Skip to main content
Brazilian Journal of Microbiology logoLink to Brazilian Journal of Microbiology
. 2023 Feb 25;54(2):907–919. doi: 10.1007/s42770-023-00936-0

Non-self glycan structures as possible modulators of cancer progression: would polysaccharides from Cryptococcus spp. impact this phenomenon?

Israel Diniz-Lima 1, Leonardo Marques da Fonseca 1, Jhenifer Santos Dos Reis 1, Debora Decote-Ricardo 2, Alexandre Morrot 3,4, Jose Osvaldo Previato 1, Lucia Mendonça Previato 1, Celio Geraldo Freire-de-Lima 1, Leonardo Freire-de-Lima 1,
PMCID: PMC10235250  PMID: 36840821

Abstract

Invasive fungal infections (IFI) are responsible for a large number of annual deaths. Most cases are closely related to patients in a state of immunosuppression, as is the case of patients undergoing chemotherapy. Cancer patients are severely affected by the worrisome proportions that an IFI can take during cancer progression, especially in an already immunologically and metabolically impaired patient. There is scarce knowledge about strategies to mitigate cancer progression in these cases, beyond conventional treatment with antifungal drugs with a narrow therapeutic range. However, in recent years, ample evidence has surfaced describing the possible interferences that IFI may have both on the progression of pre-existing cancers and in the induction of newly transformed cells. The leading gambit for modulation of tumor progression comes from the ability of fungal virulence factors to modulate the host’s immune system, since they are found in considerable concentrations in the tumor microenvironment during infection. In this context, cryptococcosis is of particular concern, since the main virulence factor of the pathogenic yeast is its polysaccharide capsule, which carries constituents with high immunomodulatory properties and cytotoxic potential. Therefore, we open a discussion on what has already been described regarding the progression of cryptococcosis in the context of cancer progression, and the possible implications that fungal glycan structures may take in both cancer development and progression.

Keywords: Invasive fungal infection, Cryptococcosis, Glycoconjugates, Immunomodulation, Cancer, Epithelial-mesenchymal transition process, Glycobiology, Metastasis

Introduction

Humans, as any other animal species, are historically afflicted by pathogenic fungal-related infections [1]. The main reason involved in the urge to study these infections is the immune evasion capacity that this pathogen displays during its life cycle [2]. Fungi environmental diversity implies an array of intrinsic resistance mechanisms that contributes to fungal resistance to competitors, like free-living organisms, and to the diversification of structural patterns associated with fungal species [3, 4]. Therefore, many environmental fungal species can become pathogenic and are capable of causing opportunistic infections. Consequently, they acquire a higher proliferation rate in an immunocompromised host, subverting the immunological response, through the action of the fungal virulence factors, at a cellular level [5]. The most common examples of fungal infections causing severe public health issues are the infections caused by Candida albicans species, comprising the higher part of global mortality by fungal diseases [6]. Interestingly, C. albicans is a microorganism from the commensal human microbiota that colonizes the skin and mucous membranes. In immunocompromised hosts, this yeast can grow considerably, promoting a superficial infection [7]. Due to this opportunistic behavior, C. albicans yeasts can develop the late stages of its life cycle, causing the formation of invasive hyphae in the host’s tissues, as is the case of common infections in post-transplant patients, as well as in chemotherapy-treated cancer patients [8]. These susceptible populations are not only subjugated to opportunistic infections by commensal microorganisms as C. albicans, but by other environmental fungi as well, causing severe cases of invasive fungal infections (IFI). Major examples are Aspergillus fumigatus and the pathogenic species of Cryptococcus, such as Cryptococcus neoformans and Cryptococcus gattii [9, 10].

Invasive fungal infections and cancer

Much is still debated about the association between IFI incidence in individuals with either immune deficiencies or chronic diseases [11]. Therefore, studies started to associate socioeconomic features with chronic diseases and cancer progression, where urbanized countries possessed a higher incidence compared to others, a phenomenon explained by the hygiene hypothesis [12, 13]. During the early stages of immune development in a prevalent hygiene status, more self-reactive immune cell populations are accumulated instead of activated antigen-specific cell clones. Then, the immune system is conditioned to self-reactivity and immune regulation; therefore, the tumor progression ends up being facilitated when the urge of autoimmunity is privileged [14]. However, the oversight by public and private health systems in underdeveloped countries, with a higher incidence of neglected tropical diseases, gives rise to new questions about cancer susceptibility [15]. The hygiene theory is challenged in this increasing scenario of microbe-related pathologies modulating the immune response against or in favor of tumor progression [13].

Naturally, some viral infections interfere with the cell cycle and proliferation and may favor the development of cancer, just as some bacterial infections, by promoting inflammatory processes, contribute to the necrosis of metastatic cells by a different mechanism. Naturally, some viral infections interfere with the cell cycle and proliferation and may favor cancer development [16]. Bacterial diseases, on the other hand, whether by inducing inflammatory mediators or directly by endotoxin production, may contribute to cancer cell necrosis [17, 18]. Furthermore, Candida albicans can also induce oral tumor progression by endogenous production of nitrosamines and release of its virulence factor, candidalysin, during infection [19, 20]. Therefore, microbial virulence factors such as endotoxins and other soluble molecules can modulate tumor progression. Furthermore, the presence of chronic infections implies epigenetic modifications that cause activation of oncogenes and inactivation of tumor suppressor genes [21]. Gastric cancer may be induced by Helicobacter pylori infection due to changes in DNA methylation, for instance [22]. Indeed, recent data suggests specific commensal fungal microbiome may be related to different types of cancer, such as pancreatic, breast, and ovarian cancers [23, 24]. Nonetheless, little is clear about the influence of IFI during oncogenesis.

Cancer patients are especially vulnerable to fungal infections for many reasons. The use of corticosteroid promotes a state of immunosuppression during their treatment [25], leading to a reduction of the host defense against infections, even promoting microbiota dysregulation [26]. The state of dysbiosis within membranes can lead to mucositis, facilitating the settling of pathogenic microorganisms [27]. Besides that, cancer chemotherapy generates a great metabolic stress and a severe immunity impairment. Therefore, cases of myelosuppression and mucous membrane ulcerations are frequently responsible for the entry of opportunistic fungi [28]. When it comes to IFI in cancer patients, a critical factor resulting in greater disease severity is the lack of control in the early stages of infection that quickly advances to graver forms, as the case of fungemia caused by C. albicans and meningoencephalitis caused by pathogenic Cryptococcus spp. [29]. IFI worsen rapidly in the more advanced stages of the infection. In addition, they tend to require prolonged use of antifungal treatment. Therefore, a late diagnosis makes it difficult to resolve the infection [30]. Furthermore, antifungal therapy may result in acute hepatotoxicity and nephrotoxicity during prolonged treatment [3133]. The combination of different types of medication (antifungals and chemotherapeutic agents) can potentiate toxic effects, because they share the same metabolization pathways or because individuals are already metabolically debilitated by cancer [34, 35].

Although not a new idea [36], more significant associations between IFI and cancer have been made recently, concerning the synergic and deleterious potential that both diseases can bring about [37]. Previous works associate the increase of metastatic potential of some types of cancer caused by opportunistic infections by C. albicans [38, 39], such as hepatic carcinoma [40, 41], and oral squamous cell carcinoma (OSCC) [42, 43]. The argument that supports this mechanism is still not fully understood, and it may be related, in the case of C. albicans, with the invasive potential that the Candida hyphae possess, leading to cell–matrix destabilization and upregulation of the epithelial-mesenchymal transition (EMT) markers [42]. This may be related to the fact that C. albicans is potentially capable of nitrosamine production, a carcinogen associated with the development of oral squamous cell carcinoma (OSCC) and other types of cancers [19]. Inquiringly, some rare associations of patients developing OSCC after C. albicans infection have been observed [43]. It is worth mentioning that much is still discussed about the real reasons why potentially pathogenic fungi can enhance or trigger metastatic processes in humans. It seems to be due to a combination of the inflammatory status and mechanisms initiated by virulence factors intrinsic to pathogenic fungi, such as acetaldehyde [44], candidalysin [45], or acid aspartyl proteinases [20].

Cryptococcosis during cancer progression

Cryptococcosis is an invasive opportunistic infectious disease, caused by the pathogenic species of the Cryptococcus genre, and it is frequently associated with severe cases of pulmonary infection in cancer patients [46]. Despite the suggested evidence that cryptococcosis has the potential to contribute to the worsening of the host’s health status during cancer progression [47], the mechanisms involved in the relation between cryptococcosis and tumoral progression are still poorly understood. The majority of the cases reported are associated with the Cryptococcus neoformans species, known to infect immunocompromised hosts [1], while for other species such as Cryptococcus gattii, the association is still scarce [48]. Interestingly, cryptococcosis more commonly affects patients suffering from hematological malignancies than those bearing solid tumors [49]. The immunomodulatory potential of the Cryptococcus spp. during the infection is vastly explored and well described [5054]. Most effects are associated to its main virulence factor, the foremost capsular polysaccharidic components, namely glucuronoxylomannan (GXM) and glucuronoxylomannogalactan (GXMGal). Both polysaccharides are capable of modulating the cell activation process and inflammatory response, with GXM being the most prevalent in the total capsule composition [55]. Interestingly, during cryptococcosis, these components with elevated immunomodulatory potential are constantly leaked by the fungus, permeating the host’s tissues. Therefore they have the capability to modulate the immune response in sites non-adjacent to the focal infection, acting systemically [56]. Although C. albicans infects primary sites of tumor progression, as occurs during OSCC, an IFI does not necessarily install itself at the same site as the neoplastic formation. As in the case of infection by Cryptococcus spp., which most often starts as a pulmonary infection, however, the vast bioavailability of its capsular polysaccharides with a high immunomodulatory effect has the potential to modulate other sites systemically and may not be limited only to the lung site [57, 58]. Some cases of coexisting Cryptococcus spp. infection with cancers, for instance, in the development of cryptococcoma together with pulmonary cancer [59], or even the treatment of breast cancer with anastrozole favoring the progression of cryptococcosis [60], point to some connection between tumoral treatment and infection progression. The relationship between Cryptococcus spp. and cancer goes both ways and breast or prostate cancer even in initial stages can facilitate cases of opportunistic infections in- and off-site [6163]. The cryptococcal infection is very frequent in patients with acquired immunodeficiency syndrome (AIDS), with focal infections colocalized with the lesions caused by the Kaposi sarcoma [64]. Lymphoproliferative neoplasms are associated with Cryptococcus spp. infections as well [65, 66]. Furthermore, because of the incapacitated status of cancer patients, cases of Cryptococcus laurentii infection has been observed. This can lead to very worrisome implications, as C. laurentii is commonly seen as an environmental fungi species not associated with infections. Therefore, cancer patients may be affected by infections caused by previously non-pathogenic fungi [67, 68]. It is possible to speculate that fungi with the ability to evade the environment can be categorized as opportunistic fungi.

