T-lymphocytes Expression of Toll-like Receptors 2 and 4 in Acute Myeloid Leukemia Patients with Invasive Fungal Infections 

Muhamad R. Abdel Hammed1*, Sherein G. Elgendy2*, Mohamed A. El-Mokhtar2, Douaa Sayed3, Samar M. Mansour3 and Abeer M. Darwish3.

Internal Medicine Department and Hematology Unit, Assiut University Hospitals and South Egypt Cancer Institute Bone Marrow Transplantation Unit, Assiut University, Assiut, Egypt.
2 Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt.
3 Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt.
* Both author equally contributed to the work.

Correspondence to: Muhamad R. Abdel Hameed, MD. Department of Internal Medicine  & Hematology Unit, Assiut University Hospitals and Bone Marrow Transplantation Unit, South Egypt Cancer Institute, Assiut University,  Assiut, Egypt. Tel:  (+2) 01097510010, Fax: +088-2080278. E-mail: dr.muhamadramadan@yahoo.com
Sherein G. Elgendy, Ph.D. Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Egypt. Tel.: (+2) 01021887728, Fax: +088-2080278. E-mail: Shereinelgendy@yahoo.com   

Published: March 1, 2022
Received: October 29, 2021
Accepted: February 8, 2022
Mediterr J Hematol Infect Dis 2022, 14(1): e2022022 DOI 10.4084/MJHID.2022.022

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background: Invasive fungal infections (IFIs) are important cause of mortality in acute myeloid leukemia (AML) patients on treatment with intensive induction chemotherapy. Toll-like receptors, mainly Toll-like receptors 2 and 4 (TLR2 and TLR4), play a considerable role in the host defense against microorganisms. The involvement of TLR signaling in modulation of innate immune response is extensively discussed, but the TLR expressions profiling on adaptive immune cells are not specified. Also, the expressions of TLRs and their association with the occurrence of IFIs in patients with AML are not studied. So, the novel aim of this study was to investigate the associations between the T-lymphocyte expression of TLR2 and TLR4 and the occurrence of IFIs in AML patients treated with intensive induction chemotherapy.
Materials and Methods:
One hundred twenty two newly diagnosed AML patients were evaluated. The laboratory diagnostic techniques for IFIs include culture, microscopic examination, histopathology, galactomannan assay and PCR. The expressions of TLR2 and TLR4 were analyzed by flow cytometry. The Control group included 20 age and sex-matched individuals. 

Results: There was a significant increase in the expression of TLR4 in AML patients with IFI compared to healthy controls (p=0.001). TLR2 and TLR4 expressions increased significantly in AML patients with mixed fungal and bacterial infection compared to healthy controls (p= 0.002 and p=0.001, respectively).
Conclusion:
TLRs expressions could be important biological markers for the occurrence of IFI in non-M3 AML patients after intensive induction chemotherapy.



Introduction

Acute myeloid leukemia (AML) represents the hematologic malignancy with the highest risk of invasive fungal infections (IFIs). The overall mortality rate in AML patients due to fungal infections was improved in recent years to 20%-30%.[1] IFIs represent a considerable clinical problem due to the high costs of the prophylaxis and treatment of fungal infections in limited resource localities.[2]
 Multiple risk factors can predispose AML  patients to develop fungal infections including old ages, pulmonary comorbidities, duration of neutropenia, relapse/refractory disease, intense chemotherapy, and a high dose of steroids.[3]
The Infectious Diseases Working Party of the German Society of Hematology and Medical Oncology (AGIHO) postulates that prolonged severe neutropenia in AML patients (<500 cells/μL of at least 8 days) post- induction/consolidation chemotherapy or allogeneic stem cell transplantation are considered as individuals at high-risk for IFI.[4]
Diagnosis of IFI is challenging, particularly in AML patients as symptoms can be absent or subtle. Fever may be the only sign. Thrombocytopenia and coagulopathy due to the underlying cause and chemotherapy may impair the ability to tissue biopsy which is the preferred method for diagnosis establishment.5 The European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group (EORTC) and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (MSG) defining IFI as proven, probable, and possible infections.[5,6]
Recognition of fungi by immune cells is mediated through pattern recognition receptors (PRRs); like Toll-like receptors (TLRs) and C-type lectins (CLRs). Binding of fungal pathogen-associated molecular patterns (PAMPs) to PRRs triggers phagocytes to the infection site, microbial killing, and dendritic cells (DCs) activation.[7,8]
Toll-like receptors are widely expressed on myeloid cells of innate immune system, such as macrophages, DCs.[9] TLR signaling in DCs triggers a maturation program that increases their ability to prime naïve T cells through up-regulation of MHC and co-stimulatory molecules and expression of pro-inflammatory cytokines, such as TNF-α, IL-1, and IL-6.[10]
TLRs have been considered traditionally to play an important role in the innate immune system. However, other few studies have found that TLRs are also expressed on various adaptive immune cells, such as B cells,[11] CD4+ and CD8+ T cells,[12] and the CD4+CD25+ regulatory T cell population.[13] Two studies, sorted CD4+CD45high T cells from C57/BL6 (B6) mice were found to express TLR1, 2, 3, 6, 7 and 8, but low levels of TLR 4, 5 and 9 mRNA.[14] Naïve CD8+ T cells from B6 mice were reported to express mRNA for TLR1, 2, 6 and 9 but not TLR4.[15] Naïve CD4+ T cells from BALB/c mice express mRNA for TLR3, 4, 5 and 9.[16] Thus, the involvement of TLR signaling in modulation of immune response is not limited to innate immune cells, but also modulate cellular and humoral adaptive immunity. TLR2 and TLR4 are two of the most studied TLRs to have an important role in the recognition of both bacterial and fungal pathogens.17 So, we have focused on the associations between T-lymphocyte expression of TLR2 and TLR4 and the occurrence of IFIs in AML patients which remains unclear.


