Abstract
Most studies on the human gut microbiome have focused on the bacterial fraction rather than fungal biomics, which as resulted in an incomplete understanding of the fungal microbiome. Recent advances in microbiota detection and next-generation sequencing technology have boosted an increase in research on fungi. Symbiotic fungi have become increasingly influential in health and disease and modulate various physiologic functions within the host. Fungal infections can result in high morbidity and mortality rates and are life-threatening in some immunocompromised patients. In addition to bacterial dysbiosis, alterations in fungal communities are important and have been linked to many diseases, including asthma, mental illness, and various cancers. When investigating cancer it is imperative to consider the role of fungi alongside viruses and bacteria. This review examined the impact of intestinal fungi and peri-tumor fungi on tumorigenesis, cancer progression, and response to anticancer therapies. The review highlights the specific involvement of some fungal species in cancers include digestive tract tumors such as colorectal, pancreatic, liver, and gastric cancers, as well as non-digestive tract tumors such as lung, melanoma, breast, and ovarian cancers. Furthermore, fungal mechanisms of action, including fungus-host recognition and immune regulation, biofilm formation, toxin and metabolite production in the tumor microenvironment, and the complex effects of fungus-bacteria interactions on tumorigenesis and development, highlight the significance of potential biomarkers in cancer diagnosis and treatment.
keywords
Introduction
Microbiomes exist in every human ecologic niche that has been examined, including the oral cavity, skin surface, intestinal tract, esophagus, and lungs. The microbiota are comprised of bacteria, archaea, viruses, phages, and fungi. Bacteria tend to be the most prominent microbiota, particularly with respect to species1. The microbiome is a term that usually refers to the community of various microorganisms that inhabit/live inside human/animal bodies or on the skin. The microbiome forms a complex ecosystem that includes trillions of commensals, symbiotics, and even pathogenic microorganisms2. A variety of bacteria, viruses, fungi, protists, archaea, and protozoa coexist within the mammalian gastrointestinal tract3. These microbes (bacteria, fungi, archaea, and viruses) are collectively referred to as the microbiota and are fundamental to physiology and immunity4. Mycobiota represent a subset of the human microbiome and have a significant role in the immune response. However, the specific role in human cancer has not been established. A pan-cancer analysis of multiple body sites has revealed the existence of tumor-specific mycobiomes comprised of up to 1 fungus per 104 tumor cells5,6. In recent years, pan-cancer analyses have suggested that the presence of some fungal communities may be associated with the development of cancer. For example, in gastrointestinal and lung tumors, specific fungal species, such as Candida tropicalis, C. albicans, and Blastomyces, have been identified. The DNA of these fungi is relatively abundant in some tumor types, suggesting that the fungi are involved in cancer onset or development7. The number of studies investigating the associations between cancer and mycobiota has increased steadily with a few studies uncovering potential mechanistic links between discrete fungal functions and cancer-related processes7,8.
The gut microbiota is a consortium of microorganisms that includes archaea, bacteria, fungi, and viruses3,5. Approximately 90% of all lethal fungi belong to 4 genera (Aspergillus, Candida, Cryptococcus, and Pneumocystis)3. Several fungal species, such as Candida, Aspergillus, Saccharomyces, Malassezia, Penicillium, Cladosporium and Cyberlindnera are associated with human diseases3,5,8–10, as shown in Table 1.
The gut is the most well-researched ecologic niche and the most frequently reported fungal taxa at this site include Candida, Saccharomyces, and Malassezia. Other body sites that have long been investigated for fungal colonization are the oral cavity, where the dominant taxa is Candida, the skin, which is mainly inhabited by Malassezia spp., and the vagina, which is primarily occupied by Candida spp. that may become pathogenic and drive vulvovaginal candidiasis5,10–12. The intestinal microbiota represents a complex ecosystem enriched with Saccharomyces and Candida. The balance of these microbial communities is crucial for maintaining gut health9,13. Candida is thought to be involved in cancer development because Candida is an opportunistic pathogen in an immunosuppressive state, especially during chemotherapy14. In addition, fungi and bacteria jointly form a complex microbial ecosystem within mammals, encompassing various body parts, such as the gastrointestinal tract, skin epithelium, respiratory tract, and reproductive organs. This ecosystem involves close interactions between microorganisms, as well as between microorganisms and their hosts. These interactions are of significant importance to human health5,6,11,15. Therefore, we have described the characteristics of fungi in tumor tissues and explored the relationship between fungi, fungal-bacterial interactions, and the growth, metastasis, and invasion of tumor cells, as well as the potential impact on treatment response and prognosis in patients.
Association between fungal dysregulation and the occurrence and development of multiple cancers
Compared to bacterial microbiota, research on fungal involvement in human carcinogenesis is still in the early stages. Although preliminary studies have indicated that some fungi may be associated with the occurrence and progression of cancer, little is known about the specific fungal mechanisms of action in the carcinogenesis process. This limitation may be attributable the relatively small distribution and quantity of fungi in the human body, as well as the different biological characteristics from bacteria, making studies more challenging8. This review provides a comprehensive analysis of relevant research on the relationship between fungi and cancer, particularly highlighting the gut, lung, liver, and pancreas as potential ecologic niches for fungal action. Fungi in these niches may participate in the development and occurrence of cancer through different mechanisms (Figure 1).
Fungal dysbiosis in colorectal cancer (CRC)
CRC is a common malignancy involving the colon and rectum and threatens the lives of approximately 1.8 million people every year16,17. Inflammatory bowel disease (IBD) increases the risk of developing CRC and over the past decade a significant understanding of the pathogenesis and treatment of colorectal tumors in IBD has been achieved3,18–20. CRC that occurs in individuals with IBD is often considered a prototype of inflammation-induced carcinogenesis18. Chronic inflammation results in oxidative stress-induced DNA damage, which may activate the oncogenes and inactivate the tumor suppressor genes. Markers of oxidative damage and DNA double-strand breaks gradually increase in the inflammation-dysplasia-carcinoma sequence18. IBD patients may develop sporadic tumors, with chronic inflammation being a key factor in risk stratification. Understanding this process helps manage inflammation in IBD patients, which in turn helps prevent cancer development.
