known to cause liver cancer. Since its association with live cancer was first reported in 1990, researchers have extensively explored the potential mechanisms by which this fungus acts as a carcinogen (39). Through the use of Multicohort Fecal Metagenomic Analytics, scientists have also identified an association between Aspergillus lambellii and CRC (40).
However, current research primarily focuses on investigating the differences in Aspergillus abundance in the feces of patients with CRC compared with healthy individuals. While it has been established that fungi, including Aspergillus lambellii, can serve as biomarkers for CRC diagnosis, the specific mechanisms underlying this relationship are not yet fully understood (40). In the exploration of the correlation between Aspergillus and lung adenocarcinoma (LUAD), researchers have observed an enrichment of tumor resident Aspergillus not only in patients with LUAD, but also in three distinct homologous lung cancer mouse models. Through these models, it has been found that Aspergillus promotes lung tumor progression by mediating the expansion and activation of myeloid-derived suppressor cells via IL-1β signaling (41). How does this fungus activate the signaling pathway, as shown in Fig. 1. This process inhibits cytotoxic T lymphocytes activity and impedes the accumulation of PD-1+CD8+T cells (41), thereby facilitating tumor growth.
Other fungi and cancer
In addition to well-known fungi such as Malassezia, C. albicans and Aspergillus, other less common fungi species have also been contributed to tumor occurrence and development through dysbiosis of the microbial community. Advanced sequencing technologies such as whole-genome shotgun sequencing and high-throughput sequencing have been used by research to identify changes in these fungi in tumor patients compared with healthy individuals. For instance, there is an observed increase in the abundance of Trichophoron and Malassezia in the intestinal mucosa of patients with CRC (42), as well as Nakaseomyces and Skeletocutis in patients with pancreatic ductal adenocarcinoma (43). Aspergillus lambellii is closely related to the CRC-enriched bacterium Fusobacterium nucleatum, and this type of fungi can promote CRC cell proliferation and tumor growth in xenograft mice (40). Through pan-pathogen array (PathoChip), the 18S ribosomal RNA signal of dendritic spores has been detected in almost all ovarian cancer samples, in addition to the distinctive features of Pneumocystis, Acremonium, Cladophialophora, Malassezia and Pleistophora microsporidia (44). It has been revealed that >95% of ovarian cancer samples correlate with the features of Rhizomucor, Rhodotorula, Alternaria and Geotrichum, but the features of Geotrichum are also detected in all control samples (44). Thus, all the aforementioned mentioned fungi except Geotrichum deserve to be noticed in ovarian cancer.
Although these findings warrant further investigation, they underscore the importance of analyzing fungal dysbiosis in comprehending the mechanisms underlying cancer occurrence. Recent studies have also suggested an association between certain fungal species and tumor transcriptional profiles (21,22) or CRC stage (22,45), but a specific causal relationship is unknown.
While the link between these less common fungal species, as shown in Table I, and carcinogenesis may require further confirmation through additional clinical studies, it is evident that the relationship between tumors and fungi is intricate. Dysbiosis of fungi can significantly impact the immune response within the tumor microenvironment, thereby influencing tumor occurrence and progression.
Effect of immune cells induced by fungi in the TME
Fungi can elicit various immune responses within the TME through interactions with immune cells. The presence of fungi in the TME can activate immune cells, leading to both pro-inflammatory and anti-inflammatory responses.
Mast cells (MCs)
MCs are important resident sentinel in TEM, recognizing fungal cells and playing a crucial role in the immune response. The response of MCs to fungal cells depends on their morphology and MC subtype (46). Connective tissue mast cells predominantly respond to the mycelial state of C. albicans by promoting the production of anti-inflammatory cytokines (47). By contrast, mucosal mast cells (MMC) tend to react differently, promoting death and damage in response to yeast state of C. albicans (48).
During C. albicans infection in vivo, mast cells exhibit a dual role. They contribute to the initial inflammatory pathology (48,49) while also playing a crucial role in controlling fungal growth and spreading, as well as activating the Th1 immune response for memory protection during reinfection (49). In leaky gut models, Candida-driven IL-9 and MMC contribute to the loss of barrier function, dissemination of the fungus (50) and inflammation (51,52). This highlights the ability of C. albicans to exploit the versatility of the IL-9/MC axis for both symbiosis and pathogenesis. The IL-9/MC axis, by integrating signals from disturbed host/microbiota homeostasis, may serve as a marker for distinguishing between pathogenic and protective roles of fungi in the gut (49).
Candida-infected MCs have been shown to enhance the crawling ability of macrophages and to promote their chemotaxis towards the site of infection, suggesting an important role in modulating macrophage responses during C. albicans infections (53). As MCs maintain equilibrium between the host and commensal fungi such as C. albicans, their modulation of macrophage activity would likely helps limit fungal growth during infections (54). However, quiescent MCs can notably inhibit phagocytosis of C. albicans by macrophages in a contact-dependent manner (47). This property may indeed be implicated in suppressing macrophage activity, potentially aiding in immune evasion within the tumors.
Neutrophils
Neutrophils are an essential component of the innate immune system, equipped with both neutrophilic and specialized granules that aid in the digestion of bacteria and foreign bodies. They possess strong chemotaxis and phagocytosis capabilities, making them quick response at fungal infection sites (55). Studies have also found that neutrophils have a close association with tumor development in the TME (56–58). Depleting neutrophils leads to increased tumor growth, proliferation and invasiveness in mouse model of inflammation-induced and sporadic colon tumors (59).
Candidalysin, a peptide toxin secreted by C. albicans, play a crucial role in the interaction between the fungus and host cells. Candidalysin activates the EGFR-ERK pathway in a ligand-dependent manner (60), leading to the release of transcription factor c-Fos, granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor. These factors are essential for recruiting neutrophils to clear infections and can also promote cancer development.
While neutrophils are generally associated with anti-tumor activity due to their ability to eliminate pathogens and produce cytotoxic molecules, the potential influence of C. albicans on the TEM through its interaction with neutrophils via candidalysin secretion raises intriguing questions (61). It is plausible that C. albicans could modulate the TME to promote tumor progression by exploiting its interaction with neutrophils.