Anticryptococcal treatment and cancer chemotherapy

Chemotherapy in cancer patients tends to immunologically weaken the individual, which makes them susceptible to various opportunistic infections, such as pulmonary IFI [69]. There is a high mortality rate associated with IFI in cancer patients; therefore, treatment with antifungal drugs is mandatory in the early stages of diagnosis to increase survival [70]. However, antifungal prophylaxis is extremely limited in terms of diversity and availability of drugs, being limited to a few drug classes [71]. The infection occurrence is mainly due to chemotherapy, which compromises CD4+ T lymphocyte count levels along with defects in T helper lymphocyte function. Therefore, patients who develop AIDS are extremely susceptible to cryptococcosis [72]. In antifungal therapy against cryptococcosis, the therapeutic strategy consists of a combination of drug classes. Azole compounds and antimycotic polyenes are the most widely used and indicated by international guidelines and organizations, such as Food and Drug Administration (FDA) and German Society for Haematology and Medical Oncology (DGHO) [70, 73]. These ergosterol synthesis inhibitors are fungistatic agents that compromise the structural integrity of the pathogenic yeast [74]. First, therapy with flucytosine and amphotericin B is started, followed by continuous treatment with fluconazole for 10 to 12 weeks [75]. Alternatively, posaconazole, voriconazole, and itraconazole can be used. However, the most efficient in reducing the yeast’s levels in the cerebrospinal fluid is fluconazole [76, 77]. It is important to note that the use of azoles during chemotherapy should be taken with caution, since the prolonged administration of this drug class can lead to an excessive increase in the bioavailability of certain chemotherapeutics, thus leading to the potentiation of the deleterious effects of chemotherapy. The main reason for those effects is due to the fact that azole compounds are inhibitors of the hepatic enzymes of the cytochrome P450 3A4 complex (CYP3A4) and many chemotherapeutics pass part of their biotransformation route through this enzyme complex [78]. Ibrutinib, midostaurin, sorafenib, and venetoclax, for example, rely upon CYP3A4 for their metabolization and are highly impacted by the action of azoles, with their recommended dose being reduced when used in combination with these antifungals, and it is also recommended that serum levels of chemotherapeutic agents should be monitored continuously [79]. Therefore, this interaction can impact the effectiveness of chemotherapy, as well as immunological aspects, culminating in a worse prognosis for fungal infections. Due to the impact on the biotransformation of hepatic metabolites, not only are chemotherapeutic compounds affected, but the synthesis of endogenous hormones is also compromised with prolonged azole treatment [80]. Adrenal insufficiency is characteristic in this case, resulting in reduced production of endogenous steroids, since ergosterol is an essential precursor for the synthesis of many glucocorticoids, mineralocorticoids, and human gonadal hormones [81, 82]. This condition can lead to further debilitation of the individual and may even go unnoticed, since the cancer progression itself causes symptoms similar to adrenal insufficiency, such as weakness, anorexia, nausea, and vomiting [83]. Interestingly, treatment with ketoconazole and fluconazole is used in Cushing’s syndrome in order to reduce the high glucocorticoid hormone levels associated with this pathology [81]. In this context, renal failure is critical because concomitant treatment with anticancer agents in these patients becomes more toxic for individuals with lower renal clearance [84]. The effect of hormonal imbalance generated by adrenal insufficiency conditions the individual to an immunosuppressed state, mainly affecting populations of cytotoxic T lymphocytes such as the natural killer cells, which directly corroborates the impairment of antitumor immunity [85]. The strong hormonal modulation of azole compounds in the context of cancer has not yet been evaluated with care and detail; however, it is important to emphasize that hormonal disorders not only strongly modulate immune aspects, but also cancer progression and malignancy aspects [86, 87]. Also, many tumors respond directly to gonadal hormones. Breast cancer progression and malignancy can be greatly modulated by the presence of estrogen [88, 89], while prostate cancers respond to circulating testosterone levels, to a point where gonadal deprivation to reduce hormone levels to a castration level is an actual therapeutical option in some cases [90, 91]. Testosterone replacement therapy remains a controversial topic for hypogonadal men suffering from prostate cancer or even at a higher risk for it [92]. Taking this into account, any effects azole treatments for cryptococcosis can exert on hormonal modulation may lead to potentially significant repercussions on tumor progression and malignancy. Also, a higher risk for squamous cell carcinoma (SCC) has been associated to use of voriconazole [9395]. Although the specific mechanisms involved are not yet known, one possible explanation might be linked to its main metabolite, a chromophore for UV-B radiation. The increased absorption of amount of ultraviolet (UV) light plays a key role in enhancing DNA damage [93]. Voriconazole is also capable of inhibiting vitamin A (vit A) metabolization and increasing the half-life of molecules such as tretinoin, leading to increased phototoxicity [96, 97]. Higher intake of vit A itself is associated with lower levels of ovarian and breast cancer, however. So, it is not a clear-cut relationship [98, 99]. It is clear, however, that chronic use of azole compounds can further enhance cancer progression if used inappropriately and without surveillance.

Repurposing of antifungals for cancer chemotherapy

Despite the metabolic stress that azole antifungals can cause in patients with concomitant chemotherapy for cancer treatment, the possibility of repurposing antifungal drugs in order to use them as adjuvant therapy in the treatment of certain types of cancer has gained traction in recent years [100102], such as miconazole in osteosarcoma [103], terbinafine in promyelocytic leukemia [104], ketoconazole in colorectal and hepatocellular carcinoma [105], and itraconazole in cutaneous squamous cell carcinoma [106], melanoma [107], and glioblastoma [108]. Many antifungals act through compromising cell wall structure, enhancing DNA damage, or inhibiting mitosis, and many of these effects, apart from those directed at the cell wall, are similar to the action mechanisms displayed by chemotherapy drugs against malignant cells [109111]. Therefore, the use of non-cancer drugs such as antifungals not only has theoretical justification for their off-label use [112, 113], but has also shown promise in oncological clinical trials [114]. In the case of azole drugs, activation of apoptosis pathways is one of the proposed mechanisms to control tumor progression [115]. Itraconazole has even been described as an inducer of a cell death pathway by inducing autophagy of metastatic cells, increasing the survival outcome in patients with colon cancer and glioblastoma [108]. Ketoconazole also elicits similar mechanisms in the context of hepatocellular carcinoma, while sertaconazole does the same in lung cancer [116, 117]. In addition to inducing cell death, the antiangiogenic effect of itraconazole limits tumor progression, since angiogenesis is a crucial mechanism for the maintenance of newly formed tumor tissue [118]. In fact, its use has already been considered beneficial in the joint treatment of acute myeloid leukemia with chemotherapy [119]. Furthermore, the inhibition of one of the pathways for cell proliferation, the Hedgehog pathway, important for tumor progression, is suppressed by the action of itraconazole [120]. Interestingly, one of the mechanisms by which itraconazole acts is by inhibiting P-glycoprotein (P-gp), an efflux protein of the ABC superfamily associated with multidrug resistance (MDR) phenotype [121]. P-gp inhibition in tumors exhibiting MDR phenotypes can lead to resistance reduction, facilitating the action of other drugs [122]. Itraconazole is also capable of inhibiting the ABC transporters, CDR1/2 in fungal pathogens. ABC transporters, or ATP-binding cassette transporters, are primary active transporters that use energy from adenosine triphosphate (ATP) hydrolysis to fuel the transport of molecules across membranes of prokaryotic and eukaryotic cells against their electrochemical gradient [123]. Furthermore, they are essential for the uptake of nutrients for cellular metabolism and are crucial in exporting molecules present inside the cytoplasm; therefore, they compose in tumor cells an essential mechanism for drug resistance, because they behave like an efflux pump for chemotherapy drugs [124]. These proteins are associated with drug efflux, reducing the effective concentration in the cytoplasmic interior of the yeast, and are therefore linked to azole resistance [125128] (Fig. 1). Concomitantly, the use of doxorubicin, a chemotherapy drug of choice, has already been described as a potent inducer of ABC transporters in C. albicans, increasing the levels of CDR1/2 proteins [129]. Similar observations can be made for P-gp and other ABC transporters in cancer models using either doxorubicin or other anticancer drugs [130133] and it is possible to theorize that drug associations between itraconazole and anticancer drugs could represent a higher chance of eliciting ABC-mediated drug resistance. Thus, in addition to the implications that fungal infection may have on antitumor immunity, drug treatment itself can modulate the response to tumor progression.

Fig. 1.

Fig. 1

The effects of antitumor and antifungal drug therapy on immunity during metastasis and proposed mechanisms for the interaction between the pathogenic fungus Cryptococcus spp. and its capsular polysaccharides during tumor progression. MMP, matrix metalloproteinase; EMT, epithelial-mesenchymal transition; ABC transporters, ATP-binding cassette transporters. Green arrows indicate activation and induction effects; red arrows indicate regulation and inhibition effects; gray arrows indicate proposed effects. Upside and downside arrows indicate respectively upregulation and downregulation of indicated factors

Anticryptococcal immunity and cancer interplay

The facilitated of Cryptococcus spp. entry in cancer patients through the lungs can lead to greater dissemination of the fungus systemically [48, 134]. There are structural factors such as mannoproteins that facilitate adhesion to the lung epithelium and greater interaction of the fungus with host cells [135]. The activation of macrophages in the pulmonary microenvironment is decisive for cryptococcosis control [136]. Naturally, the maintenance of an inflammatory profile with the Th17 cell activation and IL-6 production is responsible for the Wnt and NF-κB pathway induction, both pivotal for tumorigenesis through apoptosis regulation, a mechanism already associated with C. albicans infection [26, 42]. In the case of Cryptococcus spp. infection, the effect may vary according to the species. In infections caused by C. neoformans occurs an increase in the Th1 and Th17 profiles responsible for activating the Wnt pathway [137]. However, in infections caused by C. gattii, there is a considerable disfavor of the activation of responses associated with the Th17 profile; furthermore, the interferences in the Wnt pathway in C. gattii are still unknown [138]. Despite this possible mechanism for oncogenesis promotion during IFI, Cryptococcus spp. infections can subvert the cytokine microenvironment promoting immune regulation. Interestingly, Cryptococcus spp. infection initially induces Th1/Th17 cell profile activation and cell recruitment by IL-18 production [139]. Invariably, the production of IL-18 is closely associated with the activation of cytotoxic cells with the production of IFN-γ that mitigate the progression of tumors and autoimmunity [140]. However, chronically, Cryptococcus spp. promotes Th2 responses, strongly associated with IL-33 cytokine production [141143]. This cytokine is not only responsible for the positive feedback for Th2 maintenance, but in association with the TGF-β production, it is linked to enhanced invasiveness of malignant cells’ chemotherapy resistance during tumor progression due to their capacity to initiate EMT programming [144146]. Furthermore, the recruitment of regulatory cells such as M2 phenotype macrophages is frequent in pulmonary cryptococcosis [147, 148]. This macrophage subpopulation contributes to a worse prognosis in the context of tumors [149, 150]. The T cell profile modulation during cryptococcosis could be activated differently depending on the fungal strain or species, as seen by the C. gattii species promoting a more Th2-oriented profile induction with compromised Th1 response in comparison with C. neoformans species [138]. The differences are significant as well in the regulation of matrix metalloproteinases (MMP), also markers of the metastatic process during tumor progression. During lung infection, C. neoformans induces upregulation of MMP-3 and MMP-12, an effect not replicated for C. gattii infection [151] (Fig. 1). This strongly suggests the immunomodulatory profile promoted by different species of Cryptococcus is divergent. Not only that, but it implies concomitant infections in cancer patients could have different impacts on disease outcomes, depending on the pathogen.