Materials and Methods

Ethics Statement. This study was approved by the Regional Ethical Committee in South Egypt Cancer Institute (SECI), Assiut University, in accordance with the provisions of the Declaration of Helsinki. Informed written consent obtained from all participants before enrolment.
Study Design and Setting. This study was performed at Clinical Hematology Unit, Internal Medicine Department, Assiut University Hospital, and South Egypt Cancer Institute (SECI), Assiut University, Egypt. All newly diagnosed AML patients (aged more than 18 years old), admitted in the duration from October 2017 to July 2020 were enroll in this study. The diagnosis was performed according to the WHO criteria for AML. [18] The intensive induction chemotherapy was (idarubicin 12 mg/m2 per day for 2–3 days, and cytarabine 100 mg/m
2/day for 5–7 days). Patients received prophylactic treatment during the period of neutropenia following chemotherapy (sulfamethoxazole 400 mg/ trimethoprim 80 mg once or twice daily). Patients receiving antifungal prophylaxis or preexisting antifungal treatment were excluded. Also, AML with antecedent hematologic malignancies like Myelodysplastic syndrome, and Myeloproliferative neoplasms, AML M3, relapsed AML patients and chemotherapy courses with low-intensity regimen were excluded. Baseline demographic and clinical data, type of AML, chemotherapy courses, duration of febrile neutropenia, complete blood cell count, cytogenetic risks, radiological examination" high-resolution chest computed tomography (CT)", IFI incidence, site of fungal infection, and patients outcome were recorded. Twenty age and sex-matched healthy individuals were the control group in this study.
Diagnosis of Invasive Fungal Infection. Diagnosis of IFI was applied according to 2008 consensus criteria of the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group (EORTC) and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (MSG), which classified IFI into possible, probable, or proven IFI.[5] Proven IFI requires that a fungus be detected by culture or pathohistological blood analysis in a sterile site sample. Probable IFI requires lesions on imaging indicative of fungal infection and mycological evidence, not only culture and pathohistological analysis of a sample but also indirect tests, such as galactomannan. Possible IFI only requires imaging lesions indicating fungal infection without presence mycological evidence.
Neutropenia was defined as a neutrophil count <500 cells/ μL.[19] The duration of neutropenia in each course of chemotherapy was collected. When patients remain febrile neutropenic >72 hrs after antibacterial agent, a thorough history and physical examination were recorded, along with culture for blood and other potentially infectious focuses including oral mucositis grade ΙΙΙ or ΙѴ, or lower respiratory tract infection (LRTI). For patients with no identified focuses, high resolution computed tomography (CT) was performed, together with galactomannan (GM) assay and PCR. Broncho-alveolar lavage (BAL) was not performed routinely. Fluconazole 400 mg IV/day was given if IFI were suspected with the CT findings, positive galactomannan or PCR assays, or other clinical evidence.
Sample Collection and Processing. Blood, oral swabs, and sputum samples were collected from the patients according to their clinical presentation and different localizing signs and symptoms before the initiation of antifungal therapy.
Identification of Candida spp. Blood cultures were done by adding 5-10 mL blood to 50-100 mL Sabouraud dextrose broth (Himedia, India) and incubated at 37ºC for 10 days with subculture every other day.[20] Oral swabs and sputum samples were cultivated on Sabouraud dextrose agar (Himedia, India) with chloramphenicol (16 mg/mL). The isolates were further identified by colony morphology on CHROMagar® Candida medium (CHROMagar, Paris, France), germ tube test, chlamydospores on Tween 80 cornmeal agar (Difco) and growth at 45°C.[21]
Identification of Mold. Direct microscopic examination is performed on a fresh sample between a glass slide and coverslip. The morphological characteristics of Aspergillus spp are the presence of hyphae (hyaline and septate) with dichotomous branches at 45° angles and with uniform width (3–6 µm). However, it is hard to distinguish the species of Aspergillus because of the difficulty in distinguishing the morphology of the different fungi species. Aspergillus spp were cultivated on Sabouraud’s dextrose-agar at 37 °C for 2 to 5 days. Fungi that grew in culture were identified according to morphological and microscopic criteria and Roth’s flag technique.[22,23] In addition, Patient sera were tested for galactomannan (GM) by Galactomannan ELISA kits according to the manufacturer instructions[24] (Bio-Rad, Hercules, CA). The presence of bacterial infections was tested by VITEK® 2 system.
DNA Extraction and PCR Amplification. DNA extraction was performed using a commercial kit (QIAamp DNA Mini Kit (Qiagen, Germany)) according to the manufacturer’s instructions. PCR was performed utilizing the fungus specific, universal primer pair ITS1 (ʹ5TCCGTAGGTGAACCTGCGG3ʹ) which hybridizes at the 3ʹ end of 18S rDNA and ITS4 (ʹ5TCC TCC GCTTATGATAT GC3ʹ) which hybridizes at the 5ʹend of 28S rDNA (Sigma, USA).[25] The concentration was measured by a NanoDrop ND-1000 spectrophotometer (NanoDrop Technologies). The PCR reaction mix contained 0.5 μM of each primer, 10 μM deoxynucleotides, 1.5 mM MgCl2 and 1 x buffer (Promega). One unit of the Taq Polymerase (Promega) was added to each tube. DNA amplification was carried out in a Gene Amp9600 thermal cycler under the following conditions: 35 cycles of denaturing at 94°C for 1 min; annealing at 55.5°C for 2 min and extension at 72°C for 2 min; and final extension at 72° for 10 min.[25,26] PCR products were visualized by electrophoresis on a 1% agarose gel stained with ethidium bromide.
Flow Cytometry. Whole blood samples (anticoagulated with EDTA) were collected from enrolled individuals and stained with the following antibodies (all from BD bioscience, USA); Alexa fluor 488-conjugated anti-CD282 for detection of TLR2, PE-conjugated anti-CD284|MD-2 complex for detection of TLR4, PerCP-conjugated anti-HLA-DR, and APC-conjugated anti-CD3. RBCs were lysed with the lysis buffer, then at least 10.000 events were acquired and analyzed by FACS Caliber flow cytometer (BD bioscience, USA). Appropriate isotype-matched controls were included in the experiments to identify positive populations.[27] Data was analyzed with cell Quest software (BD bioscience, USA), (Figure 1).