Macrophages
Macrophages are known as the first line of defense against pathogens. They are responsible for recognizing, phagocytosing and degrading cellular debris and pathogens. Additionally, they play a crucial role in presenting antigens to T cells and inducing the expression of co-stimulatory molecules by other antigen-presenting cells (62). Recent research has revealed a dichotomy in macrophage polarization, identified as two distinct forms: M1 and M2. M1 macrophages produce type I pro-inflammatory cytokines, participate in antigen presentation and exert an antitumor role (63). Conversely, M2 macrophages produce type II cytokines that promote anti-inflammatory responses and possess pro-tumor functions (64). In fact, macrophages play a pivotal role in supporting various aspects of tumor progression and tumor cell invasion (65).
Mechanistically, it has been discovered that phenylpyruvic acid derived from C. albicans directly binds to sirtuin 2, leading to an increase in reactive oxygen species (ROS) production (66). ROS, in turn, modulate various cellular signaling pathways primarily involving transcription factors NF-κB and STAT3 (67), hypoxia-inducible factor-1α (68), kinases, growth factors, cytokines and other proteins and enzymes. These pathways have been implicated in various aspects of cancer biology, including cellular transformation, inflammation, tumor survival, proliferation, invasion, angiogenesis and metastasis (69,70). In a mouse model lacking the c-type lectin Dectin-3 (Dectin-3-/-), increased C. albicans load triggers glycolysis in macrophages and secretion of IL-7, which induces IL-22 production in type 3 innate lymphoid cells (ILC3) via the aryl hydrocarbon receptor and STAT3 signaling pathway (71). Notably, IL-22 has been shown to promote tumor growth in several models (72,73). This aligns with the fact that IL-22 is a potent activator of STAT3 in epithelial cells (74), which is required for the development of colitis-associated cancer (75). Based on these findings, it is reasonable to speculate that C. albicans promotes macrophages activation via the IL-22/STAT3 axis, contributing to the complex interplay between fungal infection and cancer development.
Similar effects on macrophages have been reported in another yeast species, including Malassezia. The enrichment of Malassezia in fungal communities found in mouse and human pancreatic tumors suggests a potential carcinogenic role for this organism (31), possibly through its influence on the immune system of the host, such as suppressing immunity via the inhibition of M1 macrophage differentiation. Therefore, targeting fungal populations such as Malassezia or modulating inflammatory responses associated with these fungi may hold significant promise in reshaping the TEM and enhancing cancer immunotherapy. By disrupting the immunosuppressive effects of these fungi, it may be possible to enhance the antitumor immune response and improve the effectiveness of immunotherapy for cancer treatment.
Dendritic cells (DCs)
DCs play a crucial role in both initiating the primary host immune response and enhancing the secondary host immune response. As the most potent antigen presenting cell, DCs excel at stimulating the activation and proliferation of naive T cells, making them primary initiators of specific immunity.
Recent findings have revealed that the yeast form of C. albicans induces Th17 cell responses through a mechanism that requires Dectin-1-mediated expression of IL-6 by DCs (76). This underscores the potential of DCs to not only mediate immune responses to fungal infections but also to exert influences on the TEM. However, despite the critical role of DCs in promoting T cell responses (77), the interaction between fungi and DCs within the TME remains relatively unstudied. Understanding the dynamics of this interaction, along with the broader interplay between tumors, immune cells and fungi in the TME, will continue to be a prominent focus in future studies.
Natural Killer (NK) cells
In recent years, the importance of NK cells in antifungal immunity (43,78) has been recognized, in addition to their well-known role in combating viruses and tumors. While NK cells are capable of recognizing and eliminating C. albicans, fungal cells can also inhibit NK cells by manipulating the immune checkpoint receptor T cell immunoreceptor with Ig and ITIM domains in both humans and mice (79). Despite this emerging understanding, there is a lack of studies investigating in the interaction between NK cells and C. albicans within TME. More in-depth research is needed to further explore this interaction.
T cells
Regulatory T cells (Treg cells)Treg cells, a subpopulation of CD4+ T cells, play a crucial role in immune homeostasis by preventing autoimmunity, controlling excessive inflammation and maintaining immune tolerance. In normal physiological conditions, Treg cells regulate the proliferation and activation of T and B cells, as well as the homeostasis of innate cytotoxic lymphocytes (80). However, the immunosuppressive effect of Treg cells can lead to immune escape of tumor cells, indirectly promoting tumor cell proliferation and enhancing tumor cells infiltration capacity (81).
In a study by Ahmadi et al (82), mice were divided into four groups: Normal, tumor, Candida infected and tumor/Candida groups. The results demonstrated that Candida infection led to an increase in the Treg population in the TME in the experimental group compared with the uninfected control group. Similarly, in a mouse model of lung cancer constructed by Liu et al, a higher proportion of Tregs was observed in the TME of mice exposed to Aspergillus sydowii (41). These experiments collectively suggest that fungi may contribute to the occurrence and development of tumors by inhibiting antitumor immune responses through mechanisms involving Treg cells. However, the specific signaling pathways and mechanisms underlying this response remain unclear and warrant further investigation. Understanding these mechanisms could provide insights into potential therapeutic strategies for targeting fungal-induced immunosuppression in cancer.
T helper (Th)2 cells
Th2 cells and ILC2 have been shown to stimulate tumor growth by secreting pro-tumorigenic cytokines such as IL-4, IL-5 and IL-13 (83). Notably, the cancer-cell-specific deletion of IL-33 has been found to reduce the recruitment Th2 and ILC2, thereby promoting tumor regression (84,85). Notably, the secretion of IL-33 has been found to be dependent on the intra-tumoral fungal mycobiome (86). Specifically, Malassezia in the pancreas has been implicated in promoting the occurrence of pancreatic tumors by promoting the secretion of IL-33. Genetic deletion of IL-33 or anti-fungal treatment can decrease TH2 and ILC2 infiltration and increases survival (84).
According to a 2016 report, all cases of atrophying pityriasis versicolor had Malassezia infiltration within the stratum corneum and were surrounded by focal interfacial dermatitis mediated by Th2 cell (87). These findings may reveal the generation of Malassezia-primed Th2 cells.
Th17 cells
C. albicans regulates Th17 cells proliferation, leading to increased secretion of IL-17A, IL-17F, and IL-22. IL-17 activates NF- κB through the MAP kinase pathway, enhancing its biological activity (88). IL-17A is a pro-inflammatory cytokine that recruits neutrophils and promotes their proliferation, maturation and chemotaxis. It also synergistically activates T cells and promote cell maturation (89).