Possible role of the Cryptococcus capsular polysaccharides during cancer progression

It is well established that glycosylation changes are a hallmark of both cancer cells [152157] and infiltrating immune cells in the tumor microenvironment [158160]. Over the last 10 years, our research group has demonstrated that alterations in both N- and O-linked glycan structures are able to influence the behavior of transformed cells, modulating events related to the EMT process and multidrug resistance phenotype [132, 161166]. Although some epidemiological studies have already described a correlation between infections caused by different fungi and numerous types of cancer [167171], it is not well known how the pathogen and/or its polysaccharide constituents may participate in metastatic events or the acquisition of drug resistance phenotypes, which today are the greatest obstacles faced in clinical oncology [172176]. In addition to the infection itself, the major capsular polysaccharide of Cryptococcus spp., GXM, may have significant implications for tumor progression. The potential of GXM to recruit suppressor T cells has already been described [177], as well as the reduced leukocyte translocation between epithelial barriers [178]. These characteristics are complicating factors regarding the containment of tumors by immunity, especially when associated with reduced migration of phagocytes and subversion of the production of inflammatory mediators generated by GXM [179]. Therefore, the facilitation of transepithelial passage and modulation of cell containment in the inflammatory foci due to exposure to GXM within the course of the infection itself could modulate the tumor invasiveness response. The epithelium of the host’s blood barriers can also be modulated by the Cryptococcus spp. capsular content to enhance adhesion. Despite the mechanisms through which GXM reduces macrophage phagocytosis [180], inducing cell apoptosis [181, 182], reducing T cell activation [183, 184], and generating affinity with epithelial cells via interaction with the CD14 receptor [1], it is closely associated with facilitating the passage of yeast across the blood–brain barrier [185]. Therefore, the interaction of the yeast with the epithelium seems to be important for its effective proliferation. However, data relating the epithelium dynamics between brain or lung cancer and Cryptococcus spp. infection has not yet been evaluated. In addition, other factors such as the enhanced CD44 expression caused by the hyaluronic acid that recovers the capsule and binds to the epithelium facilitate the fungal dissemination between lung and brain areas [186, 187]. Interestingly, the CD44 is a characteristic maker of mesenchymal cells that during infection is imprinted in the epithelial cell phenotype. Nevertheless, the reduction of E-cadherin expression is also a EMT hallmark, and its expression is dampened in the lung epithelium during cryptococcosis by the capsular content to enhance transepithelial passage of pathogenic yeasts, resulting in fungemia [144] (Fig. 1). Therefore, this gives rise to the possibility of persistent EMT maker expression during chronic cryptococcosis, increasing the possibility of metastatic dissemination of already established tumors or the generation of newly formed neoplasms. The capsular polysaccharides from Cryptococcus spp. are, in general, known for their pro-apoptotic potential [53]. Much of this pro-apoptotic capacity tends to downregulate cell activity by the formation of apoptotic bodies that are signals for regulatory activity [188, 189]. In the context of infection progression, this mechanism can worsen the cryptococcal infection by promoting the killing of effector cells and reducing their function. However, when it comes to cancer progression, the pro-apoptotic capacity of the capsular polysaccharides of Cryptococcus spp. could have a double effect. The downregulation of immune cells could dampen the antitumoral immunity, enhancing cancer progression. But at the same time, capsular polysaccharides could just as well induce tumor regression or impairment by the same pro-apoptotic mechanism. Nevertheless, the induction of apoptosis by capsular exopolysaccharides of Cryptococcus sp. has already been observed in non-small lung cancer cells [190]. Despite the cytotoxic activity of these capsular contents, IL-10 production generated with GXM treatment has the potential to regulate the cytotoxic cells [191]. Therefore, it reduces the cellular immunity potential of a possible important antitumoral immune response during concomitant cryptococcosis and cancer, leading to a poor prognosis. Hence, the strong association of cryptococcosis with tumor progression locally and systemically requires further evaluation and study.

Conclusion

Overall, there is little information about the immunopathogenesis of cryptococcosis associated with the progression of cancers, either in the possibility of the appearance of transformed cells or in the modulation of metastasis of pre-existing tumor cells and this relationship is still unclear. However, due to the great immunomodulatory potential that the infection and capsular factors exert on the infected host, it is very likely that the dynamics of tumor progression and metastasis can be influenced in some way in individuals affected with cryptococcosis and cancer, since other better-studied opportunistic fungal infections have a similar relationship with cancer progression. Nevertheless, promising antifungal drugs are being studied for off-label use in cancer treatment, leading to better patient prognosis. Evidently, the deleterious effect that an antifungal treatment in joint therapy with chemotherapy can bring to poor health patients should not be ignored, but rather increasingly studied to effectively implement the best strategies and care that must be taken in the tenuous balance between infection control and cancer chemotherapy.

An effort must be made in the field of cancer research to know more about the immune implications of a concomitant IFI, in order to unravel the possible effects of infection and its soluble virulence factors on tumor progression. Many questions still need to be clarified: whether the fungus and its capsular polysaccharides are involved in the EMT mechanism; whether the fungal infection modulates the glycophenotype of cancer cells; whether the promotion of oncogenes is associated with the development of systemic cryptococcosis; whether the infection itself can lead to metastasis of cancer cells, among other more elaborate issues. Obtaining answers to such questions will strongly contribute not only to better treatment protocols for cancer patients susceptible to fungal infections, but also to shed light on IFI that are neglected globally, and are responsible for high annual mortality rates, especially in oncological patients.

Author contribution

Conceptualization, Diniz-Lima I, da Fonseca LM, Freire-de-Lima CG, Freire-de-Lima L.

Technical support: Santos dos Reis J, Decote-Ricardo D, Morrot A.

Wrote the manuscript: Diniz-Lima I, da Fonseca LM, Freire-de-Lima L.

Figure elaboration: Diniz-Lima I.

Supervision: Previato JO, Mendonça Previato L, da Fonseca LM, Freire-de-Lima CG, Freire-de-Lima L.

Project administration: Freire-de-Lima L.

Funding

This work was supported by the Brazilian National Research Council (CNPq), the Brazilian Cancer Foundation, and the Rio de Janeiro State Science Foundation (FAPERJ).

Data availability

Not applicable.

Declarations

Ethical approval and consent to participate

Not applicable.

Consent for publication

All authors have read and agreed to the published version of the manuscript.

Conflict of interest

The authors declare no competing interests.