Changes in fungal populations have been linked to CRC development16,21,22. Fungal dysbiosis occurs in patients with colon polyps and CRC with a higher diversity in patients with polyps compared to healthy individuals23. Intestinal fungal abundance is higher in the CRC group than in the polyp and control groups. C. albicans was reported to be the most abundant intestinal fungus in the cancer group24. An analysis of mucosal-adherent fungi showed that the three major fungal phyla (Ascomycota, Basidiomycota, and Glomeromycota) were present in all samples8. Most fungi observed in the studies were from Ascomycetes8,21,22,25 followed by Basidiomycota was second in both the case and control groups. The Basidiomycetes-to-Ascomycetes ratio was higher in the CRC group than the control group8,17,22,26, indicating that fungal dysbiosis increases with disease progression26. Differences in fungal composition were observed between colorectal adenomas and neighboring tissues along with a decrease in diversity in the adenomas8,17,22. There was a significant reduction in Saccharomycetes and Pneumocystis fungi in CRC, with an enrichment of Malasseziomytes22,26. Six fungi genera, including Acremonium, Malassezia, Moniliophthtora, Rhodotorula, Thielaviopsis, and Pisolithus, occur in individuals with CRC22,26. Enrichment of the above six genera resulted in changes in the composition of CRC-specific fungi22,26. In a study involving the fungal core group, one group consisted of 10 fungi enriched in CRC, while the other group was comprised of 23 fungi depleted in CRC21. Aspergillus rambellii is the most enriched fungus, while A. kawachii is the most depleted fungus in CRC when compared to healthy subjects8,22. Twenty-four fungal species with varying levels of abundance were identified during screening among patients with CRC and adenomas. Among these 24 fungi, 1 species was enriched in CRC (A. rambellii), while 6 species were depleted in CRC (A. kawachii, Fusarium pseudograminearum, Lentinula edodes, Hanseniaspora guilliermondii, Saitoella complicata, and Cryptococcus neoformans). Compared to adenoma patients, the abundance of some fungi changed by > 1.4-fold compared to patients with adenomas. A total of 10 fungal species with differential abundances were identified between adenoma patients and healthy individuals. Among the 10 fungi, 5 species had an increase in abundance of at least 1.5-fold in adenoma patients21. Moreover, there was a significant increase in the abundance of C. albicans in CRC patients8,24. Overall, the number of opportunistic pathogens increased and the biodiversity changed in the disease group. Candida may, in some cases, promote or accelerate the process of disease metastasis. This may be because Candida can affect immune system function, thus increasing the risk of disease metastasis. Alternatively, Candida may directly disrupt the tissue barrier, providing a pathway for disease transfer. The presence of Candida may result in decreased cell adhesion. This may be because Candida can secrete acetaldehyde and candidalysin that interfere with cell-to-cell connections, leading to a loosening of the tight bonds between cells. Reduced cell adhesion may impair tissue integrity and make it more vulnerable to disease. Thus, Candida is considered a predictor that its presence can predict certain adverse outcomes7.
The changes in abundance of these fungi in CRC patients may provide new clues that help us understand the pathogenesis of CRC. Depletion or enrichment of these fungi may be closely related to the occurrence and development of CRC and potentially serve as a biomarker for CRC or participate in the pathologic process underlying CRC.
Thirteen fungi, listed as key biomarkers for the diagnosis of CRC, had a positive association in all samples25,27. This finding indicates that in addition to bacteria, fecal fungi could also serve as an important reference for the non-invasive diagnosis of CRC patients. Significant differences in the abundance of some fungal species between patients with CRC and healthy individuals can be detected based on fecal sample analysis. These fungi may be closely related to the occurrence and development of CRC with the potential to become new biomarkers for CRC diagnosis21,22.
In a colon tumor containing C. albicans, the tumor is characterized by downregulated transcription of tumor suppressor genes, extracellular matrix receptor interactions, and genes associated with focal adhesion8.
Fungal dysbiosis in pancreatic cancer
Fungi may enter the pancreas from the intestinal cavity through several pathways and subsequently proliferate. This abnormal colonization of fungi may be closely related to the occurrence and development of pancreatic ductal adenocarcinoma (PDA)26,28,29. In animal models of PDA and human studies involving PDA the fungal content in the pancreas is significantly increased by approximately 3000 times compared to the pancreas in normal physiologic states3,28. Specifically, the fungal communities infiltrating PDA tumors are rich in Malassezia species in rodents and humans3,19,28,30,31. The presence of mannose-binding lectin (MBL), which binds to fungal wall glycans to allow complement cascade activation, is responsible for neoplastic promotion, while MBL or C3 deletion in the extra-tumoral compartment or C3aR knockdown in cancer cells has tumor-protective effects, even in the presence of Malassezia, which leads to PDA promotion via the complement cascade through MBL activation3,19,26–28,30,32–34. Another study showed that intra-tumoral fungi (Malassezia globosa or Alternaria alternata) enhance IL-33 secretion in PDA cancer cells through the dectin-1-mediated activation of the non-receptor tyrosine kinase (src)-spleen tyrosine kinase (Syk)-caspase recruitment domain protein 9 (CARD 9)-nuclear factor kappa B (NF-κB) pathway. Secreted IL-33 recruits and activates Th 2 and ILC 2, which stimulates tumor growth through secretion of tumor-promoting cytokines, such as IL-4, IL-5, and IL-13 in the tumor microenvironment (TME)35. Other pro-inflammatory cytokines, including IL-18, IL-8, IL-6, and Th22, and chemokines, including C-C motif chemokine ligand 17 (CCL17), were also significantly increased after exposure to Malazzia, thereby exacerbating local inflammation36. Malassezia colonization has been shown to induce CARD9-S12N polymorphism and to strongly enhance the release of cytokines, such as IL-10 or tumor necrosis factor-alpha (TNF-α) in wild-type (WT) or Card9−/− colitis mice compared to negative groups37. In addition, Malassezia can activate the NLRP3 inflammasome through Dectin2/CARD9 signaling, accelerating the production of IL-1β and aggravating inflammation36. Malassezia also promotes degradation of the extracellular matrix (ECM). Malassezia may promote tumor invasion and metastasis, in which integrin aggregation promotes focal adhesion, strengthens the ERK and PI3K pathways, and promotes cell proliferation and invasion36.
More research is needed to determine the role of fungi in PDA. This discovery opens up new avenues for PDA diagnosis and treatment. For example, could detecting fungal content in the pancreas assist in PDA diagnosis? Could antifungal treatments help to intervene in PDA progression? These questions warrant further investigation in future studies.
Fungal dysbiosis in liver cancer
The gut and liver communicate bidirectionally via the biliary system and the portal vein. The gut microbiota and microbial products have a critical role in modulating liver function32,38.