A recent study revealed that C. albicans infection induces the expression of Gasdermin E (GSDME) in a specific subset of human Th17 cells (90). GSDME is a membrane pore-forming molecule that induces rapid cell death, suggesting its potential role as a cancer checkpoint (91). Investigating GSDME expression on the surface of Th17 cells following C. albicans infection could provide valuable novel insights for tumor diagnosis. Further research is required to explore the implications of C. albicans-mediated GSDME response on Th17 cells within TEM. Understanding the impact of ton tumor development, progression and therapy response could lead to innovative approaches to cancer diagnosis and treatment (90).
CD8+ T cells
CD8+ T cells play a key role in eliminating intracellular infections and malignant cells and provide long-term protective immunity (92). There are multiple subpopulations of CD8+ T cells, each with distinct effector functions and cytotoxic potential; for example, Tc1s has special cytotoxic activity and can effectively kill tumor cells and cells containing intracellular pathogens (93). Tc17 can produce IL17, which acts on tumor cells or other cells within the TME (94). These subpopulations have also been detected in the TME (95)
On the other hand, CD8+ T cells themselves can target the fungus to produce and secrete large amounts of IFN-γ and protease granzyme B to control the infection (96). For example, in a mouse model of lung adenocarcinoma, the proportion of CD8+ T cells in the TME increased after Aspergillus sydowii infection, revealing that CD8+ T cells and fungi reach a delicate balance in the TME to regulate tumor development (41).
Future perspectives
The presence of specific fungi within the TME holds promise as novel diagnostic or prognostic marker for specific types of cancer. Recent research by Narunsky Haziza et al in 2022 investigated the relationship between clinical data of various cancer patients and fungal species, revealing positive associations between fungi and various clinical outcomes. For instance, fungi were found to be positively associated with total survival time of patients with breast cancer, progression free survival time in patients with ovarian cancer, immunotherapeutic response in patients with melanoma and detection of early cancer (21). These findings suggest that fungi could serve as valuable biomarkers and potential therapeutic targets in cancer management. The analyses of gut microbiota as a screening, prognostic or predictive biomarkers have already begun to meet clinical applications, offering a promising avenue for preventing cancer, enhancing treatment efficacy and reducing adverse treatment effects through microbiota regulation (97).
Recent studies have also highlighted the significant role of fungi in cancer treatment and drug resistance. In addition to the direct approach of reducing Th2 and ILC2 infiltration through antifungal therapy, which can improve the survival rate of patients with cancer (86), the use of some fungal metabolites, such as polysaccharides and triterpenes, in conjunction with immune therapy, has garnered attention for their antitumor effects through various mechanisms (98). Polysaccharides can serve as key antitumor metabolites in numerous fungi infections through their impacts on host immunity. They exhibit diverse effects on host immunity by enhancing immunity, inhibiting tumor cell growth and inducing tumor cell apoptosis (99). Polysaccharides derived from Ganoderma atrum have been reported to activate macrophages, improve immunity and inhibit colorectal tumor growth through a TLR4 dependent signaling pathway (100). In addition to inhibiting tumor growth, Ganoderma polysaccharides have shown promise in antihepatocellular cancer treatment by modulating macrophage polarization (86) or increasing the ratio of the effector T cells to Tregs (101). Another example is Poria cocos polysaccharides, which have been implicated in exerting antitumor effects. According to experiments conducted by Jing et al, Poria cocos polysaccharides have the ability to induce apoptosis in ovarian cancer cells (102).
Triterpenes represent another common antitumor component found in fungi. Feng et al (103) studied the inhibitory effects of Ganoderma triterpenoids on cell proliferation and tumor growth, revealing their ability to induce cell death in lung cancer cells in vitro. Additionally, in a Lewis tumor-bearing mouse model, Ganoderma triterpenoids significantly delayed its tumor growth (104). Although research on this ingredient primarily focuses on in vitro and mouse experiments, lacking clinical trial data, its potential therapeutic effects warrant further exploration. Investigation whether Ganoderma triterpenoids has a specific effect on the treatment of patients with lung cancer is an important step for future exploration. Furthermore, assessment of their safety and toxicity are also worth further research. However, regardless, the combined use of these products in clinical settings is also essential.
Anticancer drug resistance is present in almost all types of cancers (105). Research on the relationship between the immune microenvironment and tumor resistance is advancing rapidly. T-cell depletion is a well-documented factor in promoting resistance to anticancer drugs, and strategies to enhance T-cell infiltration show promise in overcoming this anticancer drug resistance. Notably, fungi have been increasingly recognized for their influence on immune function and their potential role in cancer treatment. A study utilizing a fecal microbiota transplantation to boost the effectiveness of PD-1 therapy in drug-resistant melanoma patients marks a significant breakthrough (106). It underscores the potential for manipulating the microbiome, including fungi, to enhance responses to immunotherapy (107). As research continues to uncover fungal-bacterial communities capable of significantly improving tumor treatment outcomes, there is hope for extending these benefits beyond melanoma to other cancer types treated with antitumor therapies.
Both animal experiments and clinical studies have found the significant impact of gut microbiota (108) on enhancing the anticancer effects of checkpoint inhibitors such as CTLA-4, PD-1, and PD-L1 in treating lung cancer (109), renal cell carcinoma (110) and melanoma (106,107,111). Changes in gut microbiota appear to enhance the effectiveness of PD-1 inhibitors by possibly activating the IL-12 signaling pathway, which boosts the response of Th1 cells and activates T lymphocytes (108). Modulating the proportion of probiotics and fungi within the gut microbiota, potentially through antifungal drugs, could further optimize the efficacy of PD-1 inhibitors.
Numerous studies have demonstrated the capacity of gut microbiota to influence the body's immune function and bolster the efficacy of immunotherapy, indicating that manipulating gut microbiota could emerge as a crucial element in tumor prevention and immunotherapy (81,112,113). The highlighted therapies underscore the significant interplay between fungi and the immune system, presenting considerable promise in tumor prevention and treatment. However, it is essential to conduct additional clinical trials to confirm the efficacy and safety of these approaches.