Footnotes

Responsible Editor: Fernando R. Pavan

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Diniz-Lima I, Fonseca LMd, Silva-Junior EBd, Guimarães-de-Oliveira JC, Freire-de-Lima L, Nascimento DO, et al. Cryptococcus: history, epidemiology and immune evasion. Appl Sci. 2022;12(14):7086. doi: 10.3390/app12147086. [DOI] [Google Scholar]
  • 2.Zaragoza O. Basic principles of the virulence of Cryptococcus. Virulence. 2019;10(1):490–501. doi: 10.1080/21505594.2019.1614383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Diniz-Lima I, da Fonseca LM, Dos Reis JS, da Costa Rodrigues, Santos MA, Costa KM, do Nascimento Santos CA, et al. The sweet side of fungal infections: structural glycan diversity and its importance for pathogenic adaptation. Med. 2022;9(6):37. doi: 10.3390/medicines9060037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Steenbergen JN, Casadevall A. The origin and maintenance of virulence for the human pathogenic fungus Cryptococcus neoformans. Microbes Infect. 2003;5(7):667–675. doi: 10.1016/s1286-4579(03)00092-3. [DOI] [PubMed] [Google Scholar]
  • 5.Bongomin F, Gago S, Oladele RO, Denning DW. Global and multi-national prevalence of fungal diseases-estimate precision. J Fungi. 2017;3(4):57. doi: 10.3390/jof3040057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Strickland AB, Shi M. Mechanisms of fungal dissemination. Cell Mol Life Sci. 2021;78(7):3219–3238. doi: 10.1007/s00018-020-03736-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gow NA, Hube B. Importance of the Candida albicans cell wall during commensalism and infection. Curr Opin Microbiol. 2012;15(4):406–412. doi: 10.1016/j.mib.2012.04.005. [DOI] [PubMed] [Google Scholar]
  • 8.Pagani JJ, Libshitz HI. Opportunistic fungal pneumonias in cancer patients. AJR Am J Roentgenol. 1981;137(5):1033–1039. doi: 10.2214/ajr.137.5.1033. [DOI] [PubMed] [Google Scholar]
  • 9.Chaturvedi V, Chaturvedi S. Cryptococcus gattii: a resurgent fungal pathogen. Trends Microbiol. 2011;19(11):564–571. doi: 10.1016/j.tim.2011.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Doering TL. How sweet it is! Cell wall biogenesis and polysaccharide capsule formation in Cryptococcus neoformans. Annu Rev Microbiol. 2009;63:223–247. doi: 10.1146/annurev.micro.62.081307.162753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bach JF. The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat Rev Immunol. 2018;18(2):105–120. doi: 10.1038/nri.2017.111. [DOI] [PubMed] [Google Scholar]
  • 12.Garn H, Potaczek DP, Pfefferle PI. The hygiene hypothesis and new perspectives-current challenges meeting an old postulate. Front Immunol. 2021;12:637087. doi: 10.3389/fimmu.2021.637087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Oikonomopoulou K, Brinc D, Kyriacou K, Diamandis EP. Infection and cancer: revaluation of the hygiene hypothesis. Clin Cancer Res. 2013;19(11):2834–2841. doi: 10.1158/1078-0432.CCR-12-3661. [DOI] [PubMed] [Google Scholar]
  • 14.Christen U. Pathogen infection and autoimmune disease. Clin Exp Immunol. 2019;195(1):10–14. doi: 10.1111/cei.13239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ezzati M, Pearson-Stuttard J, Bennett JE, Mathers CD. Acting on non-communicable diseases in low- and middle-income tropical countries. Nature. 2018;559(7715):507–516. doi: 10.1038/s41586-018-0306-9. [DOI] [PubMed] [Google Scholar]
  • 16.Oh JK, Weiderpass E. Infection and cancer: global distribution and burden of diseases. Ann Glob Health. 2014;80(5):384–392. doi: 10.1016/j.aogh.2014.09.013. [DOI] [PubMed] [Google Scholar]
  • 17.Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A. 1975;72(9):3666–3670. doi: 10.1073/pnas.72.9.3666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lundin JI, Checkoway H. Endotoxin and cancer. Environ Health Perspect. 2009;117(9):1344–1350. doi: 10.1289/ehp.0800439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Krogh P, Hald B, Holmstrup P. Possible mycological etiology of oral mucosal cancer: catalytic potential of infecting Candida albicans and other yeasts in production of N-nitrosobenzylmethylamine. Carcinogenesis. 1987;8(10):1543–1548. doi: 10.1093/carcin/8.10.1543. [DOI] [PubMed] [Google Scholar]
  • 20.Di Cosola M, Cazzolla AP, Charitos IA, Ballini A, Inchingolo F, Santacroce L. Candida albicans and oral carcinogenesis A brief review. J Fungi. 2021;7(6):476. doi: 10.3390/jof7060476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Maeda M, Moro H, Ushijima T. Mechanisms for the induction of gastric cancer by Helicobacter pylori infection: aberrant DNA methylation pathway. Gastric Cancer. 2017;20(Suppl 1):8–15. doi: 10.1007/s10120-016-0650-0. [DOI] [PubMed] [Google Scholar]
  • 23.Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185(20):3789–806 e17. doi: 10.1016/j.cell.2022.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim JI, et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 2019;574(7777):264–267. doi: 10.1038/s41586-019-1608-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Skribek M, Rounis K, Afshar S, Grundberg O, Friesland S, Tsakonas G, et al. Effect of corticosteroids on the outcome of patients with advanced non-small cell lung cancer treated with immune-checkpoint inhibitors. Eur J Cancer. 2021;145:245–254. doi: 10.1016/j.ejca.2020.12.012. [DOI] [PubMed] [Google Scholar]
  • 26.Huet MAL, Lee CZ, Rahman S. A review on association of fungi with the development and progression of carcinogenesis in the human body. Curr Res Microb Sci. 2022;3:100090. doi: 10.1016/j.crmicr.2021.100090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Anderson PM, Lalla RV. Glutamine for amelioration of radiation and chemotherapy associated mucositis during cancer therapy. Nutrients. 2020;12(6):1675. doi: 10.3390/nu12061675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lyman GH, Abella E, Pettengell R. Risk factors for febrile neutropenia among patients with cancer receiving chemotherapy: a systematic review. Crit Rev Oncol Hematol. 2014;90(3):190–199. doi: 10.1016/j.critrevonc.2013.12.006. [DOI] [PubMed] [Google Scholar]
  • 29.Pagliano P, Esposito S, Ascione T, Spera AM. Burden of fungal meningitis. Future Microbiol. 2020;15:469–472. doi: 10.2217/fmb-2020-0006. [DOI] [PubMed] [Google Scholar]
  • 30.Singh GK, Capoor MR, Nair D, Bhowmik KT. Spectrum of fungal infection in head and neck cancer patients on chemoradiotherapy. J Egypt Natl Canc Inst. 2017;29(1):33–37. doi: 10.1016/j.jnci.2017.01.006. [DOI] [PubMed] [Google Scholar]
  • 31.Kyriakidis I, Tragiannidis A, Munchen S, Groll AH. Clinical hepatotoxicity associated with antifungal agents. Expert Opin Drug Saf. 2017;16(2):149–165. doi: 10.1080/14740338.2017.1270264. [DOI] [PubMed] [Google Scholar]
  • 32.Tragiannidis A, Gkampeta A, Vousvouki M, Vasileiou E, Groll AH. Antifungal agents and the kidney: pharmacokinetics, clinical nephrotoxicity, and interactions. Expert Opin Drug Saf. 2021;20(9):1061–1074. doi: 10.1080/14740338.2021.1922667. [DOI] [PubMed] [Google Scholar]
  • 33.Vincenzi B, Armento G, Spalato Ceruso M, Catania G, Leakos M, Santini D, et al. Drug-induced hepatotoxicity in cancer patients - implication for treatment. Expert Opin Drug Saf. 2016;15(9):1219–1238. doi: 10.1080/14740338.2016.1194824. [DOI] [PubMed] [Google Scholar]
  • 34.Perazella MA. Pharmacology behind common drug nephrotoxicities. Clin J Am Soc Nephrol. 2018;13(12):1897–1908. doi: 10.2215/CJN.00150118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rausch CR, Kontoyiannis DP. Prolonged voriconazole treatment in a patient with chronic lymphocytic leukemia resulting in a litany of chronic overlapping toxicities. J Oncol Pharm Pract. 2019;25(3):747–753. doi: 10.1177/1078155218762624. [DOI] [PubMed] [Google Scholar]
  • 36.Zimmerman LE, Rappaport H. Occurrence of Cryptococcosis in patients with malignant disease of reticuloendothelial system. Am J Clin Pathol. 1954;24(9):1050–1072. doi: 10.1093/ajcp/24.9.1050. [DOI] [PubMed] [Google Scholar]
  • 37.Angarone M. Fungal infections in cancer patients. Cancer Treat Res. 2014;161:129–155. doi: 10.1007/978-3-319-04220-6_4. [DOI] [PubMed] [Google Scholar]
  • 38.Ramirez-Garcia A, Rementeria A, Aguirre-Urizar JM, Moragues MD, Antoran A, Pellon A, et al. Candida albicans and cancer: can this yeast induce cancer development or progression? Crit Rev Microbiol. 2016;42(2):181–193. doi: 10.3109/1040841X.2014.913004. [DOI] [PubMed] [Google Scholar]
  • 39.Ramirez-Garcia A, Arteta B, Abad-Diaz-de-Cerio A, Pellon A, Antoran A, Marquez J, et al. Candida albicans increases tumor cell adhesion to endothelial cells in vitro: intraspecific differences and importance of the mannose receptor. PLoS One. 2013;8(1):e53584. doi: 10.1371/journal.pone.0053584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rodriguez-Cuesta J, Hernando FL, Mendoza L, Gallot N, de Cerio AA, Martinez-de-Tejada G, et al. Candida albicans enhances experimental hepatic melanoma metastasis. Clin Exp Metastasis. 2010;27(1):35–42. doi: 10.1007/s10585-009-9300-9. [DOI] [PubMed] [Google Scholar]
  • 41.Ramirez-Garcia A, Gallot N, Abad A, Mendoza L, Rementeria A, Hernando FL. Molecular fractionation and characterization of a Candida albicans fraction that increases tumor cell adhesion to hepatic endothelium. Appl Microbiol Biotechnol. 2011;92(1):133–145. doi: 10.1007/s00253-011-3540-8. [DOI] [PubMed] [Google Scholar]
  • 42.Vadovics M, Ho J, Igaz N, Alfoldi R, Rakk D, Veres E, et al (2022) Candida albicans enhances the progression of oral squamous cell carcinoma in vitro and in vivo. mBio. e0314421. 10.1128/mBio.03144-21. [DOI] [PMC free article] [PubMed]
  • 43.Kharadi U, Parkarwar P, Khairnar S, Arur P, Kulkarni T. Oral candidiasis turns to oral cancer - a rare clinical presentation. Clin Oncol. 2016;1:1126. [Google Scholar]