In comparison to healthy controls and cirrhotic patients, hepatocellular carcinoma (HCC) patients exhibit intestinal fungal dysregulation and significant enrichment of opportunistic fungi, such as Malassezia and C. albicans. Alpha diversity analysis revealed reduced fungal diversity in HCC and cirrhosis patients compared to healthy controls. Beta diversity analysis showed a notable separation clustering among the three groups32. Among these groups, the abundance of C. albicans was significantly higher in stage III–IV HCC patients compared to stage I–II32. C. albicans reprograms HCC metabolism and promotes NLRP6-dependent HCC progression39. HCC can also be caused by M. furfur3,32.
It is known that aflatoxins (a fungal toxin produced by the secondary metabolism of A. favus and A. parasiticus) induce HCC32,40. Aflatoxins are usually ingested and cleared quickly from the blood. Experiments have shown that AFB1 damages intestinal epithelial cells, leading to destruction of intestinal structures40. Metabolism of the CYP450 system forms DNA adducts that lead to mutations in the p53 tumor suppressor gene, a key mechanism in tumor development. AFB1 also induces ROS, inhibits protein, RNA, and DNA synthesis, and ultimately leads to apoptosis of microglia by activating NF-κB during oxidation38,40,41.
Fungal dysbiosis in gastric cancer
The hallmark of GC-associated mycobiome imbalance is alteration in fungal composition and ecology. C. albicans dysbiosis is associated with gastric carcinogenesis34. Candida and Alternaria are significantly increased in GC, suggesting an important role in distinguishing between GC and non-GC tissues and potentially serving as accurate biomarkers3,8,30.
Fungal dysbiosis in lung cancer
The lungs were once thought to be sterile in a healthy state, leading to research into the lung microbiome to lag behind other aspects of human microbiomics5,42–45. Blastomyces dermitidis/gilchristii are associated with tumor tissue in lung cancer6–8. Researchers found evidence of Blastomyces DNA in 6 out of 50 patients with squamous cell lung cancer6. Intratumor mycobiome dysbiosis is correlated with lung adenocarcinoma (LUAD) pathology and is enriched with a live fungus (A. sydowii). Tumor-resident A. sydowii promotes lung tumor progression by inducing an immunosuppressive TME46.
Fungal dysbiosis in other cancers
Narunsky-Haziza et al.7 characterized cancer mycobiomes in tissues and blood. All tumor types (breast cancer, lung cancer, melanoma, ovarian cancer, colon cancer, GBM, bone cancer and pancreatic cancer) tested had a higher fungal load than negative controls and the fungal load differed among tumor types. Fungi in human tumors have been observed by staining microarrays of melanoma and pancreas, breast, lung, and ovarian cancer tissues. Interestingly, images showed cancer type-specific localization patterns. Although fungal staining was mainly evident within cancer cells in pancreas, breast, and ovarian cancer, staining was mostly localized to macrophages in melanoma and lung cancer. The associations of fungi with clinical parameters, including overall survival in breast cancer, progression-free survival in ovarian cancer, the immunotherapy response in melanoma tumors, and detection of early-stage cancers, support the clinical utility as potential biomarkers and therapeutic targets7.
Potential roles of fungi in the pathogenesis of cancer
The gut microbiota, including the bacterial and viral communities, undergoes dynamic maturation from birth47. The immune system is an important defense line that is responsible for identifying and resisting viruses, bacteria, and other foreign pathogens12. Miranda et al.48 extracted Candida from the blood of patients with candidiasis and compared the Candida obtained from the rectal area or skin of the same patient. In most cases, C. albicans strains in blood sample-matched strains were present in the rectal samples obtained from these patients. These findings support a model in which commensal organisms from the gastrointestinal tract can translocate from this niche and to the bloodstream48–50.
Upon reaching organs through the bloodstream, C. albicans first adheres to endothelial cells in the form of blastoconidia or hypham51. C. albicans and its mannoproteins activate hepatic sinusoidal endothelial mainly through mannose receptor, and as a consequence, trigger a cytokine-mediated pro-inflammatory response that increases tumor cell adhesion to the hepatic endothelium and metastasis51. C. albicans stimulates endothelial cells to synthesize TNF-α, which further induces these infected cells to secrete IL-8. Additionally, through an autocrine mechanism, these endothelial cells express e-selectin, an essential cell adhesion molecule that facilitates the adhesion of white blood cells to vascular endothelial cells, further promoting progression of the inflammatory response. IL-8, a pro-inflammatory cytokine that has a crucial role in inflammation and immune response generation, is particularly known for its pro-inflammatory effects on neutrophils. When intestinal epithelial cells are stimulated by hyphae, intestinal epithelial cells release IL-8 as part of the natural immune response. However, excessive inflammatory reactions can be harmful to the body. For example, in IBDs, intestinal epithelial cells are constantly exposed to an inflammatory environment, leading to the excessive secretion of pro-inflammatory cytokines, such as IL-847.
Vascular cell adhesion molecule-1 [VCAM-1 (CD106)] is a membrane protein that contributes critical physiologic functional roles in the cellular immune response, including leukocyte extravasation in inflamed and infected tissues52. The combined activities of TNF-α, IL-1α, and IL-1β significantly enhance VCAM-1 expression in endothelial cells. This combined effect is likely achieved through the activation of specific signaling pathways, which can influence the expression and transcription of the VCAM-1 gene51,53. Mature IL-1β stimulates IL-18 release, which increases VCAM-1 expression and cancer cell adhesion54. The effect of VCAM-1 on the metastatic potential of tumors was first demonstrated in the adhesion of melanoma cells to endothelial cells. Since then, VCAM-1 overexpression has been related to metastasis of multiple adult-onset cancers, including gliomas and breast, ovarian, and colorectal cancers, and VCAM-1 was negatively correlated to patient prognosis52. Newly disseminated cancer cells expressing VCAM-1 thrive in leukocyte-rich microenvironments through juxtacrine activation of a VCAM-1-Ezrin-PI3K/Akt survival pathway55, as shown in Figure 2. Tumor expression of VCAM-1 might promote T cell migration away from tumors, thereby minimizing the contact between T and tumor cells. The decreased accumulation of T cells around tumor cells might contribute to the ability of tumor cells to escape immune attack56. VCAM1 expression is upregulated in human CRC tissues compared to matched adjacent normal intestinal epithelium. In addition, VCAM1 significantly affects the invasion and metastasis of CRC cells in vitro and in vivo and activates the EMT program by which cancer cells adhere to the endothelium and cross the blood vessel wall by forming pseudopodia and invasive pseudopodia57.