Conclusions
In conclusion, the relationship between fungi, immunity and the TEM is intricate and significant. Fungi possess the ability to modulate immunity through diverse mechanisms, consequently influencing the TME. The cytokines produced by different immune cell phenotypes play a crucial mediator in determining tumor outcomes.
Studying the interaction between the tumor immune system and microbiota holds promise for uncovering novel therapeutic targets. It is essential to gain a deeper understanding of molecular mechanisms underlying fungal actions in tumors. This knowledge will contribute to the discovery of novel therapeutic targets and such endeavors will not only facilitate the discovery of new therapeutic targets but also aid in identifying valuable biomarkers for improved cancer treatment strategies.
Acknowledgements
Not applicable.
Funding
This work was supported by grants from the National Nature Science Foundation of China (grant no. NM 82272358), the Key Research and Development Plan of Jining (grant nos. NM 2023YXNS001 and NM 2022YXNS127) and the Medicine health science and Technology Development Plan of Shandong Province (grant no. 202202070556).
Availability of data and materials
Not applicable.
Authors' contributions
JZ wrote the original draft. YF participated in the revision of the draft manuscript. DM supervised the study, visualized the data, and reviewed and edited the manuscript. DS provided resources, visualized the data, and reviewed and edited the paper. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
El-Sayed A, Aleya L and Kamel M: Microbiota's role in health and diseases. Environ Sci Pollut Res Int. 28:36967–36983. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gao L, Xu T, Huang G, Jiang S, Gu Y and Chen F: Oral microbiomes: More and more importance in oral cavity and whole body. Protein Cell. 9:488–500. 2018. View Article : Google Scholar : PubMed/NCBI | |
Byrd AL, Belkaid Y and Segre JA: The human skin microbiome. Nat Rev Microbiol. 16:143–155. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sommer F, Anderson JM, Bharti R, Raes J and Rosenstiel P: The resilience of the intestinal microbiota influences health and disease. Nat Rev Microbiol. 15:630–638. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chen C, Song X, Wei W, Zhong H, Dai J, Lan Z, Li F, Yu X, Feng Q, Wang Z, et al: The microbiota continuum along the female reproductive tract and its relation to uterine-related diseases. Nat Commun. 8:8752017. View Article : Google Scholar : PubMed/NCBI | |
Ottman N, Smidt H, de Vos WH and Belzer C: The function of our microbiota: Who is out there and what do they do? Front Cell Infect Microbiol. 2:1042012. View Article : Google Scholar : PubMed/NCBI | |
Zoetendal EG, Rajilic-Stojanovic M and de Vos WM: High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut. 57:1605–1615. 2008. View Article : Google Scholar : PubMed/NCBI | |
Davenport ER, Sanders JG, Song SJ, Amato KR, Clark AG and Knight R: The human microbiome in evolution. BMC Biol. 15:1272017. View Article : Google Scholar : PubMed/NCBI | |
Garrett WS: Cancer and the microbiota. Science. 348:80–86. 2015. View Article : Google Scholar : PubMed/NCBI | |
Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J and Chen ZS: Microbiota in health and diseases. Signal Transduct Target Ther. 7:1352022. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Zhou J and Wang L: Role and mechanism of gut microbiota in human disease. Front Cell Infect Microbiol. 11:6259132021. View Article : Google Scholar : PubMed/NCBI | |
Belvoncikova P, Maronek M and Gardlik R: Gut dysbiosis and fecal microbiota transplantation in autoimmune diseases. Int J Mol Sci. 23:107292022. View Article : Google Scholar : PubMed/NCBI | |
Paget S: The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 8:98–101. 1989.PubMed/NCBI | |
Preisler HD, Bjornsson S, Mori M and Barcos M: Granulocyte differentiation by friend leukemia cells. Cell Differ. 4:273–283. 1975. View Article : Google Scholar : PubMed/NCBI | |
Izraely S and Witz IP: Site-specific metastasis: A cooperation between cancer cells and the metastatic microenvironment. Int J Cancer. 148:1308–1322. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bicher HI, Hetzel FW, Sandhu TS, Frinak S, Vaupel P, O'Hara MD and O'Brien T: Effects of hyperthermia on normal and tumor microenvironment. Radiology. 137:523–530. 1980. View Article : Google Scholar : PubMed/NCBI | |
Xie Y, Xie F, Zhou X, Zhang L, Yang B, Huang J, Wang F, Yan H, Zeng L, Zhang L and Zhou F: Microbiota in tumors: From understanding to application. Adv Sci (Weinh). 9:e22004702022. View Article : Google Scholar : PubMed/NCBI | |
Huang C, Liu H, Gong X, Wen B, Chen D, Liu J and Hu F: Analysis of different components in the peritumoral tissue microenvironment of colorectal cancer: A potential prospect in tumorigenesis. Mol Med Rep. 14:2555–2565. 2016. View Article : Google Scholar : PubMed/NCBI | |
de Martel C, Ferlay J, Franceschi S, Vignat J, Bray F, Forman D and Plummer M: Global burden of cancers attributable to infections in 2008: A review and synthetic analysis. Lancet Oncol. 13:607–615. 2012. View Article : Google Scholar : PubMed/NCBI | |
Vallianou N, Kounatidis D, Christodoulatos GS, Panagopoulos F, Karampela I and Dalamaga M: Mycobiome and cancer: What is the evidence? Cancers (Basel). 13:31492021. View Article : Google Scholar : PubMed/NCBI | |
Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, Gavert N, Stajich JE, Amit G, González A, et al: Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 185:3789–3806.e17. 2022. View Article : Google Scholar : PubMed/NCBI | |