  • 44.Alnuaimi AD, Ramdzan AN, Wiesenfeld D, O’Brien-Simpson NM, Kolev SD, Reynolds EC, et al. Candida virulence and ethanol-derived acetaldehyde production in oral cancer and non-cancer subjects. Oral Dis. 2016;22(8):805–814. doi: 10.1111/odi.12565. [DOI] [PubMed] [Google Scholar]
  • 45.Engku NasrullahSatiman EAF, Ahmad H, Ramzi AB, Abdul Wahab R, Kaderi MA, Wan Harun WHA, et al. The role of Candida albicans candidalysin ECE1 gene in oral carcinogenesis. J Oral Pathol Med. 2020;49(9):835–41. doi: 10.1111/jop.13014. [DOI] [PubMed] [Google Scholar]
  • 46.Buchanan KL, Murphy JW. What makes Cryptococcus neoformans a pathogen? Emerg Infect Dis. 1998;4(1):71–83. doi: 10.3201/eid0401.980109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Caira M, Trecarichi EM, Tumbarello M, Leone G, Pagano L. Uncommon yeast infections in hematological patients: from diagnosis to treatment. Expert Rev Anti Infect Ther. 2011;9(11):1067–1075. doi: 10.1586/eri.11.124. [DOI] [PubMed] [Google Scholar]
  • 48.Paccoud O, Bougnoux ME, Desnos-Ollivier M, Varet B, Lortholary O, Lanternier F. Cryptococcus gattii in patients with lymphoid neoplasms: an illustration of evolutive host-fungus interactions. J Fungi. 2021;7(3):212. doi: 10.3390/jof7030212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Schmalzle SA, Buchwald UK, Gilliam BL, Riedel DJ. Cryptococcus neoformans infection in malignancy. Mycoses. 2016;59(9):542–552. doi: 10.1111/myc.12496. [DOI] [PubMed] [Google Scholar]
  • 50.Almeida F, Wolf JM, da Silva TA, DeLeon-Rodriguez CM, Rezende CP, Pessoni AM, et al. Galectin-3 impacts Cryptococcus neoformans infection through direct antifungal effects. Nat Commun. 2017;8(1):1968. doi: 10.1038/s41467-017-02126-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.LaRocque-de-Freitas IF, Rocha JDB, Nunes MP, Oliveira PAV, Nascimento DO, Freire-de-Lima L, et al. Involvement of the capsular GalXM-induced IL-17 cytokine in the control of Cryptococcus neoformans infection. Sci Rep. 2018;8(1):16378. doi: 10.1038/s41598-018-34649-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rocha JD, Nascimento MT, Decote-Ricardo D, Corte-Real S, Morrot A, Heise N, et al. Capsular polysaccharides from Cryptococcus neoformans modulate production of neutrophil extracellular traps (NETs) by human neutrophils. Sci Rep. 2015;5:8008. doi: 10.1038/srep08008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Villena SN, Pinheiro RO, Pinheiro CS, Nunes MP, Takiya CM, DosReis GA, et al. Capsular polysaccharides galactoxylomannan and glucuronoxylomannan from Cryptococcus neoformans induce macrophage apoptosis mediated by Fas ligand. Cell Microbiol. 2008;10(6):1274–1285. doi: 10.1111/j.1462-5822.2008.01125.x. [DOI] [PubMed] [Google Scholar]
  • 54.Albuquerque PC, Fonseca FL, Dutra FF, Bozza MT, Frases S, Casadevall A, et al. Cryptococcus neoformans glucuronoxylomannan fractions of different molecular masses are functionally distinct. Future Microbiol. 2014;9(2):147–161. doi: 10.2217/fmb.13.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Decote-Ricardo D, LaRocque-de-Freitas IF, Rocha JDB, Nascimento DO, Nunes MP, Morrot A, et al. Immunomodulatory role of capsular polysaccharides constituents of Cryptococcus neoformans. Front Med. 2019;6:129. doi: 10.3389/fmed.2019.00129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Goldman DL, Lee SC, Casadevall A. Tissue localization of Cryptococcus neoformans glucuronoxylomannan in the presence and absence of specific antibody. Infect Immun. 1995;63(9):3448–3453. doi: 10.1128/iai.63.9.3448-3453.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yamamoto S, Niki Y, Soejima R. Fungal infection in hepatobiliary and pancreatic diseases: clinical evaluation in autopsy cases. Kansenshogaku Zasshi. 1994;68(5):612–616. doi: 10.11150/kansenshogakuzasshi1970.68.612. [DOI] [PubMed] [Google Scholar]
  • 58.Grinsell M, Weinhold LC, Cutler JE, Han Y, Kozel TR. In vivo clearance of glucuronoxylomannan, the major capsular polysaccharide of Cryptococcus neoformans: a critical role for tissue macrophages. J Infect Dis. 2001;184(4):479–487. doi: 10.1086/322787. [DOI] [PubMed] [Google Scholar]
  • 59.Yao K, Qiu X, Hu H, Han Y, Zhang W, Xia R, et al. Pulmonary cryptococcosis coexisting with central type lung cancer in an immuocompetent patient: a case report and literature review. BMC Pulm Med. 2020;20(1):161. doi: 10.1186/s12890-020-01200-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wei M, Xu YR, Liu K, Wen P. Anastrozole-induced pulmonary cryptococcosis in a patient with early breast cancer: a case report. Medicine. 2020;99(2):e18688. doi: 10.1097/MD.0000000000018688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ou KW, Hsu KF, Cheng YL, Hsu GC, Hsu HM, Yu JC. Asymptomatic pulmonary nodules in a patient with early-stage breast cancer: Cryptococcus infection. Int J Infect Dis. 2010;14(1):e77–80. doi: 10.1016/j.ijid.2009.03.007. [DOI] [PubMed] [Google Scholar]
  • 62.Shah SI, Bui H, Velasco N, Rungta S. Incidental finding of Cryptococcus on prostate biopsy for prostate adenocarcinoma following cardiac transplant: case report and review of the literature. Am J Case Rep. 2017;18:1171–1180. doi: 10.12659/ajcr.905528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wang X, Zhou Z, Turner D, Lilly M, Ou T, Jiang W. Differential circulating fungal microbiome in prostate cancer patients compared to healthy control individuals. J Immunol Res. 2022;2022:2574964. doi: 10.1155/2022/2574964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Williams SC, Sweeney J, Parameswaran L. Diagnostic and management considerations in the modern patient with AIDS: a case of concurrent disseminated Kaposi sarcoma and colesional Cryptococcus neoformans. BMJ Case Rep. 2020;13(4):e233860. doi: 10.1136/bcr-2019-233860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Silva LM, Ferreira WA, Filho R, Lacerda MVG, Ferreira GMA, Saunier MN, et al. New ST623 of Cryptococcus neoformans isolated from a patient with non-Hodgkin’s lymphoma in the Brazilian Amazon. Ann Clin Microbiol Antimicrob. 2020;19(1):20. doi: 10.1186/s12941-020-00361-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dioverti MV, Parikh SA, Osmon DR, Habermann TM, Tande AJ. Cryptococcus neoformans infections in patients with lymphoproliferative neoplasms. Leuk Lymphoma. 2019;60(4):920–926. doi: 10.1080/10428194.2018.1508666. [DOI] [PubMed] [Google Scholar]
  • 67.Averbuch D, Boekhoutt T, Falk R, Engelhard D, Shapiro M, Block C, et al. Fungemia in a cancer patient caused by fluconazole-resistant Cryptococcus laurentii. Med Mycol. 2002;40(5):479–484. doi: 10.1080/mmy.40.5.479.484. [DOI] [PubMed] [Google Scholar]
  • 68.Neves RP, Lima Neto RG, Leite MC, Silva VK, Santos Fde A, Macedo DP. Cryptococcus laurentii fungaemia in a cervical cancer patient. Braz J Infect Dis. 2015;19(6):660–663. doi: 10.1016/j.bjid.2015.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Teoh F, Pavelka N. How chemotherapy increases the risk of systemic candidiasis in cancer patients: current paradigm and future directions. Pathog. 2016;5(1):6. doi: 10.3390/pathogens5010006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Infectious Diseases Working Party of the German Society of H, Oncology, Bohme A, Ruhnke M, Buchheidt D, Cornely OA, et al. Treatment of invasive fungal infections in cancer patients--recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Ann Hematol. 2009;88(2):97-11010.1007/s00277-008-0622-5 [DOI] [PubMed]
  • 71.Mota Fernandes C, Dasilva D, Haranahalli K, McCarthy JB, Mallamo J, Ojima I, et al. The future of antifungal drug therapy: novel compounds and targets. Antimicrob Agents Chemother. 2021;65(2). 10.1128/AAC.01719-20. [DOI] [PMC free article] [PubMed]
  • 72.Brouwer AE, Rajanuwong A, Chierakul W, Griffin GE, Larsen RA, White NJ, et al. Combination antifungal therapies for HIV-associated cryptococcal meningitis: a randomised trial. Lancet. 2004;363(9423):1764–1767. doi: 10.1016/S0140-6736(04)16301-0. [DOI] [PubMed] [Google Scholar]
  • 73.McKeny PT, Nessel TA, Zito PM (2022) Antifungal antibiotics. StatPearls. Treasure Island (FL) [PubMed]
  • 74.Cross JT, Jr, Hickerson SL, Yamauchi T. Antifungal drugs. Pediatr Rev. 1995;16(4):123–129. doi: 10.1542/pir.16-4-123. [DOI] [PubMed] [Google Scholar]
  • 75.van der Horst CM, Saag MS, Cloud GA, Hamill RJ, Graybill JR, Sobel JD, et al. Treatment of cryptococcal meningitis associated with the acquired immunodeficiency syndrome. National Institute of Allergy and Infectious Diseases Mycoses Study Group and AIDS Clinical Trials Group. N Engl J Med. 1997;337(1):15–21. doi: 10.1056/NEJM199707033370103. [DOI] [PubMed] [Google Scholar]
  • 76.Pitisuttithum P, Negroni R, Graybill JR, Bustamante B, Pappas P, Chapman S, et al. Activity of posaconazole in the treatment of central nervous system fungal infections. J Antimicrob Chemother. 2005;56(4):745–755. doi: 10.1093/jac/dki288. [DOI] [PubMed] [Google Scholar]
  • 77.Saag MS, Graybill RJ, Larsen RA, Pappas PG, Perfect JR, Powderly WG, et al. Practice guidelines for the management of cryptococcal disease. Infectious Diseases Society of America. Clin Infect Dis. 2000;30(4):710–8. doi: 10.1086/313757. [DOI] [PubMed] [Google Scholar]
  • 78.Dvorak Z. Drug-drug interactions by azole antifungals: beyond a dogma of CYP3A4 enzyme activity inhibition. Toxicol Lett. 2011;202(2):129–132. doi: 10.1016/j.toxlet.2011.01.027. [DOI] [PubMed] [Google Scholar]
  • 79.Lindsay J, Teh BW, Micklethwaite K, Slavin M. Azole antifungals and new targeted therapies for hematological malignancy. Curr Opin Infect Dis. 2019;32(6):538–545. doi: 10.1097/QCO.0000000000000611. [DOI] [PubMed] [Google Scholar]
  • 80.Benitez LL, Carver PL. Adverse effects associated with long-term administration of azole antifungal agents. Drugs. 2019;79(8):833–853. doi: 10.1007/s40265-019-01127-8. [DOI] [PubMed] [Google Scholar]