Secretion of fungal carcinogenic metabolites
In 2016 the first cytolysin toxin produced by the human pathogenic fungus, C. albicans, was given the name candidalysin8. Candidalysin is a cytolytic enzyme that damages the host cell membrane by forming a porous structure, resulting in the release of LDH and the flow of calcium into the cytoplasm, which damages the host cell membrane. These signaling pathways trigger hazard response signaling pathways that activate epithelial immunity and the secretion of IL-1β by macrophages58. C. albicans and candidalysin activate the human epithelial EGFR receptor, whereas fungal mutants lacking candidalysin exhibit a weaker ability to induce EGFR phosphorylation during oropharyngeal candidiasis in mice59. EGFR activation leads to the induction of the c-Fos transcription factor in the candidalysin-induced mitogen-activated protein kinase (MAPK) signaling pathway, ultimately triggering the recruitment of neutrophils via G-CSF and GM-CSF, as shown in Figure 2. This process depends on the release of EGFR ligands and an influx of calcium40,47,59. In addition, candidalysin indirectly activates EGFR through a complex mechanism of matrix metalloproteinases and EGFR ligands, leading to downstream immune activation60. EGFR may be activated in an atypical manner, thereby affecting intracellular processes when cells are exposed to stress stimuli, such as transport, autophagy, and energy metabolism. This atypical EGFR activity may be related to cancer cell survival and treatment resistance. Specifically, atypical EGFR activity may help cancer cells adapt to environmental changes, enhance the ability to survive, and reduce the sensitivity to anticancer drugs. One of the best-studied metabolic changes in cancer cells is the increase in glycolysis, even in the presence of oxygen, which is also known as the Warburg effect61. Lim et al.62 reported that EGF signaling activates the first step of glycolysis but blocks the last step, leading to the accumulation of metabolic intermediates in this pathway. In addition, Lim et al.62 showed that one of these intermediates, fructose 1,6 diphosphate (F1,6BP), directly binds and enhances the activity of EGFR, thereby enhancing the inhibition of local cytotoxic T cell activity caused by a lactic acid overdose, which promotes cancer cell growth and metastasis62. The PI3K signaling pathway regulates key metabolic activities in EGFR-mutated LUAD. The PI3K/AKT/mTOR signaling pathway is crucial for regulating aerobic glycolysis in EGFR-mutated LUAD cells. EGFR activation triggers a series of reactions that activate PI3K, further activating the PI3K/AKT/mTOR signaling pathway. This signaling pathway provides tumor cells with a significant amount of energy and biosynthetic precursors required for tumor growth and invasion63. Compelling evidence has shown that Akt, a serine/threonine kinase downstream of PI3K, is involved in the Warburg effect64. Active Akt inhibits autophagy and blocks GLUT1 endocytosis. The latter results in increased GLUT1 levels at the plasma membrane, which increases glucose uptake61. In cancer cells, Akt induces GLUT1 and GLUT3 expression and is an established mediator for the translocation of GLUT4 to the plasma membrane, which stimulates glucose transport64.
C. albicans also produces nitrosamines and metabolizes ethanol into acetaldehyde3. Nitrosamines activate specific proto-oncogenes that trigger abnormal proliferative changes in oral epithelial cells or cancers65. Acetaldehyde, a carcinogenic substance, can cause mutations by binding directly to DNA and proteins, induce oxidative stress, and lead to DNA damage and genetic mutations40, as shown in Figure 2. Acetaldehyde treatment induces oxidative stress and Ca2+ overload in SH-SY5Y cells, leading to the phosphorylation of Drp1 at Ser616 and translocation of Drp1 to mitochondria, subsequently causing excessive fission and fragmentation of mitochondria. The resulting imbalance of mitochondrial dynamics eventually impairs mitochondrial function and causes cytotoxicity. The mitochondrial electron transport chain is the major source of intracellular ROS. When the mitochondrial respiratory chain complex is damaged, ROS can be overproduced and accumulated, causing oxidative stress and triggering more mitochondrial alterations (e.g., ΔΨm disruption and ATP depletion)66.
Bacterial species and fungi often form multi-microbial biofilms40. These biofilms act as a barrier that protects microbes from the host immune system and potentially promotes local inflammatory responses3,8,28. Studies show that mucosal biofilms in the colon directly trigger CRC3,8. Therefore, biofilms are strongly associated with CRC because biofilms evade host immune responses, increase epithelial cell permeability or promote pre-cancerous inflammation, and have a strong ability to invade and spread3,40.
Fungi activate the immune response
The immune system detects ubiquitous fungi and requires a good balance between pro- and anti-inflammatory signals to maintain a stable host-fungal relationship. A disruption of this balance can lead to pathologic consequences67. Fungi are usually effectively cleared by a functioning immune system. However, patients with impaired immune systems and patients receiving immunosuppressive therapy have increased susceptibility to fungal infections, which are associated with higher mortality60,68. By masking or subverting the host detection systems, fungi may avoid inflammation, which contributes to fungal adaptation and opportunism67. The cell wall of C. albicans is rich in various complex carbohydrate structures, including chitin, glucan, and mannan. These components can all be considered pathogen-associated molecular patterns (PAMPs). PAMPs are recognized by the host immune system during the infection process, triggering a series of immune responses19,69,70. A PRR is a multiline-encoded receptor that recognizes a variety of pathogen-associated molecules expressed by invading microorganisms (PAMPs)68. Four receptor families can help us identify a wide range of species, including fungi68,69.
After the recognition of PAMPs by PRRs, PRRs activate a series of complex intracellular signaling pathways. These signaling pathways involve key molecules, such as adaptor proteins, kinases, and transcription factors, which together convert external signals into intracellular biochemical reactions. The signals induced by PRRs activate gene expression and promote the synthesis of a range of molecules, including cytokines, cell adhesion molecules, and immune receptors, as shown in Figure 3.