Dohlman AB, Klug J, Mesko M, Gao IH, Lipkin SM, Shen X and Iliev ID: A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 185:3807–3822. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhong M, Xiong Y, Zhao J, Gao Z, Ma J, Wu Z, Song Y and Hong X: Candida albicans disorder is associated with gastric carcinogenesis. Theranostics. 11:4945–4956. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ramirez-Garcia A, Rementeria A, Aguirre-Urizar JM, Moragues MD, Antoran A, Pellon A, Abad-Diaz-de-Cerio A and Hernando FL: Candida albicans and cancer: Can this yeast induce cancer development or progression? Crit Rev Microbiol. 42:181–193. 2016.PubMed/NCBI | |
Conti HR and Gaffen SL: Host responses to Candida albicans: Th17 cells and mucosal candidiasis. Microbes Infect. 12:518–527. 2010. View Article : Google Scholar : PubMed/NCBI | |
Williamson DM: Chronic hyperplastic candidiasis and squamous carcinoma. Br J Dermatol. 81:125–127. 1969. View Article : Google Scholar : PubMed/NCBI | |
Gupta SR, Gupta N, Sharma A, Xess I, Singh G and Mani K: The association of Candida and antifungal therapy with pro-inflammatory cytokines in oral leukoplakia. Clin Oral Investig. 25:6287–6296. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bastiaan RJ and Reade PC: The prevalence of Candida albicans in the mouths of tobacco smokers with and without oral mucous membrane keratoses. Oral Surg Oral Med Oral Pathol. 53:148–151. 1982. View Article : Google Scholar : PubMed/NCBI | |
Daftary DK, Mehta FS, Gupta PC and Pindborg JJ: The presence of Candida in 723 oral leukoplakias among Indian villagers. Scand J Dent Res. 80:75–79. 1972.PubMed/NCBI | |
Healy CM and Moran GP: The microbiome and oral cancer: More questions than answers. Oral Oncol. 89:30–33. 2019. View Article : Google Scholar : PubMed/NCBI | |
Alnuaimi AD, Wiesenfeld D, O'Brien-Simpson NM, Reynolds EC and McCullough MJ: Oral Candida colonization in oral cancer patients and its relationship with traditional risk factors of oral cancer: A matched case-control study. Oral Oncol. 51:139–145. 2015. View Article : Google Scholar : PubMed/NCBI | |
McCullough M, Jaber M, Barrett AW, Bain L, Speight PM and Porter SR: Oral yeast carriage correlates with presence of oral epithelial dysplasia. Oral Oncol. 38:391–393. 2002. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Chen C, Chai D, Li C, Qiu Z, Kuang T, Liu L, Deng W and Wang W: Characterization of the intestinal fungal microbiome in patients with hepatocellular carcinoma. J Transl Med. 21:1262023. View Article : Google Scholar : PubMed/NCBI | |
Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim JI, Shadaloey SA, Wu D, Preiss P, Verma N, et al: The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 574:264–267. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ishikawa T, Itoh F, Yoshida S, Saijo S, Matsuzawa T, Gonoi T, Saito T, Okawa Y, Shibata N, Miyamoto T and Yamasaki S: Identification of distinct ligands for the C-type lectin receptors mincle and dectin-2 in the pathogenic Fungus Malassezia. Cell Host Microbe. 13:477–488. 2013. View Article : Google Scholar : PubMed/NCBI | |
Zambirinis CP, Pushalkar S, Saxena D and Miller G: Pancreatic cancer, inflammation, and microbiome. Cancer J. 20:195–202. 2014. View Article : Google Scholar : PubMed/NCBI | |
Sparmann A and Bar-Sagi D: Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell. 6:447–458. 2004. View Article : Google Scholar : PubMed/NCBI | |
Limon JJ, Tang J, Li D, Wolf AJ, Michelsen KS, Funari V, Gargus M, Nguyen C, Sharma P, Maymi VI, et al: Malassezia Is associated with Crohn's disease and exacerbates colitis in mouse models. Cell Host Microbe. 25:377–388.e6. 2019. View Article : Google Scholar : PubMed/NCBI | |
Nixon MW: Aflatoxin and liver cancer. Lancet. 335:11651990. View Article : Google Scholar : PubMed/NCBI | |
Lin Y, Lau HC, Liu Y, Kang X, Wang Y, Ting NL, Kwong TN, Han J, Liu W, Liu C, et al: Altered mycobiota signatures and enriched pathogenic Aspergillus rambellii are associated with colorectal cancer based on multicohort fecal metagenomic analyses. Gastroenterology. 163:908–921. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liu NN, Yi CX, Wei LQ, Zhou JA, Jiang T, Hu CC, Wang L, Wang YY, Zou Y, Zhao YK, et al: The intratumor mycobiome promotes lung cancer progression via myeloid-derived suppressor cells. Cancer Cell. 41:1927–1944. 2023. View Article : Google Scholar : PubMed/NCBI | |
Gao R, Kong C, Li H, Huang L, Qu X, Qin N and Qin H: Dysbiosis signature of mycobiota in colon polyp and colorectal cancer. Eur J Clin Microbiol Infect Dis. 36:2457–2468. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Shi ZW, Strickland AB and Shi M: Cryptococcus neoformans infection in the central nervous system: The battle between host and pathogen. J Fungi (Basel). 8:10692022. View Article : Google Scholar : PubMed/NCBI | |
Banerjee S, Tian T, Wei Z, Shih N, Feldman MD, Alwine JC, Coukos G and Robertson ES: The ovarian cancer oncobiome. Oncotarget. 8:36225–36245. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu NN, Jiao N, Tan JC, Wang Z, Wu D, Wang AJ, Chen J, Tao L, Zhou C, Fang W, et al: Multi-kingdom microbiota analyses identify bacterial-fungal interactions and biomarkers of colorectal cancer across cohorts. Nat Microbiol. 7:238–250. 2022. View Article : Google Scholar : PubMed/NCBI | |
Dudeck A, Köberle M, Goldmann O, Meyer N, Dudeck J, Lemmens S, Rohde M, Roldán NG, Dietze-Schwonberg K, Orinska Z, et al: Mast cells as protectors of health. J Allergy Clin Immunol. 144:S4–S18. 2019. View Article : Google Scholar : PubMed/NCBI | |
De Zuani M, Paolicelli G, Zelante T, Renga G, Romani L, Arzese A, Pucillo CEM and Frossi B: Mast cells respond to candida albicans infections and modulate macrophages phagocytosis of the Fungus. Front Immunol. 9:28292018. View Article : Google Scholar : PubMed/NCBI | |