Figure 1 Figure 1. Representative gating strategy used to detect the TLR2+ and TLR4+ T cells. Blood samples were stained with PerCP-conjugated anti-HLA-DR, APC-conjugated anti-CD3, Alexa fluor 488-conjugated anti-CD282 (TLR2), and PE-conjugated anti CD284 (TLR4). Lymphocytes were selected (R1) and then gated on CD3+ cells (R2) and HLA-DR+ cells (R3). R2 was further analyzed for TLR2 and TLR4 expression.

Statistical Data. Descriptive results of continuous variables were expressed as mean±SE for non-parametric variables and as mean±SD for parametric variables. Comparison of the demographic characteristics between cases and control was calculated using the chi-square test for categorical data and independent sample t-test for numerical variables. Qualitative variables were expressed as the number of positive cases (%). Differences in mean values of TLR2 and TLR4 level of expression between different groups were calculated using the Mann-Whitney test. P-value was considered significant at ˂ 0.05. Statistical calculation was performed with the statistical package for social science software (SPSS version 16.0 Inc, Chicago, III).

Results

From 2017 to 2020, 122 newly diagnosed non-M3 AML patients (aged more than 18) who received induction chemotherapy were admitted to Clinical Hematology Unit, Internal Medicine Department, Assiut University Hospital, South Egypt Cancer Institute (SECI). Forty patients (32.78%) developed IFIs. The demographic and clinical characteristics of these 40 non-M3 AML patients with IFIs were presented in Table 1. The median age was 38.8 years (range, 18–65 years); male patients were 27 (67.5%). The diagnosis was applied according to the WHO criteria for AML. There were mainly AML4, AML1 and AML2 (30 %, 25% and 22.5% respectively). Eleven (27.5%) patients were favorable-cytogenetic group, 9 (22.5%) poor group, and 20 (50%) the intermediate- risk group. No significant differences were found between TLR4 or TLR2 expressions and age, sex, Type of AML, cytogenetic risk and Patient’s outcome (P > 0.05).
The most common sites of infection were the lower respiratory tract 47.5% (19/40), and oral mucosa (mucositis grade ӀӀ or ӀѴ) 37.5% (15/40). Mixed infection sites (bloodstream, oral, and LRTI) were detected only in 15% (6/40), Table 1.