  • 81.van der Pas R, Hofland LJ, Hofland J, Taylor AE, Arlt W, Steenbergen J, et al. Fluconazole inhibits human adrenocortical steroidogenesis in vitro. J Endocrinol. 2012;215(3):403–412. doi: 10.1530/JOE-12-0310. [DOI] [PubMed] [Google Scholar]
  • 82.Albert SG, DeLeon MJ, Silverberg AB. Possible association between high-dose fluconazole and adrenal insufficiency in critically ill patients. Crit Care Med. 2001;29(3):668–670. doi: 10.1097/00003246-200103000-00039. [DOI] [PubMed] [Google Scholar]
  • 83.Carvalho F, Louro F, Zakout R. Adrenal insufficiency in metastatic lung cancer. World J Oncol. 2015;6(3):375–377. doi: 10.14740/wjon890w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wirk B. Renal failure in multiple myeloma: a medical emergency. Bone Marrow Transplant. 2011;46(6):771–783. doi: 10.1038/bmt.2011.8. [DOI] [PubMed] [Google Scholar]
  • 85.Bancos I, Hazeldine J, Chortis V, Hampson P, Taylor AE, Lord JM, et al. Primary adrenal insufficiency is associated with impaired natural killer cell function: a potential link to increased mortality. Eur J Endocrinol. 2017;176(4):471–480. doi: 10.1530/EJE-16-0969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Belvisi MG. Regulation of inflammatory cell function by corticosteroids. Proc Am Thorac Soc. 2004;1(3):207–214. doi: 10.1513/pats.200402-002MS. [DOI] [PubMed] [Google Scholar]
  • 87.Obradovic MMS, Hamelin B, Manevski N, Couto JP, Sethi A, Coissieux MM, et al. Glucocorticoids promote breast cancer metastasis. Nature. 2019;567(7749):540–544. doi: 10.1038/s41586-019-1019-4. [DOI] [PubMed] [Google Scholar]
  • 88.Gompel A. Hormone and breast cancer. Presse Med. 2019;48(10):1085–1091. doi: 10.1016/j.lpm.2019.09.021. [DOI] [PubMed] [Google Scholar]
  • 89.Starek-Swiechowicz B, Budziszewska B, Starek A. Endogenous estrogens-breast cancer and chemoprevention. Pharmacol Rep. 2021;73(6):1497–1512. doi: 10.1007/s43440-021-00317-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Desai K, McManus JM, Sharifi N. Hormonal therapy for prostate cancer. Endocr Rev. 2021;42(3):354–373. doi: 10.1210/endrev/bnab002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yassin A, AlRumaihi K, Alzubaidi R, Alkadhi S, Al AA. Testosterone, testosterone therapy and prostate cancer. Aging Male. 2019;22(4):219–227. doi: 10.1080/13685538.2018.1524456. [DOI] [PubMed] [Google Scholar]
  • 92.Lenfant L, Leon P, Cancel-Tassin G, Audouin M, Staerman F, Roupret M, et al. Testosterone replacement therapy (TRT) and prostate cancer: an updated systematic review with a focus on previous or active localized prostate cancer. Urol Oncol. 2020;38(8):661–670. doi: 10.1016/j.urolonc.2020.04.008. [DOI] [PubMed] [Google Scholar]
  • 93.Williams K, Arron ST. Association of CYP2C19 *17/*17 genotype with the risk of voriconazole-associated squamous cell carcinoma. JAMA Dermatol. 2016;152(6):719–720. doi: 10.1001/jamadermatol.2016.0351. [DOI] [PubMed] [Google Scholar]
  • 94.Mansh M, Binstock M, Williams K, Hafeez F, Kim J, Glidden D, et al. Voriconazole exposure and risk of cutaneous squamous cell carcinoma, Aspergillus colonization, invasive aspergillosis and death in lung transplant recipients. Am J Transplant. 2016;16(1):262–270. doi: 10.1111/ajt.13431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Patel VA, Parikh SA, Nayyar PM, Ratner D. Voriconazole-induced multiple squamous cell carcinomas in a patient with chronic lymphocytic leukemia. Dermatol Surg. 2015;41(6):747–749. doi: 10.1097/DSS.0000000000000349. [DOI] [PubMed] [Google Scholar]
  • 96.Epaulard O, Leccia MT, Blanche S, Chosidow O, Mamzer-Bruneel MF, Ravaud P, et al. Phototoxicity and photocarcinogenesis associated with voriconazole. Med Mal Infect. 2011;41(12):639–645. doi: 10.1016/j.medmal.2011.09.016. [DOI] [PubMed] [Google Scholar]
  • 97.Cheng MP, Paquette K, Lands LC, Ovetchkine P, Theoret Y, Quach C. Voriconazole inhibition of vitamin A metabolism: are adverse events increased in cystic fibrosis patients? Pediatr Pulmonol. 2010;45(7):661–666. doi: 10.1002/ppul.21234. [DOI] [PubMed] [Google Scholar]
  • 98.Wang Q, He C (2020) Dietary vitamin A intake and the risk of ovarian cancer: a meta-analysis. Biosci Rep 40(4). 10.1042/BSR20193979 [DOI] [PMC free article] [PubMed]
  • 99.Bitsie KR, Cheng TD, McCann SE, Zirpoli G, Yao S, Bandera EV, et al. Dietary vitamin A and breast cancer risk in Black women: the African American Breast Cancer Epidemiology and Risk (AMBER) consortium. J Nutr. 2021;151(12):3725–3737. doi: 10.1093/jn/nxab278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zhang Z, Zhou L, Xie N, Nice EC, Zhang T, Cui Y, et al. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct Target Ther. 2020;5(1):113. doi: 10.1038/s41392-020-00213-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Khachigian LM. Repurposing drugs for skin cancer. Curr Med Chem. 2020;27(42):7214–7221. doi: 10.2174/0929867327666191220103901. [DOI] [PubMed] [Google Scholar]
  • 102.Weng N, Zhang Z, Tan Y, Zhang X, Wei X, Zhu Q. Repurposing antifungal drugs for cancer therapy. J Adv Res. 2022 doi: 10.1016/j.jare.2022.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Chang HT, Chen WC, Chen JS, Lu YC, Hsu SS, Wang JL, et al. Effect of miconazole on intracellular Ca2+ levels and proliferation in human osteosarcoma cells. Life Sci. 2005;76(18):2091–2101. doi: 10.1016/j.lfs.2004.09.033. [DOI] [PubMed] [Google Scholar]
  • 104.Yang KC, Wu CC, Wu CH, Chen JH, Chu CH, Chen CH, et al. Involvement of proapoptotic Bcl-2 family members in terbinafine-induced mitochondrial dysfunction and apoptosis in HL60 cells. Food Chem Toxicol. 2006;44(2):214–226. doi: 10.1016/j.fct.2005.07.008. [DOI] [PubMed] [Google Scholar]
  • 105.Chen RJ, Lee WS, Liang YC, Lin JK, Wang YJ, Lin CH, et al. Ketoconazole induces G0/G1 arrest in human colorectal and hepatocellular carcinoma cell lines. Toxicol Appl Pharmacol. 2000;169(2):132–141. doi: 10.1006/taap.2000.9062. [DOI] [PubMed] [Google Scholar]
  • 106.Xu C, Zhuo Y, Liu Y, Chen H. Itraconazole inhibits the growth of cutaneous squamous cell carcinoma by targeting HMGCS1/ACSL4 axis. Front Pharmacol. 2022;13:828983. doi: 10.3389/fphar.2022.828983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Liang G, Liu M, Wang Q, Shen Y, Mei H, Li D, et al. Itraconazole exerts its anti-melanoma effect by suppressing Hedgehog, Wnt, and PI3K/mTOR signaling pathways. Oncotarget. 2017;8(17):28510–28525. doi: 10.18632/oncotarget.15324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Liu R, Li J, Zhang T, Zou L, Chen Y, Wang K, et al. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: involvement of abnormal cholesterol trafficking. Autophagy. 2014;10(7):1241–1255. doi: 10.4161/auto.28912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Young DH, Wang NX, Meyer ST, Avila-Adame C. Characterization of the mechanism of action of the fungicide fenpicoxamid and its metabolite UK-2A. Pest Manag Sci. 2018;74(2):489–498. doi: 10.1002/ps.4743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zhou Y, Xu J, Zhu Y, Duan Y, Zhou M. Mechanism of action of the benzimidazole fungicide on Fusarium graminearum: interfering with polymerization of monomeric tubulin but not polymerized microtubule. Phytopathol. 2016;106(8):807–813. doi: 10.1094/PHYTO-08-15-0186-R. [DOI] [PubMed] [Google Scholar]
  • 111.Bae SH, Park JH, Choi HG, Kim H, Kim SH. Imidazole antifungal drugs inhibit the cell proliferation and invasion of human breast cancer cells. Biomol Ther. 2018;26(5):494–502. doi: 10.4062/biomolther.2018.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Sarkar S, Singh PC. Mechanistic aspects of fungicide-induced DNA damage: spectroscopic and molecular dynamics simulation studies. J Phys Chem B. 2019;123(41):8653–8661. doi: 10.1021/acs.jpcb.9b06009. [DOI] [PubMed] [Google Scholar]
  • 113.Li R, Liu B, Xu W, Yu L, Zhang C, Cheng J, et al. DNA damage and cell apoptosis induced by fungicide difenoconazole in mouse mononuclear macrophage RAW264.7. Environ Toxicol. 2022;37(3):650–9. doi: 10.1002/tox.23432. [DOI] [PubMed] [Google Scholar]
  • 114.Kim DJ, Kim J, Spaunhurst K, Montoya J, Khodosh R, Chandra K, et al. Open-label, exploratory phase II trial of oral itraconazole for the treatment of basal cell carcinoma. J Clin Oncol. 2014;32(8):745–751. doi: 10.1200/JCO.2013.49.9525. [DOI] [PubMed] [Google Scholar]
  • 115.Shen PW, Chou YM, Li CL, Liao EC, Huang HS, Yin CH, et al. Itraconazole improves survival outcomes in patients with colon cancer by inducing autophagic cell death and inhibiting transketolase expression. Oncol Lett. 2021;22(5):768. doi: 10.3892/ol.2021.13029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Zhang W, Zhou L, Qin S, Jiang J, Huang Z, Zhang Z, et al. Sertaconazole provokes proapoptotic autophagy via stabilizing TRADD in nonsmall cell lung cancer cells. MedComm. 2021;2(4):821–37. doi: 10.1002/mco2.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Chen Y, Chen HN, Wang K, Zhang L, Huang Z, Liu J, et al. Ketoconazole exacerbates mitophagy to induce apoptosis by downregulating cyclooxygenase-2 in hepatocellular carcinoma. J Hepatol. 2019;70(1):66–77. doi: 10.1016/j.jhep.2018.09.022. [DOI] [PubMed] [Google Scholar]
  • 118.Harris A. Antiangiogenesis for cancer therapy. Lancet. 1997;349:13–15. doi: 10.1016/s0140-6736(97)90014-3. [DOI] [PubMed] [Google Scholar]
  • 119.Copley MS, Waldron M, Athans V, Welch SC, Brizendine KD, Cober E, et al. Itraconazole vs. posaconazole for antifungal prophylaxis in patients with acute myeloid leukemia undergoing intensive chemotherapy: a retrospective study. Int J Antimicrob Agents. 2020;55(3):105886. doi: 10.1016/j.ijantimicag.2020.105886. [DOI] [PubMed] [Google Scholar]