Abraham SN and St John AL: Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol. 10:440–452. 2010. View Article : Google Scholar : PubMed/NCBI | |
Renga G, Moretti S, Oikonomou V, Borghi M, Zelante T, Paolicelli G, Costantini C, De Zuani M, Villella VR, Raia V, et al: IL-9 and mast cells are key players of candida albicans commensalism and pathogenesis in the gut. Cell Rep. 23:1767–1778. 2018. View Article : Google Scholar : PubMed/NCBI | |
Gerard R, Sendid B, Colombel JF, Poulain D and Jouault T: An immunological link between Candida albicanscolonization and Crohn's disease. Crit Rev Microbiol. 41:135–139. 2013. View Article : Google Scholar : PubMed/NCBI | |
Liu B, Yang MQ, Yu TY, Yin YY, Liu Y, Wang XD, He ZG, Yin L, Chen CQ and Li JY: Mast cell tryptase promotes inflammatory bowel disease-induced intestinal fibrosis. Inflamm Bowel Dis. 27:242–255. 2021. View Article : Google Scholar : PubMed/NCBI | |
Nalleweg N, Chiriac MT, Podstawa E, Lehmann C, Rau TT, Atreya R, Krauss E, Hundorfean G, Fichtner-Feigl S, Hartmann A, et al: IL-9 and its receptor are predominantly involved in the pathogenesis of UC. Gut. 64:743–755. 2015. View Article : Google Scholar : PubMed/NCBI | |
Richardson JP, Moyes DL, Ho J and Naglik JR: Candida innate immunity at the mucosa. Semin Cell Dev Biol. 89:58–70. 2019. View Article : Google Scholar : PubMed/NCBI | |
Jiao Q, Luo Y, Scheffel J, Zhao Z and Maurer M: The complex role of mast cells in fungal infections. Exp Dermatol. 28:749–755. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hu SC, Yu HS, Yen FL, Lin CL, Chen GS and Lan CC: Neutrophil extracellular trap formation is increased in psoriasis and induces human beta-defensin-2 production in epidermal keratinocytes. Sci Rep. 6:311192016. View Article : Google Scholar : PubMed/NCBI | |
Miksch RC, Schoenberg MB, Weniger M, Bösch F, Ormanns S, Mayer B, Werner J, Bazhin AV and D'Haese JG: Prognostic impact of tumor-infiltrating lymphocytes and neutrophils on survival of patients with upfront resection of pancreatic cancer. Cancers (Basel). 11:392019. View Article : Google Scholar : PubMed/NCBI | |
Raftopoulou S, Valadez-Cosmes P, Mihalic ZN, Schicho R and Kargl J: Tumor-mediated neutrophil polarization and therapeutic implications. Int J Mol Sci. 23:32182022. View Article : Google Scholar : PubMed/NCBI | |
Que H, Fu Q, Lan T, Tian X and Wei X: Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochim Biophys Acta Rev Cancer. 1877:1887622022. View Article : Google Scholar : PubMed/NCBI | |
Triner D, Devenport SN, Ramakrishnan SK, Ma X, Frieler RA, Greenson JK, Inohara N, Nunez G, Colacino JA, Mortensen RM and Shah YM: Neutrophils restrict tumor-associated microbiota to reduce growth and invasion of colon tumors in mice. Gastroenterology. 156:1467–1482. 2019. View Article : Google Scholar : PubMed/NCBI | |
Nikou SA, Zhou C, Griffiths JS, Kotowicz NK, Coleman BM, Green MJ, Moyes DL, Gaffen SL, Naglik JR and Parker PJ: The Candida albicans toxin candidalysin mediates distinct epithelial inflammatory responses through p38 and EGFR-ERK pathways. Sci Signal. 15:eabj69152022. View Article : Google Scholar : PubMed/NCBI | |
Swamydas M, Gao JL, Break TJ, Johnson MD, Jaeger M, Rodriguez CA, Lim JK, Green NM, Collar AL, Fischer BG, et al: CXCR1-mediated neutrophil degranulation and fungal killing promote Candida clearance and host survival. Sci Transl Med. 8:322ra102016. View Article : Google Scholar : PubMed/NCBI | |
Wynn TA and Vannella KM: Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 44:450–462. 2016. View Article : Google Scholar : PubMed/NCBI | |
Strizova Z, Benesova I, Bartolini R, Novysedlak R, Cecrdlova E, Foley LK and Striz I: M1/M2 macrophages and their overlaps-myth or reality? Clin Sci (Lond). 137:1067–1093. 2023. View Article : Google Scholar : PubMed/NCBI | |
Biswas SK and Mantovani A: Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat Immunol. 11:889–896. 2010. View Article : Google Scholar : PubMed/NCBI | |
Condeelis J and Pollard JW: Macrophages: Obligate partners for tumor cell migration, invasion, and metastasis. Cell. 124:263–266. 2006. View Article : Google Scholar : PubMed/NCBI | |
Gu P, Liu R, Yang Q, Xie L, Wei R, Li J, Mei F, Chen T, Zeng Z, He Y, et al: A metabolite from commensal Candida albicans enhances the bactericidal activity of macrophages and protects against sepsis. Cell Mol Immunol. 20:1156–1170. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li J, Lan T, Zhang C, Zeng C, Hou J, Yang Z, Zhang M, Liu J and Liu B: Reciprocal activation between IL-6/STAT3 and NOX4/Akt signalings promotes proliferation and survival of non-small cell lung cancer cells. Oncotarget. 6:1031–1048. 2015. View Article : Google Scholar : PubMed/NCBI | |
Cheng CW, Kuo CY, Fan CC, Fang WC, Jiang SS, Lo YK, Wang TY, Kao MC and Lee AY: Overexpression of Lon contributes to survival and aggressive phenotype of cancer cells through mitochondrial complex I-mediated generation of reactive oxygen species. Cell Death Dis. 4:e6812013. View Article : Google Scholar : PubMed/NCBI | |
Prasad S, Gupta SC and Tyagi AK: Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 387:95–105. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Cao S, Toole BP and Xu Y: Cancer may be a pathway to cell survival under persistent hypoxia and elevated ROS: A model for solid-cancer initiation and early development. Int J Cancer. 136:2001–2011. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhu Y, Shi T, Lu X, Xu Z, Qu J, Zhang Z, Shi G, Shen S, Hou Y, Chen Y and Wang T: Fungal-induced glycolysis in macrophages promotes colon cancer by enhancing innate lymphoid cell secretion of IL-22. EMBO J. 40:e1053202021. View Article : Google Scholar : PubMed/NCBI | |