Table 1 Table 1. Demographic and clinical characterization of the AML patients with IFI

The fungal pathogens among the 40 AML patients was identified as 2 (5%) proven, 28 (70%) probable, and 10 (25%) possible IFIs. The pure fungal growth was observed in 24 patients, whereas mixed bacterial and fungal growth was encountered in 16 patients. Candida species was the most encountered fungi. It was present in 21 specimens (2 specimens were mixed candida and mold pathogen) followed by Aspargillus in 19 specimens then penicillum in 2 specimen, Table 2.

Table 2 Table 2. Identified fungal pathogens* among the 40 AML patients.

TLR2 expression in AML patients with IFIs in comparison to healthy controls showed no significant difference (p ꞊ 0.659), while there was a significant increase in the expression of TLR4 in AML patients with IFI compared to healthy controls (p = 0.001). TLR2 and TLR4 expression in AML patients with no IFI in comparison to healthy controls had no significant difference (p ꞊ 0.72, 0.69 respectively), Table 3.

Table 3 Table 3. TLR2 and TLR4 expression level in comparison between patients and control group.

Moreover, we observed that TLR2 expression increased significantly in AML patients with mixed fungal and bacterial infections compared to healthy controls (p = 0.002). Also, TLR4 expression in AML patients with mixed fungal and bacterial infection was significantly increased (p = 0.001), Table 4.

Table 4 Table 4. TLR2 and TLR4 expression level in comparison between AML patients with mixed fungal and bacterial infection and healthy controls.