  • 120.Xin M, Ji X, De La Cruz LK, Thareja S, Wang B. Strategies to target the Hedgehog signaling pathway for cancer therapy. Med Res Rev. 2018;38(3):870–913. doi: 10.1002/med.21482. [DOI] [PubMed] [Google Scholar]
  • 121.Tsubamoto H, Ueda T, Inoue K, Sakata K, Shibahara H, Sonoda T. Repurposing itraconazole as an anticancer agent. Oncol Lett. 2017;14(2):1240–1246. doi: 10.3892/ol.2017.6325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lee J, Kang J, Kwon NY, Sivaraman A, Naik R, Jin SY, et al. Dual inhibition of P-gp and BCRP improves oral topotecan bioavailability in rodents. Pharm. 2021;13(4):559. doi: 10.3390/pharmaceutics13040559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Locher KP. Mechanistic diversity in ATP-binding cassette (ABC) transporters. Nat Struct Mol Biol. 2016;23(6):487–493. doi: 10.1038/nsmb.3216. [DOI] [PubMed] [Google Scholar]
  • 124.Beis K. Structural basis for the mechanism of ABC transporters. Biochem Soc Trans. 2015;43(5):889–893. doi: 10.1042/BST20150047. [DOI] [PubMed] [Google Scholar]
  • 125.Prasad R, Balzi E, Banerjee A, Khandelwal NK. All about CDR transporters: past, present, and future. Yeast. 2019;36(4):223–233. doi: 10.1002/yea.3356. [DOI] [PubMed] [Google Scholar]
  • 126.Prasad R, Murthy SK, Gupta V, Prasad R. Multiple drug resistance in Candida albicans. Acta Biochim Pol. 1995;42(4):497–504. doi: 10.18388/abp.1995_4902. [DOI] [PubMed] [Google Scholar]
  • 127.Schuetzer-Muehlbauer M, Willinger B, Egner R, Ecker G, Kuchler K. Reversal of antifungal resistance mediated by ABC efflux pumps from Candida albicans functionally expressed in yeast. Int J Antimicrob Agents. 2003;22(3):291–300. doi: 10.1016/s0924-8579(03)00213-9. [DOI] [PubMed] [Google Scholar]
  • 128.White TC, Marr KA, Bowden RA. Clinical, cellular, and molecular factors that contribute to antifungal drug resistance. Clin Microbiol Rev. 1998;11(2):382–402. doi: 10.1128/CMR.11.2.382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kofla G, Turner V, Schulz B, Storch U, Froelich D, Rognon B, et al. Doxorubicin induces drug efflux pumps in Candida albicans. Med Mycol. 2011;49(2):132–142. doi: 10.3109/13693786.2010.512022. [DOI] [PubMed] [Google Scholar]
  • 130.Amawi H, Sim HM, Tiwari AK, Ambudkar SV, Shukla S. ABC transporter-mediated multidrug-resistant cancer. Adv Exp Med Biol. 2019;1141:549–580. doi: 10.1007/978-981-13-7647-4_12. [DOI] [PubMed] [Google Scholar]
  • 131.Lu X, Wang Z, Huang H, Wang H. Hedgehog signaling promotes multidrug resistance by regulation of ABC transporters in oral squamous cell carcinoma. J Oral Pathol Med. 2020;49(9):897–906. doi: 10.1111/jop.13050. [DOI] [PubMed] [Google Scholar]
  • 132.da Fonseca LM, da Silva VA, da Costa KM, Dos Reis JS, Previato JO, Previato LM, et al. Resistance to cisplatin in human lung adenocarcinoma cells: effects on the glycophenotype and epithelial to mesenchymal transition markers. Glycoconj J. 2022;39(2):247–259. doi: 10.1007/s10719-022-10042-2. [DOI] [PubMed] [Google Scholar]
  • 133.Wang T, Dong J, Yuan X, Wen H, Wu L, Liu J, et al. A new chalcone derivative C49 reverses doxorubicin resistance in MCF-7/DOX cells by inhibiting P-glycoprotein expression. Front Pharmacol. 2021;12:653306. doi: 10.3389/fphar.2021.653306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Pruitt AA. Central nervous system infections in cancer patients. Neurol Clin. 1991;9(4):867–888. doi: 10.1016/S0733-8619(18)30254-8. [DOI] [PubMed] [Google Scholar]
  • 135.Teixeira PA, Penha LL, Mendonca-Previato L, Previato JO. Mannoprotein MP84 mediates the adhesion of Cryptococcus neoformans to epithelial lung cells. Front Cell Infect Microbiol. 2014;4:106. doi: 10.3389/fcimb.2014.00106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Heung LJ, Hohl TM. Inflammatory monocytes are detrimental to the host immune response during acute infection with Cryptococcus neoformans. PLoS Pathog. 2019;15(3):e1007627. doi: 10.1371/journal.ppat.1007627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Qin HJ, Feng QM, Fang Y, Shen L. Type-I interferon secretion in the acute phase promotes Cryptococcus neoformans infection-induced Th17 cell polarization in vitro. Exp Ther Med. 2014;7(4):869–872. doi: 10.3892/etm.2014.1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Angkasekwinai P, Sringkarin N, Supasorn O, Fungkrajai M, Wang YH, Chayakulkeeree M, et al. Cryptococcus gattii infection dampens Th1 and Th17 responses by attenuating dendritic cell function and pulmonary chemokine expression in the immunocompetent hosts. Infect Immun. 2014;82(9):3880–3890. doi: 10.1128/IAI.01773-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kawakami K, Qureshi MH, Zhang T, Koguchi Y, Yara S, Takeda K, et al. Involvement of endogenously synthesized interleukin (IL)-18 in the protective effects of IL-12 against pulmonary infection with Cryptococcus neoformans in mice. FEMS Immunol Med Microbiol. 2000;27(3):191–200. doi: 10.1111/j.1574-695X.2000.tb01430.x. [DOI] [PubMed] [Google Scholar]
  • 140.Esmailbeig M, Ghaderi A. Interleukin-18: a regulator of cancer and autoimmune diseases. Eur Cytokine Netw. 2017;28(4):127–140. doi: 10.1684/ecn.2018.0401. [DOI] [PubMed] [Google Scholar]
  • 141.Flaczyk A, Duerr CU, Shourian M, Lafferty EI, Fritz JH, Qureshi ST. IL-33 signaling regulates innate and adaptive immunity to Cryptococcus neoformans. J Immunol. 2013;191(5):2503–2513. doi: 10.4049/jimmunol.1300426. [DOI] [PubMed] [Google Scholar]
  • 142.Diniz-Lima I, da Rosa PR, da Silva-Junior EB, Guimaraes-de-Oliveira JC, de Freitas EO, de Oliveira ND, et al. X-linked immunodeficient (XID) mice exhibit high susceptibility to Cryptococcus gattii infection. Sci Rep. 2021;11(1):18397. doi: 10.1038/s41598-021-97041-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Garcia-Barbazan I, Trevijano-Contador N, Rueda C, de Andres B, Perez-Tavarez R, Herrero-Fernandez I, et al. The formation of titan cells in Cryptococcus neoformans depends on the mouse strain and correlates with induction of Th2-type responses. Cell Microbiol. 2016;18(1):111–124. doi: 10.1111/cmi.12488. [DOI] [PubMed] [Google Scholar]
  • 144.Heyen L, Muller U, Siegemund S, Schulze B, Protschka M, Alber G, et al. Lung epithelium is the major source of IL-33 and is regulated by IL-33-dependent and IL-33-independent mechanisms in pulmonary cryptococcosis. Pathog Dis. 2016;74(7):ftw086. doi: 10.1093/femspd/ftw086. [DOI] [PubMed] [Google Scholar]
  • 145.Sun Z, Ji N, Ma Q, Zhu R, Chen Z, Wang Z, et al. Epithelial-mesenchymal transition in asthma airway remodeling is regulated by the IL-33/CD146 axis. Front Immunol. 2020;11:1598. doi: 10.3389/fimmu.2020.01598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Taniguchi S, Elhance A, Van Duzer A, Kumar S, Leitenberger JJ, Oshimori N (2020) Tumor-initiating cells establish an IL-33-TGF-beta niche signaling loop to promote cancer progression. Science. 369(6501). 10.1126/science.aay1813. [DOI] [PMC free article] [PubMed]
  • 147.Wiesner DL, Smith KD, Kotov DI, Nielsen JN, Bohjanen PR, Nielsen K. Regulatory T cell induction and retention in the lungs drives suppression of detrimental type 2 Th cells during pulmonary cryptococcal infection. J Immunol. 2016;196(1):365–374. doi: 10.4049/jimmunol.1501871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Eastman AJ, He X, Qiu Y, Davis MJ, Vedula P, Lyons DM, et al. Cryptococcal heat shock protein 70 homolog Ssa1 contributes to pulmonary expansion of Cryptococcus neoformans during the afferent phase of the immune response by promoting macrophage M2 polarization. J Immunol. 2015;194(12):5999–6010. doi: 10.4049/jimmunol.1402719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Dong F, Ruan S, Wang J, Xia Y, Le K, Xiao X, et al. M2 macrophage-induced lncRNA PCAT6 facilitates tumorigenesis and angiogenesis of triple-negative breast cancer through modulation of VEGFR2. Cell Death Dis. 2020;11(9):728. doi: 10.1038/s41419-020-02926-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27(1):109–118. doi: 10.1038/cr.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Supasorn O, Sringkarin N, Srimanote P, Angkasekwinai P. Matrix metalloproteinases contribute to the regulation of chemokine expression and pulmonary inflammation in Cryptococcus infection. Clin Exp Immunol. 2016;183(3):431–440. doi: 10.1111/cei.12725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Berois N, Pittini A, Osinaga E. Targeting tumor glycans for cancer therapy: successes, limitations, and perspectives. Cancers. 2022;14(3):645. doi: 10.3390/cancers14030645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Munkley J. Aberrant sialylation in cancer: therapeutic opportunities. Cancers. 2022;14(17):4248. doi: 10.3390/cancers14174248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Fernandes A, Azevedo CM, Silva MC, Faria G, Dantas CS, Vicente MM, et al. Glycans as shapers of tumour microenvironment: a sweet driver of T-cell-mediated anti-tumour immune response. Immunology10.1111/imm.13494 [DOI] [PubMed]
  • 155.Vajaria BN, Patel PS. Glycosylation: a hallmark of cancer? Glycoconj J. 2017;34(2):147–156. doi: 10.1007/s10719-016-9755-2. [DOI] [PubMed] [Google Scholar]
  • 156.Dos Reis JS, da Costa Rodrigues, Santos MA, Mendonca DP, do Martins Nascimento SI, Barcelos PM, de Correia Lima RG, et al. Glycobiology of cancer: sugar drives the show. Med. 2022;9(6):34. doi: 10.3390/medicines9060034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.dos Reis JS, Diniz-Lima I, Santos MARdC, Barcelos PM, da Costa KM, Valente RdC et al (2023) The blessed union of glycobiology and immunology: a marriage that worked. Med 10(2):15. 10.3390/medicines10020015 [DOI] [PMC free article] [PubMed]