The signals induced by PRRs not only help recruit and activate more immune cells but also enhance the ability of cells to kill pathogens14. C-type lectin domain family 7 member A (Dectin-1), CARD9, IL-17, and IL-22 have been identified as key components in the host defense against fungal infections. Mutations in these molecular components have been linked to increased susceptibility to fungal infections in humans12. CLRs, such as Dectin-1, -2, and Mincle, have a major role in anti-fungal immunity because CLRs are specific to various carbohydrate-rich moieties in the fungal cell wall47. Dectin-1 is a well-defined CLR that recognizes beta-1,3-glucan present in the cell walls of Aspergillus, Candida, and many Saccharomyces and Trichosporon68,71. When Dectin-1 recognizes and binds to specific components on the fungal cell wall, Dectin-1 signals through the Syk-CARD9-nuclear factor-κB (NF-κB) axis to promote the assembly of the inflammasome, driving the production of pro-IL-1β and pro-IL-18. During this process, when the NLRP3 inflammasome is activated, the NLRP3 inflammasome forms a complex with caspase-1 or in some cases, caspase-8. This complex cleaves the pro-IL-1β and pro-IL-18 fragments, converting them into mature forms, such as IL-1β and IL-1823,40. Bergmann et al.72 reported that IL-1β regulates the production of IL-22 by group 3 innate lymphoid cells (ILC3s), ultimately promoting tumorigenesis by activating the epithelial signaling transducer and activator of transcription 3 (STAT3). C. albicans promotes the proliferation of intestinal epithelial cells (IECs) in a Dectin-1- and WNT-dependent manner through its interaction with Dectin-1 in IECs. The abnormal activation of the Wnt/β-catenin signaling pathway is closely related to the tumorigenesis of multiple organs, including the abnormalities triggered by mutations in genes, such as APC or CTNNB1 (encoding β-catenin)23,24,73,74, as shown in Figure 2. C. albicans infection in mouse colorectal tissue activates the Wnt pathway. IECs may recognize C. albicans activation of the Wnt pathway through Dectin-1, thus promoting the occurrence of CRC. C. albicans induces the proliferation of IECs by activating the Wnt signaling pathway and the Wnt pathway contributes to the development of CRC24. Wnt binds to Frizzled receptor and attracts LRP binding, which then undergoes phosphorylation. After Dvl binds to phosphorylated LRP, Axin is attracted to LRP, leading to disruption of the destruction complex. This disruption blocks the β-TrCP ubiquitination, resulting in the stabilization of β-catenin. Stabilized β-catenin is transported to the nucleus to activate transcription factors, which regulate target genes and ultimately lead to cell proliferation and differentiation75. It should be noted that the specific mechanism by which fungi directly activate the Wnt pathway is not fully understood. While most studies have focused on Wnt signaling in mammalian cells, fungal activation of the Wnt pathway may involve more complex interaction and regulatory networks. In addition, different species of fungi may activate the Wnt pathway through different mechanisms, so specific cases need to be analyzed.
Genetic polymorphisms in CLEC7A, which encodes the antifungal receptor, Dectin-1, are associated with ulcerative colitis severity in humans12,19,71. A Dectin-1 deficiency increases the severity of dextran sodium sulphate (DSS) colitis, which enhances the general expansion of fungi, such as Candida and Trichomonas, by suppressing the immune response to pathogens12,19,47,71,76,77. CLEC7A−/− mice experienced weight loss and histologic changes, manifesting as mucosal erosion, crypt destruction, infiltration of inflammatory cells, and increased production of TNF-α, IFN-γ, and IL-17 in the colon71. TNF-α exhibits tumorigenic characteristics by activating representative c-JunN terminal kinase (JNK) and NF-κB signaling pathways, leading to enhanced epithelial-to-mesenchymal transformation (EMT) and accelerating tumor cell invasion36. Antifungal therapy eliminates this susceptibility, further supporting the important role of intestinal fungal immunity in colitis5,12.
Increased tumorigenesis is associated with the C. albicans burden in Dectin-3−/− mice. The increase in abundance of C. albicans induces upregulation of macrophage glycolysis through the HIF-1 pathway, leading to the secretion of IL-7. IL-7 then induces the production of IL-22 by RORγt ILC3s through the aryl hydrocarbon receptor and STAT33,23,40,78. CARD9 is a signaling molecule downstream of Dectin-1 and Mincle, Dectin-3, DCIR, DC-SIGN, and Lox-1, and CARD9 loss has also been linked to human susceptibility to IBD8,12,19,71.
CARD9 deficiency results in impaired bactericidal ability and increased fungal burden, especially of C. tropicalis. Subsequently, CARD9 deficiency leads to increased accumulation of myeloid-derived suppressor cells (MDSCs), which suppress effector T cells and promote the development of CRC3,19,23,78,79. C. tropicalis induces NLRP3 inflammasome activation in MDSCs via Dectin-380. Mannans are highly conserved components in the fungal cell wall where Candida induces Th17 and IL-23 responses by activating the PRRs, thereby exacerbating GVHD and mouse colitis76. The mannoprotein portion of C. albicans has specifically been shown to enhance tumor adherence by stimulating endothelial cells and inflammation14,51. Blocking the mannose receptor reduces the ability of C. albicans to influence tumor cell adhesion and inhibits IL-18 synthesis8,14,51.
Colonization of the stomach in mice by C. albicans leads to the expansion of regulatory T cell populations, which are associated with immunosuppressive effects in the host48. C. albicans increases the production of IL-17 and IL-23 in the stomach and oral tissues in mice48. IL-23 promotes angiogenesis, which contributes to tumor initiation and growth. Furthermore, this cytokine antagonizes IL-12 and IFN-γ, both of which are crucial in Th1-mediated antitumor immune responses14. Thus, Candida colonization can enhance the inflammatory response by increasing the levels of these cytokines. Th17 cells are known to produce IL-17, which is necessary to fight off C. albicans. Although the pathologic mechanisms mediated by Il-17 are unknown, the role of IL-17 in inducing antimicrobial peptides, pro-inflammatory cytokines, and downstream neutrophil recruitment is considered significant60. IL-17 promotes gastrointestinal tumorigenesis by binding to its receptor and this signal activates ERK, p38 MAPK, and NF-κB signaling pathways. IL-17 stimulates the proliferation of tumorigenic enterocytes that just lost expression of the APC tumor suppressor, as well as the proliferation of colon epithelial cells, further supporting the malignant transformation of mice81.
Interactions between fungi and other microorganisms in cancer
Fungal-bacterial interactions in cancer
Symbiotic fungi and bacteria co-exist in the gut in a complex and delicate ecosystem. The interaction patterns between these microorganisms in the colon are crucial for maintaining intestinal health. Therefore, studying the interaction patterns of symbiotic fungi and bacteria in the gut, as well as the potential roles in CRC, is of great significance for understanding the pathogenesis of CRC and developing new treatment methods. A complex network of interactions among microbes across kingdoms can influence cancer behavior and treatment responsiveness8.
In lower gastrointestinal tract tumors, there is a positive correlation between intratumoral Candida and Dialister, while a negative correlation exists with Ruminococcus, Akkermansia, and Barnesiella intestine-hominis8. There were fewer and mostly positive correlations between fungi and bacteria in the control group compared to the CRC group, and a stronger co-exclusion relationship was observed in CRC22. Compared to individuals with adenomas and healthy individuals, the co-occurrence interactions among fungi enriched in CRC are stronger in CRC. Similarly, the strength of fungus-bacterial interactions increases as CRC progresses21. As a control group, fungi and bacteria have a synergistic association.