Discussion

This is the first study about T-lymphocytes expressions of TLRs and the development of IFIs in AML patients receiving induction chemotherapy in Assiut University Hospitals, and up to our knowledge in Egypt. We reported that the overall incidence of IFIs in AML patients is 40/122 (32.78%), this incidence is considered high in comparison with other reports from different countries.[28-30] In these reports, the incidences of IFIs in AML patients varied from 4.0% to 48.4%. This variation is due to differences in patient populations, chemotherapy regimens, antifungal prophylaxis, and geographic variation. Recent study reported that (29%) of AML patients developed an IFI. Patients with AML remain at risk for IFI despite the use of several different antifungal agents for prophylaxis.[31]
The high incidence in our study can be explained by many factors, including limited health resources, the lack of routinely administered anti-fungal prophylaxis, and environmental factors such as high temperature, which facilitates fungal growth. This high incidence of IFIs should start a new cost-effectiveness consideration about the requirement of anti-fungal prophylaxis in AML patients with induction chemotherapy. Hagiwara et al.[32] reported that AML in developing countries with limited health resources, favors the health authorities to use their low budget preferentially in another illness that has a higher incidence and a better chance for achievement of higher social impact.
The fungal pathogens among the 40 AML patients was identified as 2 (5%) proven, 28 (70%) probable, and 10 (25%) possible IFIs. Tang et al.[33] reported that the incidence of all-category IFIs was 34.6% (5.7% proven IFIs, 5.0% probable IFIs and 23.8% possible IFIs). Nucci et al.[34] report a Brazilian incidence of (18.7%) for proven/probable IFIs in AML patients after diagnosis.  Kim et al.[19] reported (9.6%) with 20 IFI diagnosed following HMA (three proven, four probable, 13 possible).
In our study the pure fungal growth was observed in 24 (60%) patients, whereas mixed bacterial and fungal growth was encountered in 16 (40%) patients. Candida species was the most encountered fungi. It was present in 21 (50%) specimens (including 2 specimens were mixed candida and mold pathogen) followed by Aspargillus in 19 (45.2%) specimens then penicillum in 2 (4.8%) specimens. This result was different from Tang et al.[33] who reported that Candida spp still predominated and almost twice as common as Aspergillus spp. The reasons for this difference are mostly due to difference in number of patients enrolled, different specimen types and the absence of anti-yeast azole prophylaxis.
The reports in Egypt are very limited; an Egyptian study conducted on high-risk pediatric cancer patients by EL-mahallawy et al.[35] reported that yeast was isolated in (78.6%) of specimen and molds in (21.11%). Among yeasts, Candida was the commonest, while the most encountered molds were Aspergillus spp. They found that polymicrobial (mixed bacterial and fungal growth) was encountered in 62.5% of specimen, which is in great accordance with our results.
In this study, non-albicans Candida spp. (C. tropicalis and C. parapsilosis) were common 8/21 (38.1%) as C. albicans 13/21 (61.9%). Another study with similar findings postulated that neutropenia is correlated with non-albican Candida infections.[36]
An Egyptian study reported that 75 (44.1%) Candida spp (25 (33.3%) non-albicans Candida spp and 50 (66.6%) C. albicans) were isolated from AML patients on induction chemotherapy.[37] The common site of IFI was the lower respiratory tracts (47.5%, 19/40), and oral mucosa (mucositis grade ӀӀ or ӀѴ) (37.5%, 15/40) followed by mixed infection sites (bloodstream, oral and LRTI) (15%, 6/40). Pagano et al.[38] and Tang et al.[33] reported that lower respiratory tract was the most common site for IFIs (80% and 75.4% respectively); also EL-mahallawy et al.[35] and Kurosawa et al.[30] found an incidence of (35.7% and 55.3% respectively) for IFIs affecting the lung. Few articles evaluated the risk factors of IFIs in AML patients during induction chemotherapy. In this study, we have determined these risk factors as standard induction chemotherapy, febrile neutropenia, elderly and male gender. Tang et al.[35] postulated similar risk factors including standard induction chemotherapy, younger than 40 or older than 60 years, and a poor chemotherapy response for all-category IFIs. Neofytos et al.[39] postulated that mucositis and organ dysfunctions are important risk factors for invasive candidiasis during induction chemotherapy, and male gender is the only risk factor for mold infection. Hammond et al.[29] also reported male gender as risk factors for IFIs. Chen et al.[40] stated that AML patients have multiple risk factors for developing invasive fungal diseases, such a including advanced age, prolonged and profound neutropenia, the presence of indwelling catheters, and individual genetic susceptibilities. Previous results indicate the heterogeneity of the study subjects and treatment protocols.