  • 158.Rodrigues Mantuano N, Stanczak MA, Oliveira IA, Kirchhammer N, Filardy AA, Monaco G, et al. Hyperglycemia enhances cancer immune evasion by inducing alternative macrophage polarization through increased O-GlcNAcylation. Cancer Immunol Res. 2020;8(10):1262–1272. doi: 10.1158/2326-6066.CIR-19-0904. [DOI] [PubMed] [Google Scholar]
  • 159.Mantuano NR, Oliveira-Nunes MC, Alisson-Silva F, Dias WB, Todeschini AR. Emerging role of glycosylation in the polarization of tumor-associated macrophages. Pharmacol Res. 2019;146:104285. doi: 10.1016/j.phrs.2019.104285. [DOI] [PubMed] [Google Scholar]
  • 160.da Costa Santos MAR, Dos Reis JS, do Nascimento Santos CA, Costa KM, Barcelos PM, de Oliveira Francisco KQ, et al. Expression of O-glycosylated oncofetal fibronectin in alternatively activated human macrophages. Immunol Res. 2023;71(1):92–104. doi: 10.1007/s12026-022-09321-9. [DOI] [PubMed] [Google Scholar]
  • 161.Freire-de-Lima L, Gelfenbeyn K, Ding Y, Mandel U, Clausen H, Handa K, et al. Involvement of O-glycosylation defining oncofetal fibronectin in epithelial-mesenchymal transition process. Proc Natl Acad Sci U S A. 2011;108(43):17690–17695. doi: 10.1073/pnas.1115191108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Ding Y, Gelfenbeyn K, Freire-de-Lima L, Handa K, Hakomori SI. Induction of epithelial-mesenchymal transition with O-glycosylated oncofetal fibronectin. FEBS Lett. 2012;586(13):1813–1820. doi: 10.1016/j.febslet.2012.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Alisson-Silva F, Freire-de-Lima L, Donadio JL, Lucena MC, Penha L, Sa-Diniz JN, et al. Increase of O-glycosylated oncofetal fibronectin in high glucose-induced epithelial-mesenchymal transition of cultured human epithelial cells. PLoS One. 2013;8(4):e60471. doi: 10.1371/journal.pone.0060471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.da Fonseca LM, Calvalhan DM, Previato JO, Mendonca Previato L, Freire-de-Lima L. Resistance to paclitaxel induces glycophenotype changes and mesenchymal-to-epithelial transition activation in the human prostate cancer cell line PC-3. Tumour Biol. 2020;42(9):1010428320957506. doi: 10.1177/1010428320957506. [DOI] [PubMed] [Google Scholar]
  • 165.Freire-de-Lima L. Sweet and sour: the impact of differential glycosylation in cancer cells undergoing epithelial-mesenchymal transition. Front Oncol. 2014;4:59. doi: 10.3389/fonc.2014.00059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.da Fonseca LM, da Silva VA, Freire-de-Lima L, Previato JO, Mendonca-Previato L, Capella MA. Glycosylation in cancer: interplay between multidrug resistance and epithelial-to-mesenchymal transition? Front Oncol. 2016;6:158. doi: 10.3389/fonc.2016.00158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Huang J, Lan C, Li H, Chen S, Lin Q, Weng H. Concomitant lung adenocarcinoma and pulmonary cryptococcosis confirmed by pathologic examinations. Med. 2019;98(50):e18316. doi: 10.1097/MD.0000000000018316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ugrakli M, Araz M, Demirkiran A, Celik AF, Karakurt Eryilmaz M, Karaagac M, et al. Pneumonitis associated with trastuzumab emtansine in a patient with metastatic breast cancer. J Oncol Pharm Pract. 2022;28(3):740–745. doi: 10.1177/10781552211066073. [DOI] [PubMed] [Google Scholar]
  • 169.Ji YZ, Ma ZM, Chang L, Liu SR. A novel case of cutaneous squamous cell carcinoma at the site of previous sporotrichosis. J Cosmet Dermatol. 2020;19(6):1487–1489. doi: 10.1111/jocd.13158. [DOI] [PubMed] [Google Scholar]
  • 170.Aslam S, Ghafoor S. Association of Candida species with novel SARS-CoV-2 and biomarkers for fungal premalignant oral lesions. J Pak Med Assoc. 2022;72(9):1827–1830. doi: 10.47391/JPMA.4632. [DOI] [PubMed] [Google Scholar]
  • 171.Badheeb A, Al Gharem N, Al Hammadi S, Elsagheer S, Badheeb M, Ahmed F. Primary pulmonary leiomyosarcoma with coexistent pulmonary aspergillosis: a case report. Pan Afr Med J. 2022;42:135. doi: 10.11604/pamj.2022.42.135.34116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Scimeca M, Urbano N, Bonfiglio R, Duggento A, Toschi N, Schillaci O, et al. Novel insights into breast cancer progression and metastasis: a multidisciplinary opportunity to transition from biology to clinical oncology. Biochim Biophys Acta Rev Cancer. 2019;1872(1):138–148. doi: 10.1016/j.bbcan.2019.07.002. [DOI] [PubMed] [Google Scholar]
  • 173.Barco I, Garcia-Font M, Garcia-Fernandez A, Fraile M, Gimenez N, Gonzalez S, et al. Breast cancer patients developing distant metastasis at follow-up: mortality-related factors. Breast J. 2021;27(3):291–293. doi: 10.1111/tbj.14159. [DOI] [PubMed] [Google Scholar]
  • 174.Ward RA, Fawell S, Floc’h N, Flemington V, McKerrecher D, Smith PD. Challenges and opportunities in cancer drug resistance. Chem Rev. 2021;121(6):3297–351. doi: 10.1021/acs.chemrev.0c00383. [DOI] [PubMed] [Google Scholar]
  • 175.De Las RJ, Brozovic A, Izraely S, Casas-Pais A, Witz IP, Figueroa A. Cancer drug resistance induced by EMT: novel therapeutic strategies. Arch Toxicol. 2021;95(7):2279–2297. doi: 10.1007/s00204-021-03063-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Li Y, Wang Z, Ajani JA, Song S. Drug resistance and cancer stem cells. Cell Commun Signal. 2021;19(1):19. doi: 10.1186/s12964-020-00627-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Blackstock R, Casadevall A. Presentation of cryptococcal capsular polysaccharide (GXM) on activated antigen-presenting cells inhibits the T-suppressor response and enhances delayed-type hypersensitivity and survival. Immunol. 1997;92(3):334–339. doi: 10.1046/j.1365-2567.1997.00357.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lipovsky MM, Tsenova L, Coenjaerts FE, Kaplan G, Cherniak R, Hoepelman AI. Cryptococcal glucuronoxylomannan delays translocation of leukocytes across the blood-brain barrier in an animal model of acute bacterial meningitis. J Neuroimmunol. 2000;111(1–2):10–14. doi: 10.1016/s0165-5728(00)00354-4. [DOI] [PubMed] [Google Scholar]
  • 179.Ellerbroek PM, Walenkamp AM, Hoepelman AI, Coenjaerts FE. Effects of the capsular polysaccharides of Cryptococcus neoformans on phagocyte migration and inflammatory mediators. Curr Med Chem. 2004;11(2):253–266. doi: 10.2174/0929867043456188. [DOI] [PubMed] [Google Scholar]
  • 180.Ibrahim AS, Filler SG, Alcouloumre MS, Kozel TR, Edwards JE, Jr, Ghannoum MA. Adherence to and damage of endothelial cells by Cryptococcus neoformans in vitro: role of the capsule. Infect Immun. 1995;63(11):4368–4374. doi: 10.1128/iai.63.11.4368-4374.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Chiapello LS, Baronetti JL, Aoki MP, Gea S, Rubinstein H, Masih DT. Immunosuppression, interleukin-10 synthesis and apoptosis are induced in rats inoculated with Cryptococcus neoformans glucuronoxylomannan. Immunol. 2004;113(3):392–400. doi: 10.1111/j.1365-2567.2004.01970.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Chiapello LS, Aoki MP, Rubinstein HR, Masih DT. Apoptosis induction by glucuronoxylomannan of Cryptococcus neoformans. Med Mycol. 2003;41(4):347–353. doi: 10.1080/1369378031000137260. [DOI] [PubMed] [Google Scholar]
  • 183.Retini C, Vecchiarelli A, Monari C, Bistoni F, Kozel TR. Encapsulation of Cryptococcus neoformans with glucuronoxylomannan inhibits the antigen-presenting capacity of monocytes. Infect Immun. 1998;66(2):664–669. doi: 10.1128/IAI.66.2.664-669.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Yauch LE, Lam JS, Levitz SM. Direct inhibition of T-cell responses by the Cryptococcus capsular polysaccharide glucuronoxylomannan. PLoS Pathog. 2006;2(11):e120. doi: 10.1371/journal.ppat.0020120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Jain N, Li L, McFadden DC, Banarjee U, Wang X, Cook E, et al. Phenotypic switching in a Cryptococcus neoformans variety gattii strain is associated with changes in virulence and promotes dissemination to the central nervous system. Infect Immun. 2006;74(2):896–903. doi: 10.1128/iai.74.2.896-903.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Jong A, Wu CH, Gonzales-Gomez I, Kwon-Chung KJ, Chang YC, Tseng HK, et al. Hyaluronic acid receptor CD44 deficiency is associated with decreased Cryptococcus neoformans brain infection. J Biol Chem. 2012;287(19):15298–15306. doi: 10.1074/jbc.M112.353375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Chen C, Zhao S, Karnad A, Freeman JW. The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol. 2018;11(1):64. doi: 10.1186/s13045-018-0605-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Freire-de-Lima CG, Nascimento DO, Soares MB, Bozza PT, Castro-Faria-Neto HC, de Mello FG, et al. Uptake of apoptotic cells drives the growth of a pathogenic trypanosome in macrophages. Nature. 2000;403(6766):199–203. doi: 10.1038/35003208. [DOI] [PubMed] [Google Scholar]
  • 189.Xiao YQ, Freire-de-Lima CG, Schiemann WP, Bratton DL, Vandivier RW, Henson PM. Transcriptional and translational regulation of TGF-beta production in response to apoptotic cells. J Immunol. 2008;181(5):3575–3585. doi: 10.4049/jimmunol.181.5.3575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Hao Y, Huang Y, Chen J, Li J, Yuan Y, Wang M, et al. Exopolysaccharide from Cryptococcus heimaeyensis S20 induces autophagic cell death in non-small cell lung cancer cells via ROS/p38 and ROS/ERK signalling. Cell Prolif. 2020;53(8):e12869. doi: 10.1111/cpr.12869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Retini C, Kozel TR, Pietrella D, Monari C, Bistoni F, Vecchiarelli A. Interdependency of interleukin-10 and interleukin-12 in regulation of T-cell differentiation and effector function of monocytes in response to stimulation with Cryptococcus neoformans. Infect Immun. 2001;69(10):6064–6073. doi: 10.1128/IAI.69.10.6064-6073.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Brazilian Journal of Microbiology are provided here courtesy of Brazilian Society of Microbiology

RESOURCES