Some novel inter-kingdom interactions that involve various fungal classes, such as Pucciniomycetes, Exobasidiomycetes, Malasseziomycetes, and the Mucoromycota phylum, have been discovered in CRC. These fungi form an important co-exclusive network with the primary bacterial species, suggesting an ecologic imbalance in the gut microbiota in CRC. Specifically, cross-kingdom co-occurrence interactions involving fungi, such as Leotiomycetes, sodium-absorbing fungi, and Eurotiomycetes, were reversed to co-exclusive interactions in CRC. This shift may indicate a disruption in gut microbiota homeostasis, probably resulting from a balanced and synergistic relationship between different kingdoms. The disruption of this relationship may further contribute to colorectal tumor development22. Conversely, co-occurrence interactions were observed between fungi (Chaetomiaceae) and bacteria (Ruminicoccaceae), as well as between fungi (Pseudeurotiaceae) and bacteria (Alteromonadaceae and Geobacteraceae). These interactions between symbiotic kingdoms contribute significantly to the homeostasis of colonic microbiota. The microbial community in the colon represents a complex ecosystem, where diverse microorganisms work synergistically to maintain the balance of the intestinal environment. This balance is crucial for human health. In the context of CRC, the interaction between bacteria and fungi becomes particularly complex. With the enrichment of bacteria, the previously positive correlation between CRC-enriched fungi (Chaetomiaceae) and CRC-lacking bacteria (Ruminococcaceae), as well as between CRC-enriched fungi (Pseudourotiaceae) and CRC-lacking bacteria (Alteromonadaceae), is no longer present. This finding reveals the unique changes in the gut microbiota in CRC. Such changes may indicate an imbalance or reconfiguration of the gut microbiota during the occurrence and development of CRC. Instead, the positive correlation between the fungus, Pseudourotiaceae, and the bacterium, Geobacteraceae, reverses to a negative correlation, despite both being CRC-enriched22.
A hallmark of gastric tumors is that Lactobacilli are only associated with Candida clusters, which also tend to be isolated from H. pylori. The significant positive correlation between Streptococci and Clostridia with Candida indicates the potential occurrence of a symbiotic or collaborative relationship between these organisms. This relationship may promote the growth and reproduction in the gastric tumor environment. However, the significant negative correlation between Streptococci and Clostridia with Saccharomyces cerevisiae suggests a competitive or inhibitory relationship between these two types of microorganisms in the gastric tumor environment. The decrease in yeast populations may be related to the occurrence and development of gastric tumors, but specific mechanisms require further investigation8. Fusobacterium nucleatum is a bacterium often found in the intestines of patients with CRC. F. nucleatum is closely related to the occurrence and development of CRC. The close association between A. rambellii and F. nucleatum further suggests that A. rambellii and F. nucleatum may collaborate or influence each other during the pathogenesis of CRC. Additionally, A. rambellii has been shown to promote the growth of CRC cells in vitro and facilitate tumor growth in xenograft mice, providing a new perspective on the pathogenesis of CRC. This finding indicates that modulating the intestinal microbial community, especially targeting key microorganisms, such as A. rambellii and F. nucleatum, may aid in the prevention and treatment of CRC21. The diagnostic panel utilizing a combination of fungal and bacterial markers has demonstrated higher accuracy for the diagnosis of intestinal diseases, emphasizing the potential value and importance of intestinal fungi in clinical applications21,82.
The presence and changes in fungi and bacteria in the TME have been widely studied. These microorganisms have been detected in the intestine, other mucosal surfaces, and within tumors, with community composition potentially evolving alongside cancer development. These changes may be related to the occurrence, development, and metastasis of cancer. However, current research on cross-kingdom interactions between fungi and bacteria in cancer is still in its infancy, and many mechanisms are not yet fully understood. Therefore, more research is needed to explore this field in depth to understand and address cancer as a global health issue more effectively.
Fungal-virus interaction in cancer
To date, the interaction between viruses and fungi remains poorly studied. The intestinal fungal diversity of patients with hepatitis B cirrhosis was higher than that of patients with chronic hepatitis B, which was higher than that of HBV carriers and healthy volunteers. There was no significant difference in intestinal fungal diversity between HBV carriers and healthy volunteers. The diversity of intestinal fungi was positively correlated with disease progression in patients with different degrees of chronic HBV infection83. The abundance of fungal species was significantly reduced in the HIV/HCV co-infected group compared to the healthy control group, while no significant difference was found in the single-infected group. The opportunistic pathogenic fungal spectrum and fungal inter-relationships in the co-infected group became less characteristic but more complex than in the single-infected group. Intestinal fungal disorders occur in HIV- and HCV-infected patients and this disorder is further complicated in HIV/HCV co-infected patients. With the existence of some new fungal genera, some unique fungal interrelationships have emerged, which can be either positive or negative84. In this era of efficacious antiretroviral therapy and consequent immune reconstitution, oropharyngeal and esophageal candidiasis remain two clinically relevant presentations in the global HIV setting. Both diseases are predominantly caused by C. albicans. HIV infection generates an environment selected for overexpression of the virulence potential by the fungus, particularly concerning the secreted aspartyl proteinases (Saps). For these reasons, new therapeutics targeting virulence factors and specific immune interventions are being addressed85.
Interaction between cancer treatment responsiveness and the mycobiome
Symbiotic fungi regulate radiotherapy response through interaction with immune cells in the TME
It is well-established that tumor-associated immune cells rely on an immune microenvironment that promotes or hinders tumor progression. This microenvironment can be regulated in part by interactions between the immune system and the microbiome86. Other components of the gut microbiome, particularly fungi, have recently been found to modulate inflammatory responses in various diseases, including colitis, asthma, and inflammatory colon cancer, because of the effects on dendritic and myeloid cells86.
The effect of radiotherapy is adversely affected by the presence of symbiotic bacteria and fungi in the gut8,86. The elimination of specific bacteria by antibiotics can significantly diminish the effectiveness of chemotherapy or immunotherapy, while the elimination of fungi may improve the efficacy of radiation therapy86. Symbiotic fungi can bind to the receptor, Dectin-1, upregulating tumor-promoting macrophages and downregulating anti-tumor T cells, thus inhibiting the anti-tumor immune response after radiotherapy8,86.