The exact role of TLRs in the development of invasive fungal infection in AML patients is unknown. Numerous endogenous and exogenous factors affect cell proliferation and play critical roles in cancer development. The expression level of TLRs may depend on the environment, subset, cell type, stimulus and probably age group.[41
In the current study, no statistically significant differences were found between TLR4 or TLR2 expressions and age, sex, cytogenetic risk and Patient’s outcome (
P > 0.05). Similar results showed by Ramzi et al.[42] postulated that expressions of TLR2 did not show significant differences in cytogenetic abnormalities status (P = 0.67). The expression of TLR4 was not different in favorable, intermediate and poor risk groups (P = 0.97). Renshaw et al[41] reported that old age could have negative effects on TLR expression and function, and therefore leads to increased susceptibility to infections and poor adaptive immune responses. 
The current study included 122 newly diagnosed non-M3 AML patients and reporting no statistically significant differences between TLR4 or TLR2 expressions and type of AML (
P > 0.05). In the same context, Ramzi et al.[42] observe a higher expression of TLR2 in AML-M3 cases compared to non-M3 AML patients (P = 0.015).
Human T cells isolated from peripheral blood reported to express mRNA for most TLRs, with considerable variation in the reported expression levels. Protein expression of TLR2, 3, 4, 5 and 9 has also been detected by flow cytometry.[43] The current study revealed that TLR2 expression in AML patients with IFIs in comparison to healthy controls presented no significant difference (p ꞊ 0.659), while there was a significant increase in the expression of TLR4 in the same patients group compared to healthy controls (p = 0.001). Consistent with these findings is the study of Bellocchio, Montagnoli. They reported that TLR4 but not TLR2 participated in host defense against A. fumigatus.[44]  In addition, Chai et al.[45]  stated that after stimulation with A. fumigatus conidia, surface TLR2 expression is markedly reduced compared to TLR4 expression, this suggests that A. fumigatus conidia induced depletion and downregulation of the TLR2-mediated pathway involved in the receptor internalization together with Aspergillus conidia into the phagosome, resulting in decreased TLR2 expression on the cell membrane. Chai et al [45] suggested a possible explanation for these findings as they postulated that the balance between TH1 and TH2 immune system pathways is necessary for the pathogen clearance and limitation of inflammation. TLR4 favors the production of TH1 response with pro-inflammatory cytokine production such as IFN-γ and 1L- 12, which induces protective antifungal defense mechanisms. T regulatory cells induced by TH2 response mediated by TLR2 signaling are needed to lower immune response and to avoid collateral damage after antifungal TH1 response mediated by TLR4 signaling.
Our result revealed that TLR2 and TLR4 expression in patients with polymicrobial infection (fungus and bacteria) are significantly increased as compared to healthy controls. This result agreed with the result of Armstrong et al.[46] who reported that expression of TLR2 and TLR4 in septic patients was significantly up-regulated compared with the expression of these receptors in healthy individuals. Tsujimoto et al.[47] stated also that septic patients display significantly up-regulated TLR expression in various organs.
We can conclude that in polymicrobial infection (fungus and bacteria) there is a marked increase of both TLR2 and TLR4 expression and this may be due to the powerful effect of bacterial LPS and other bacterial PAMPs that augment the stimulatory effect of fungal PAMPs.
Susceptibility to infections is determined by the malignant disease and its treatment, environmental factors (e.g. nutritional status and hygiene of the patient), and genetically determined variations of the immune system. Some genetic polymorphisms in the innate immune system, such as profound mannose-binding lectin deficiency and TLR polymorphism associated with an increased risk of infections. Mutations in genes encoding TLRs or downstream signaling proteins increase the risk of infection.[48]
Numerous polymorphisms and mutational inactivation have been described in TLRs and appear to have clinical significance,[48,47] reported that severely septic patients with bad general conditions and the unfavorable clinical outcome did not have increased expression of TLRs,[49] have observed that a decrease in TLR2 expression in patients with invasive candidiasis can lead to severe disseminated infection. On the other side,[50] found that mice with non-functional TLR4 showed increased fungal load in the kidneys and deficiencies in neutrophil upon C. albicans challenge when compared to TLR4 responsive mice.