Dmitrieva-Posocco O, Dzutsev A, Posocco DF, Hou V, Yuan W, Thovarai V, Mufazalov IA, Gunzer M, Shilovskiy IP, Khaitov MR, et al: Cell-type-specific responses to interleukin-1 control microbial invasion and tumor-elicited inflammation in colorectal cancer. Immunity. 50:166–180.e7. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kirchberger S, Royston DJ, Boulard O, Thornton E, Franchini F, Szabady RL, Harrison O and Powrie F: Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J Exp Med. 210:917–931. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bollrath J, Phesse TJ, von Burstin VA, Putoczki T, Bennecke M, Bateman T, Nebelsiek T, Lundgren-May T, Canli O, Schwitalla S, et al: gp130-mediated stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer Cell. 15:91–102. 2009. View Article : Google Scholar : PubMed/NCBI | |
Keir ME, Yi T, Lu TT and Ghilardi N: The role of IL-22 in intestinal health and disease. J Exp Med. 217:e201921952020. View Article : Google Scholar : PubMed/NCBI | |
Kashem SW, Igyarto BZ, Gerami-Nejad M, Kumamoto Y, Mohammed JA, Jarrett E, Drummond RA, Zurawski SM, Zurawski G, Berman J, et al: Candida albicans morphology and dendritic cell subsets determine T helper cell differentiation. Immunity. 42:356–366. 2015. View Article : Google Scholar : PubMed/NCBI | |
Pittet MJ, Di Pilato M, Garris C and Mempel TR: Dendritic cells as shepherds of T cell immunity in cancer. Immunity. 56:2218–2230. 2023. View Article : Google Scholar : PubMed/NCBI | |
Schmidt S, Condorelli A, Koltze A and Lehrnbecher T: NK cells and their role in invasive mold infection. J Fungi (Basel). 3:252017. View Article : Google Scholar : PubMed/NCBI | |
Charpak-Amikam Y, Lapidus T, Isaacson B, Duev-Cohen A, Levinson T, Elbaz A, Levi-Schaffer F, Osherov N, Bachrach G, Hoyer LL, et al: Candida albicans evades NK cell elimination via binding of Agglutinin-Like Sequence proteins to the checkpoint receptor TIGIT. Nat Commun. 13:24632022. View Article : Google Scholar : PubMed/NCBI | |
Horii M and Matsushita T: Regulatory B cells and T cell regulation in cancer. J Mol Biol. 433:1666852021. View Article : Google Scholar : PubMed/NCBI | |
Wu J, Wang S, Zheng B, Qiu X, Wang H and Chen L: Modulation of gut microbiota to enhance effect of checkpoint inhibitor immunotherapy. Front Immunol. 12:6691502021. View Article : Google Scholar : PubMed/NCBI | |
Ahmadi N, Ahmadi A, Kheirali E, Yadegari MH, Bayat M, Shajiei A, Amini AA, Ashrafi S, Abolhassani M, Faezi S, et al: Systemic infection with Candida albicans in breast tumor bearing mice: Cytokines dysregulation and induction of regulatory T cells. J Mycol Med. 29:49–55. 2019. View Article : Google Scholar : PubMed/NCBI | |
Protti MP and De Monte L: Thymic stromal lymphopoietin and cancer: Th2-dependent and -independent mechanisms. Front Immunol. 11:20882020. View Article : Google Scholar : PubMed/NCBI | |
Alam A, Levanduski E, Denz P, Villavicencio HS, Bhatta M, Alhorebi L, Zhang Y, Gomez EC, Morreale B, Senchanthisai S, et al: Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell. 40:153–167.e11. 2022. View Article : Google Scholar : PubMed/NCBI | |
Jou E, Rodriguez-Rodriguez N, Ferreira AF, Jolin HE, Clark PA, Sawmynaden K, Ko M, Murphy JE, Mannion J, Ward C, et al: An innate IL-25-ILC2-MDSC axis creates a cancer-permissive microenvironment for Apc mutation-driven intestinal tumorigenesis. Sci Immunol. 7:eabn01752022. View Article : Google Scholar : PubMed/NCBI | |
Li GL, Tang JF, Tan WL, Zhang T, Zeng D, Zhao S, Ran JH, Li J, Wang YP and Chen DL: The anti-hepatocellular carcinoma effects of polysaccharides from Ganoderma lucidum by regulating macrophage polarization via the MAPK/NF-κB signaling pathway. Food Funct. 14:3155–3168. 2023. View Article : Google Scholar : PubMed/NCBI | |
Levy JMS and Magro C: Atrophying pityriasis versicolor as an idiosyncratic T cell-mediated response to Malassezia: A case series. J Am Acad Dermatol. 76:730–735. 2017. View Article : Google Scholar : PubMed/NCBI | |
Poggi A, Catellani S, Musso A and Zocchi MR: Gammadelta T lymphocytes producing IFNgamma and IL-17 in response to Candida albicans or mycobacterial antigens possible implications for acute and chronic inflammation. Curr Med Chem. 16:4743–4749. 2009. View Article : Google Scholar : PubMed/NCBI | |
Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, Verstegen NJM, Ciampricotti M, Hawinkels LJAC, Jonkers J and de Visser KE: IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 522:345–348. 2015. View Article : Google Scholar : PubMed/NCBI | |
Chao YY, Puhach A, Frieser D, Arunkumar M, Lehner L, Seeholzer T, Garcia-Lopez A, van der Wal M, Fibi-Smetana S, Dietschmann A, et al: Human TH17 cells engage gasdermin E pores to release IL-1α on NLRP3 inflammasome activation. Nat Immunol. 24:295–308. 2023. View Article : Google Scholar : PubMed/NCBI | |
Liang WF, Gong YX, Li HF, Sun FL, Li WL, Chen DQ, Xie DP, Ren CX, Guo XY, Wang ZY, et al: Curcumin activates ros signaling to promote pyroptosis in hepatocellular carcinoma HepG2 cells. In Vivo. 35:249–257. 2021. View Article : Google Scholar : PubMed/NCBI | |
Reina-Campos M, Scharping NE and Goldrath AW: CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol. 21:718–738. 2021. View Article : Google Scholar : PubMed/NCBI | |
Farhood B, Najafi M and Mortezaee K: CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review. J Cell Physiol. 234:8509–8521. 2019. View Article : Google Scholar : PubMed/NCBI | |