Active metabolites secreted by fungi indirectly influence the efficacy of immunotherapy by regulating the host immune system
Immune checkpoint inhibitors (ICIs) represent a breakthrough in cancer treatment87. ICIs significantly improve patient outcomes in various tumors, but the efficacy has been inconsistent88. In recent years increasing evidence has pointed out that the composition of intestinal bacteria can affect the effectiveness of ICIs, but the predictive accuracy is unsatisfactory87,89. In contrast, the role of intestinal fungi in predicting the efficacy of tumor immunotherapy is more accurate than intestinal bacteria87.
Recent studies have shown that the imbalance in intestinal flora caused by antibiotics may affect the efficacy of ICIs88,90,91, the development of tolerance to PD-1 inhibitors, and significantly reduce progression-free survival and overall survival rates90,91. Using fecal DNA analysis techniques, such as 16S and shotgun sequencing, can help uncover significant differences in microbiota composition between responsive and non-responsive patients. This disparity not only provides clues about the potential role of microbiota in tumor immunity but also lays the foundation for further investigating the mechanistic links between microbiota and anti-tumor immunity89. The development of tumors is intricately connected to the immune system. The significant systemic impact of gut microbiota on tumor progression and treatment is likely mediated through immune system modulation. The gut microbiota is indeed an extremely complex system that profoundly affects the immune system not only through direct interactions with host cells but also via the release of various metabolites into the circulation. Specifically, short-chain fatty acids (SCFAs), important metabolites in the gut microbiota, have particularly significant effects. SCFAs have a significant role in the differentiation and activation of anti-inflammatory Treg cells or pro-inflammatory TH1 and TH17 cells. These cells have crucial roles in the immune responses and SCFAs regulate the balance, thus affecting the overall state of the immune system. For example, SCFAs may reduce inflammatory reactions by inhibiting the activity of pro-inflammatory cells or enhance immune tolerance by promoting the generation of anti-inflammatory cells. Additionally, SCFAs regulate the differentiation of plasma cells and the secretion of IgA. By modulating the differentiation of plasma cells and IgA secretion, SCFAs enhance the function of the intestinal mucosal barrier, thereby protecting the host from infection. Finally, SCFAs also affects the polarization of pro-inflammatory M1 and anti-inflammatory M2 macrophages. Macrophages are an important class of immune cells that have a vital role in inflammatory responses and immune regulation. By regulating macrophage polarization, SCFAs impact the magnitude and duration of inflammatory reactions, thus protecting the host from the damage caused by excessive inflammatory responses5,26,89,91,92. A recent study focusing on patients with PD-1-responsive and non-responsive solid tumors revealed that patients with a responsive status had higher levels of SCFAs, including butyrate93. It is well-known that fungi produce SCFAs, hence the impact of fungi on immunotherapy needs to be studied further.
Strategies targeting fungi for cancer
Bacteria and bacteria secretions have an indirect anti-tumor role by regulating intestinal fungi
SCFAs produced by symbiotic bacteria can act on neighboring fungi, affecting replication and morphogenesis. Acetate, butyrate, and propionate have all been shown to inhibit the growth dynamics and mycelium formation of C. albicans and Pichia pastoris in vitro, thereby impacting stable colonization of the intestine. In addition, some strains of E. coli release soluble factors that directly kill C. albicans, adding to the well-known role of this bacterium in inhibiting C. albicans replication, biofilm formation, and intestinal colonization47. Enterococcus faecalis is a common resident of the intestine and an important opportunistic pathogen. E. faecalis secretes the peptide, EntV, which can effectively inhibit the formation of mycelia19. Pichigia inhibits the growth of Candida, Aspergillus, Fusarium, and Cryptococcus by regulating virulence factors, such as nutrient restriction, biofilm formation, development, and adhesion. This inhibitory activity is related to secreted protein(s)58,94,95. In the presence of Lactobacillus rhamnosus GG, cytokines prevent intestinal epithelial cell-induced apoptosis, activate the anti-apoptotic Akt/protein kinase B pathway, and inhibit pro-apoptotic P38/mitogen-activated protein kinase activation via TNF-α, IFN-γ, or IL-196. L. rhamnosus limits the processes required for C. albicans invasions by secreting metabolites and inhibits C. albicans by starving C. albicans of nutrients that sustain the invasion phenotype19. Therefore, studying these antibacterial substances may provide new ideas for the development of novel antifungal drugs.
Probiotics prevent cancer by inhibiting carcinogenic production, enhancing the immune system, regulating the balance of intestinal flora, and acting directly against cancer
Probiotics are defined as a class of living organisms that result in numerous health benefits to the host when consumed in sufficient quantities92,97,98. In particular, the role of probiotics in promoting gut health and preventing gastrointestinal diseases, including cancer, has been studied and reported extensively30. Probiotics not only help maintain the balance of gut microbiota but also potentially reduce the risk of CRC. Furthermore, probiotics can be potentially used in cancer treatment to manage the side effects of traditional cancer therapies, such as surgery, radiotherapy, and chemotherapy, thereby improving the safety of these treatments98–100. Probiotics include bacterial species, such as Lactobacillus, Bifidobacterium, and a strain of yeast (Saccharomyces boulardii)100,101. Currently, most probiotic products contain lactic acid bacteria belonging to the genera, Lactobacillus and Bifidobacterium99. The genus, Lactobacillus, is generally considered safe and is a source of biologically active secondary metabolites. Extracellular polysaccharides from the genus, Lactobacillus, act as antioxidants, anti-biofilms, immunomodulators, antibacterial agents, and anticancer properties8,98,102. Most of the probiotics in the human microbiota belong to the genus, Lactobacillus, but there are also some yeast strains present in dairy and fermented products that are classified as probiotics103.
Intestinal ecologic disorders may cause changes in mucosal barriers and immune functions, resulting in increased intestinal permeability and bacterial translocation. Therefore, bacterial products originating from the intestine, such as endotoxins, enter the bloodstream through the portal vein system and general circulation104. S. boulardii significantly reduced the levels of costimulatory molecules (CD40 and CD80), as well as the lipopolysaccharide (LPS)-induced DC mobilization marker CC-chemokine receptor CCR7 (also known as CD197). This finding may contribute to the regulation of immune and inflammatory responses, thus protecting the host from potential tissue damage and inflammation. Protein(s) secreted by S. boulardii have demonstrated the anti-inflammatory potential through several mechanisms. Protein(s) secreted by S. boulardii inhibit the production of pro-inflammatory cytokines mediated by the inflammatory NF-κB. These microorganisms also simultaneously regulate the activity of ERK1/2 and p38 from the MAPK family, which further affects inflammatory response. Specifically, the microorganisms stimulate the production of IL-8, an important chemokine involved in leukocyte migration in inflammatory responses, but also lead to cell necrosis97,105. In addition, S. boulardii inhibits the proliferation of naive T cells, which helps to control overactivation of the immune response97,105,106. More importantly, these microorganisms activate the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ). PPAR-γ is a nuclear receptor that participates in the regulation of various biological processes, including inflammation, metabolism, and cell proliferation105.