Conclusions

The incidence of IFIs is high in AML patients who received induction chemotherapy in Assiut University Hospitals. TLR2 and TLR4 expressions in AML patients with IFI are related to invasiveness and dissemination of fungal infection. TLRs expressions could be important biological markers for the occurrence of IFI in non-M3 AML patients after intensive induction chemotherapy. Additional larger studies including a larger number of patients and detection of proinflammatory cytokines are necessary to confirm the immunological relation between TLR and fungal infection in AML patients.

Data Availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Author Contributions

M.R.A. and D.S. conceived and designed the research. D.S., M.R.A. and S.M.M. recruited patients, carried out the clinical investigations, collected clinical data. S.G.E. and M.A.E. contributed in the interpretation of data for the work. D.S., M.R.A., S.M.M., S.G.E. and M.A.E. prepared the original draft of the manuscript. All authors contributed to data analysis, drafting or revising the article, have agreed on the journal to which the article will be submitted, gave final approval of the version to be published, and agree to be accountable for all aspects of the work.

References   

  1. Wang ES. Common fungal infections in patients with leukemia. Clin Adv Hematol Oncol. 2017;15(5):352-4.
  2. Heimann SM, Vehreschild MJ, Cornely OA, Franke B, von Bergwelt-Baildon M, Wisplinghoff H, et al. A cost and resource utilization analysis of micafungin bridging for hemato-oncological high-risk patients undergoing allogeneic stem cell transplantation. Eur J Haematol. 2015;94(6):526-31. https://doi.org/10.1111/ejh.12466 PMid:25310918
  3. Rambaldi B, Russo D, Pagano L. Defining Invasive Fungal Infection Risk in Hematological Malignancies: A New Tool for Clinical Practice. Mediterr J Hematol Infect Dis. 2017;9(1):e2017012. https://doi.org/10.4084/mjhid.2017.012 PMid:28101316 PMCid:PMC5224802
  4. Heinz WJ, Buchheidt D, Christopeit M, von Lilienfeld-Toal M, Cornely OA, Einsele H, et al. Diagnosis and empirical treatment of fever of unknown origin (FUO) in adult neutropenic patients: guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO). Ann Hematol. 2017;96(11):1775-92. https://doi.org/10.1007/s00277-017-3098-3 PMid:28856437 PMCid:PMC5645428
  5. DePauw B, Walsh TJ, Donnelly JP, Stevens DA, Edwards JE, Calandra T, et al. Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) C. Clin Infect Dis. 2008;46(12): 1813-21. https://doi.org/10.1086/588660 PMid:18462102 PMCid:PMC2671227
  6. J Peter Donnelly, Sharon C Chen, Carol A Kauffman, William J Steinbach, John W Baddley, Paul E Verweij, Cornelius J Clancy et al. Revision and Update of the Consensus Definitions of Invasive Fungal Disease From the European Organization for Research and Treatment of Cancer and the Mycoses Study Group Education and Research Consortium, Clinical Infectious Diseases, 2020; 71( 6)15: 1367-1376.
  7. Ferwerda G, Netea MG, Joosten LA, van der Meer JW, Romani L, Kullberg BJ. The role of Toll-like receptors and C-type lectins for vaccination against Candida albicans. Vaccine. 2010 Jan 8;28(3):614-22. doi: 10.1016/j.vaccine.2009.10.082. Epub 2009 Nov 1. PMid:19887129
  8. Goyal S, Castrillón-Betancur JC, Klaile E, Slevogt H. The Interaction of Human Pathogenic Fungi With C-Type Lectin Receptors. Front Immunol. 2018 Jun 4;9:1261. doi: 10.3389/fimmu.2018.01261. https://doi.org/10.3389/fimmu.2018.01261 PMid:29915598 PMCid:PMC5994417
  9. McGettrick AF, O'Neill LA. Tolllike receptors: key activators of leucocytes and regulator of haematopoiesis. Br JHaematol (2007) 139:185. doi: 10.1111/j.1365-2141.2007.06802.x https://doi.org/10.1111/j.1365-2141.2007.06802.x PMid:17897294
  10. Zanin-Zhorov A and Cohen IR (2013) Signaling via TLR2 and TLR4 directly down-regulates T cell effector functions: the regulatory face of danger signals. Front. Immunol. 4:211. doi: 10.3389/fimmu.2013.00211 https://doi.org/10.3389/fimmu.2013.00211 PMid:23898332 PMCid:PMC3722573
  11. Gururajan M, Jacob J, Pulendran B. Toll-like receptor expression and responsiveness of distinct murine splenic and mucosal B-cell subsets. PLoS ONE (2007) 2:e863. doi:10. 1371/journal.pone.0000863 https://doi.org/10.1371/journal.pone.0000863 PMid:17848994 PMCid:PMC1955832
  12. Zanin-Zhorov A, Nussbaum G, Franitza S, Cohen IR, Lider O. Tcells respondtoheatshockprotein60via TLR2: activation of adhesion and inhibition of chemokine receptors. FASEB J (2003) 17:1567. https://doi.org/10.1096/fj.02-1139fje PMid:12824285
  13. Zanin-Zhorov A, Cahalon L, Tal G, Margalit R, Lider O, Cohen IR. Heat shock protein 60 enhances CD4+ CD25+ regulatory T cell function via innate TLR2 signaling. J Clin Invest (2006) 116:2022. doi: 10.1172/JCI28423 https://doi.org/10.1172/JCI28423 PMid:16767222 PMCid:PMC1474819
  14. Tomita T, Kanai T, Fujii T, Nemoto Y, Okamoto R, Tsuchiya K, et al. MyD88-dependent pathway in T cells directly modulates the expansion of colitogenic CD4+ T cells in chronic colitis. J Immunol. 2008; 180:5291. https://doi.org/10.4049/jimmunol.180.8.5291 PMid:18390710