Picard FSR, Lutz V, Brichkina A, Neuhaus F, Ruckenbrod T, Hupfer A, Raifer H, Klein M, Bopp T, Pfefferle PI, et al: IL-17A-producing CD8+ T cells promote PDAC via induction of inflammatory cancer-associated fibroblasts. Gut. 72:1510–1522. 2023. View Article : Google Scholar : PubMed/NCBI | |
Han J, Khatwani N, Searles TG, Turk MJ and Angeles CV: Memory CD8+ T cell responses to cancer. Semin Immunol. 49:1014352020. View Article : Google Scholar : PubMed/NCBI | |
Pearce EL and Shen H: Making sense of inflammation, epigenetics, and memory CD8+ T-cell differentiation in the context of infection. Immunol Rev. 211:197–202. 2006. View Article : Google Scholar : PubMed/NCBI | |
Fernandes MR, Aggarwal P, Costa RG, Cole AM and Trinchieri G: Targeting the gut microbiota for cancer therapy. Nat Rev Cancer. 22:703–722. 2022. View Article : Google Scholar : PubMed/NCBI | |
Xu J, Chen F, Wang G, Liu B, Song H and Ma T: The versatile functions of G. Lucidum Polysaccharides and G. Lucidum Triterpenes in cancer radiotherapy and chemotherapy. Cancer Manag Res. 13:6507–6516. 2021. View Article : Google Scholar : PubMed/NCBI | |
Tian W, Huang J, Zhang W, Wang Y, Jin R, Guo H, Tang Y, Wang Y, Lai H and Leung EL: Harnessing natural product polysaccharides against lung cancer and revisit its novel mechanism. Pharmacol Res. 199:1070342024. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, Nie S, Huang D, Li W and Xie M: Immunomodulatory effect of Ganoderma atrum polysaccharide on CT26 tumor-bearing mice. Food Chem. 136:1213–1219. 2013. View Article : Google Scholar : PubMed/NCBI | |
Li A, Shuai X, Jia Z, Li H, Liang X, Su D and Guo W: Ganoderma lucidum polysaccharide extract inhibits hepatocellular carcinoma growth by downregulating regulatory T cells accumulation and function by inducing microRNA-125b. J Transl Med. 13:1002015. View Article : Google Scholar : PubMed/NCBI | |
Jing T, Guo Y and Wei Y: Carboxymethylated pachyman induces ferroptosis in ovarian cancer by suppressing NRF1/HO-1 signaling. Oncol Lett. 23:1612022. View Article : Google Scholar : PubMed/NCBI | |
Feng L, Yuan L, Du M, Chen Y, Zhang MH, Gu JF, He JJ, Wang Y and Cao W: Anti-lung cancer activity through enhancement of immunomodulation and induction of cell apoptosis of total triterpenes extracted from Ganoderma luncidum (Leyss. ex Fr.) Karst. Molecules. 18:9966–9981. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bin G and Gui-Zhen Y: Effects of Ganoderma applanatum polysaccharide on cellular and humoral immunity in normal and sarcoma-180 transplanted mice. Phytother. 5:134–138. 1991. View Article : Google Scholar | |
Wu Q, Yang Z, Nie Y, Shi Y and Fan D: Multi-drug resistance in cancer chemotherapeutics: Mechanisms and lab approaches. Cancer Lett. 347:159–166. 2014. View Article : Google Scholar : PubMed/NCBI | |
Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al: Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 371:596–602. 2021. View Article : Google Scholar | |
Routy B, Lenehan JG, Miller WH Jr, Jamal R, Messaoudene M, Daisley BA, Hes C, Al KF, Martinez-Gili L, Punčochář M, et al: Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: A phase I trial. Nat Med. 29:2121–2132. 2023. View Article : Google Scholar : PubMed/NCBI | |
Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al: Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 359:91–97. 2018. View Article : Google Scholar : PubMed/NCBI | |
Qian X, Zhang HY, Li QL, Ma GJ, Chen Z, Ji XM, Li CY and Zhang AQ: Integrated microbiome, metabolome, and proteome analysis identifies a novel interplay among commensal bacteria, metabolites and candidate targets in non-small cell lung cancer. Clin Transl Med. 12:e9472022. View Article : Google Scholar : PubMed/NCBI | |
Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, Segata N, Desnoyer A, Pietrantonio F, Ferrere G, et al: Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol. 78:195–206. 2020. View Article : Google Scholar : PubMed/NCBI | |
Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ and Gajewski TF: The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 359:104–108. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lu Y, Yuan X, Wang M, He Z, Li H, Wang J and Li Q: Gut microbiota influence immunotherapy responses: Mechanisms and therapeutic strategies. J Hematol Oncol. 15:472022. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Hong Y, Wu T, Ben E, Li S, Hu L and Xie T: Role of gut microbiota in regulating immune checkpoint inhibitor therapy for glioblastoma. Front Immunol. 15:14019672024. View Article : Google Scholar : PubMed/NCBI | |
Kohi S, Macgregor-Das A, Dbouk M, Yoshida T, Chuidian M, Abe T, Borges M, Lennon AM, Shin EJ, Canto MI and Goggins M: Alterations in the duodenal fluid microbiome of patients with pancreatic cancer. Clin Gastroenterol Hepatol. 20:e196–e227. 2022. View Article : Google Scholar : PubMed/NCBI | |
Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, Quesada P, Sahin I, Chandra V, Lucas AS, et al: Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell. 178:795–806.e12. 2019. View Article : Google Scholar : PubMed/NCBI | |
Apostolou P, Tsantsaridou A, Papasotiriou I, Toloudi M, Chatziioannou M and Giamouzis G: Bacterial and fungal microflora in surgically removed lung cancer samples. J Cardiothorac Surg. 6:1372011. View Article : Google Scholar : PubMed/NCBI | |
Banerjee S, Alwine JC, Wei Z, Tian T, Shih N, Sperling C, Guzzo T, Feldman MD and Robertson ES: Microbiome signatures in prostate cancer. Carcinogenesis. 40:749–764. 2019. View Article : Google Scholar : PubMed/NCBI | |
Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY and Yu J: Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 68:654–662. 2019. View Article : Google Scholar : PubMed/NCBI |