Antigen penetration caused by intestinal mucosal destruction is one of the main factors contributing to the pathogenesis of many inflammatory diseases. Treatment with S. boulardii reduced the intestinal permeability and plasma endotoxin in CTC-treated rats104. Probiotics enhance intestinal barrier function by regulating the cytoskeleton and phosphorylation of tight junction proteins, such as ZO-196,105. S. boulardii-induced intestinal nutritional effects include enhanced brush boundary enzyme activity, increased sIgA secretion in intestinal fluids, and enhanced production of polymeric Ig receptor in villi and crypt cells101,106,107. In addition, the release of spermidine, which is necessary for cell division and differentiation, also has specific effects on the regeneration of mucosal epithelium107,108. Many digestive enzymes and nutrient transporters may be induced by polyamines secreted by S. boulardii, which promote RNA binding and stabilization, thereby promoting growth and protein synthesis92. S. boulardii regulates the gut microbiota, stimulates the production of SCFAs, inhibits the release of and various enzymes, such as lactase, alkaline phosphatase, glucose amylase, and A-glucosidase maltase, from the brush boundary and also induces the maturation of IECs60,92,100. SCFAs, especially butyrate, inhibit histone deacetylation. This inhibitory causes the group egg white to be in a more acetylated state, thus affecting chromatin organization and activating genes involved in cell differentiation, apoptosis, and cell cycle arrest in malignant cells and downregulating the expression of inflammatory factors in intestinal mucosa cells109. S. boulardii helps to increase the levels of the anti-inflammatory cytokine, IL-10, and decreases the levels of the proinflammatory cytokines, such as IL-6 and TNF-α92,104,106,110. Apparently, beneficial fungi are capable of creating a favorable environment for the antitumor effect.
At present, three clinical trials of S. boulardii have been completed, and the project numbers are NCT01895530, NCT01609660, and NCT03358511. The first two studies determined the impact of probiotics on the intestinal microbiota and the association with postoperative outcome after colorectal surgery. The third study looked at modifying the gut microbiome to target breast cancer.
Conclusions
Awareness regarding the important role of fungi in health and disease has grown in recent years. Fungi, such as C. albicans and Saccharomyces, are known to colonize healthy individuals. When the intestinal epithelial barrier is breached or the immune response is insufficient, fungi can induce cancer by influencing the immune system or releasing specific substances. Fungi can also interact with bacteria and viruses to cause diseases in the host. With in-depth research on the relationship between fungi and tumors, detecting the types and quantities of fungi in tumor tissues may help assess the malignancy and prognosis of tumors. Furthermore, utilizing antitumor components from medicinal fungi may lead to the development of new antitumor drugs or adjuvant therapeutic methods. With the continuous development of high-throughput sequencing technology and bioinformatics, more types of intestinal fungi and tumor-specific fungi will be discovered and identified in the future. The presence and distribution patterns of these fungi in the tumor microenvironment will be more fully revealed. The interaction between intestinal fungi and tumor fungi in the TME involves knowledge and technology from multiple disciplinary fields. Therefore, we need to strengthen the cross-fusion and collaborative innovation between microbiology, oncology, immunology, and other multidisciplinary disciplines to jointly provide new ideas and methods for tumor prevention and treatment. Although studying the interaction between intestinal fungi and tumor fungi in the TME has broad prospects, many challenges still exist. For example, the specific mechanism of fungal action in the TME is not fully understood. It is still difficult to accurately detect and quantitatively evaluate the variation of species and quantity of fungi. How to effectively transform the basic research results into clinical applications is essential.
Conflict of interest statement
No potential conflicts of interest are disclosed.
Author contributions
Conceived and designed the analysis: Yue Wang, Yiwen Wang, Tao Sun, Junnan Xu.
Collected the data: Yue Wang, Yuhang Zhou.
Contributed data or analysis tools: Yiwen Wang, Yun Feng, Tao Sun.
Performed the analysis: Yue Wang, Yiwen Wang, Yuhang Zhou, Junnan Xu.
Wrote the paper: Yue Wang, Yiwen Wang.
- Received June 28, 2024.
- Accepted October 25, 2024.
- Copyright: © 2024, The Authors
This work is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License.
References
- 1.↵
- 2.↵
- 3.↵
- 4.↵
- 5.↵
- 6.↵
- 7.↵
- 8.↵
- 9.↵
- 10.↵
- 11.↵
- 12.↵
- 13.↵
- 14.↵
- 15.↵
- 16.↵
- 17.↵
- 18.↵
- 19.↵
- 20.↵
- 21.↵
- 22.↵
- 23.↵
- 24.↵
- 25.↵
- 26.↵
- 27.↵
- 28.↵
- 29.↵
- 30.↵
- 31.↵
- 32.↵
- 33.
- 34.↵
- 35.↵
- 36.↵
- 37.↵
- 38.↵
- 39.↵
- 40.↵
- 41.↵
- 42.↵
- 43.
- 44.
- 45.↵
- 46.↵
- 47.↵
- 48.↵
- 49.
- 50.↵
- 51.↵
- 52.↵
- 53.↵
- 54.↵
- 55.↵
- 56.↵
- 57.↵
- 58.↵
- 59.↵
- 60.↵
- 61.↵
- 62.↵
- 63.↵
- 64.↵
- 65.↵
- 66.↵
- 67.↵
- 68.↵
- 69.↵
- 70.↵
- 71.↵
- 72.↵
- 73.↵
- 74.↵
- 75.↵
- 76.↵
- 77.↵
- 78.↵
- 79.↵
- 80.↵
- 81.↵
- 82.↵
- 83.↵
- 84.↵
- 85.↵
- 86.↵
- 87.↵
- 88.↵
- 89.↵
- 90.↵
- 91.↵
- 92.↵
- 93.↵
- 94.↵
- 95.↵
- 96.↵
- 97.↵
- 98.↵
- 99.↵
- 100.↵
- 101.↵
- 102.↵
- 103.↵
- 104.↵
- 105.↵
- 106.↵
- 107.↵
- 108.↵
- 109.↵
- 110.↵