  15. Cottalorda A, Verschelde C, Marcais A, Tomkowiak M, Musette P, Uematsu S, et al. TLR2 engagement on CD8 T cells lowers the threshold for optimal antigen-induced T cell activation. Eur J Immunol. 2006; 36:1684. https://doi.org/10.1002/eji.200636181 PMid:16761317
  16. Gelman AE, Zhang J, Choi Y, Turka LA. Toll-like receptor ligands directly promote activated CD4+ T cell survival. J Immunol. 2004; 172:6065.
    https://doi.org/10.4049/jimmunol.172.10.6065 PMid:15128790 PMCid:PMC2833313
  17. Netea MG, Van der Meer JW, Kullberg BJ (2007). Recognition of fungal pathogens by Toll-like receptors. In Immunology of Fungal Infections. Eur J Clin Microbiol Infect Dis, 23, 672-6. https://doi.org/10.1007/1-4020-5492-0_11
  18. Sabattini E, Bacci F, Sagramoso C, Pileri SA. WHO classification of tumours of haematopoietic and lymphoid tissues in 2008: an overview. Pathologica. 2010;102(3):83-7.
  19. Kim GYG, Burns J, Freyer CW, Hamilton KW, Frey NV, Gill SI, et al. Risk of invasive fungal infections in patients with high-risk MDS and AML receiving hypomethylating agents. Am J Hematol. 2020;95(7):792-8. https://doi.org/10.1002/ajh.25808 PMid:32242967
  20. Power D, Johnson J. Difco™ and BBL™ manual: manual of microbiological culture media. Becton Dickinson and Company, Sparks; 2009.
  21. Pincus DH, Orenga S, Chatellier S. Yeast identification--past, present, and future methods. Med Mycol. 2007;45(2):97-121. https://doi.org/10.1080/13693780601059936 PMid:17365647
  22. Fromtling R, Rhodes J, Dixon D. Taxonomy, classification, and morphology of the Fungi. J Manual of clinical microbiology. 2003;2:1653-8.
  23. Desoubeaux G, Bailly E, Chandenier J. Diagnosis of invasive pulmonary aspergillosis: updates and recommendations. Med Mal Infect. 2014;44(3):89-101. https://doi.org/10.1016/j.medmal.2013.11.006 PMid:24548415
  24. Ascioglu S, Rex JH, de Pauw B, Bennett JE, Bille J, Crokaert F, et al. Defining opportunistic invasive fungal infections in immunocompromised patients with cancer and hematopoietic stem cell transplants: an international consensus. Clin Infect Dis. 2002;34(1):7-14. https://doi.org/10.1086/323335 PMid:11731939
  25. Karakousis A, Tan L, Ellis D, Alexiou H, Wormald PJ. An assessment of the efficiency of fungal DNA extraction methods for maximizing the detection of medically important fungi using PCR. J Microbiol Methods. 2006;65(1):38-48 https://doi.org/10.1016/j.mimet.2005.06.008 PMid:16099520
  26. Williamson EC, Leeming JP, Palmer HM, et al. Diagnosis of invasive aspergillosis in bone marrow transplant recipients by polymerase chain reaction. Br. J. Haematol. 2000;108 (1):132-139. https://doi.org/10.1046/j.1365-2141.2000.01795.x PMid:10651736
  27. Kurt-Jones EA, Mandell L, Whitney C, Padgett A, Gosselin K, Newburger PE, et al. Role of toll-like receptor 2 (TLR2) in neutrophil activation: GM-CSF enhances TLR2 expression and TLR2-mediated interleukin 8 responses in neutrophils. Blood. 2002;100(5):1860-8. https://doi.org/10.1182/blood.V100.5.1860.h81702001860_1860_1868 PMid:12176910
  28. Gomes MZ, Mulanovich VE, Jiang Y, Lewis RE, Kontoyiannis DP. Incidence density of invasive fungal infections during primary antifungal prophylaxis in newly diagnosed acute myeloid leukemia patients in a tertiary cancer center, 2009 to 2011. Antimicrob Agents Chemother. 2014;58(2):865-73. https://doi.org/10.1128/AAC.01525-13 PMid:24277033 PMCid:PMC3910838
  29. Hammond SP, Marty FM, Bryar JM, DeAngelo DJ, Baden LR. Invasive fungal disease in patients treated for newly diagnosed acute leukemia. Am J Hematol. 2010;85(9):695-9. https://doi.org/10.1002/ajh.21776 PMid:20652970
  30. Kurosawa M, Yonezumi M, Hashino S, Tanaka J, Nishio M, Kaneda M, et al. Epidemiology and treatment outcome of invasive fungal infections in patients with hematological malignancies. Int J Hematol. 2012;96(6):748-57. https://doi.org/10.1007/s12185-012-1210-y PMid:23111539
  31. Wasylyshyn A, Linder KA, Castillo CG, Zhou S, Kauffman CA, Miceli MH. Breakthrough Invasive Fungal Infections in Patients with Acute Myeloid Leukemia. Mycopathologia. 2020;185(2):299-306. https://doi.org/10.1007/s11046-019-00418-8 PMid:31939052
  32. Hagiwara M, Sharma A, Chung KC, Delea TE. Burden of acute myeloid leukemia (AML) in a US commercially insured population. American Society of Clinical Oncology; 2017. https://doi.org/10.1200/JCO.2017.35.15_suppl.e18330
  33. Tang JL, Kung HC, Lei WC, Yao M, Wu UI, Hsu SC, et al. High Incidences of Invasive Fungal Infections in Acute Myeloid Leukemia Patients Receiving Induction Chemotherapy without Systemic Antifungal Prophylaxis: A Prospective Observational Study in Taiwan. PLoS One. 2015;10(6):e0128410. https://doi.org/10.1371/journal.pone.0128410 PMid:26061179 PMCid:PMC4462587
  34. Nucci M, Garnica M, Gloria AB, Lehugeur DS, Dias VC, Palma LC, et al. Invasive fungal diseases in haematopoietic cell transplant recipients and in patients with acute myeloid leukaemia or myelodysplasia in Brazil. Clin Microbiol Infect. 2013;19(8):745-51. https://doi.org/10.1111/1469-0691.12002 PMid:23009319
  35. El-Mahallawy HA, Shaker HH, Ali Helmy H, Mostafa T, Razak Abo-Sedah A. Evaluation of pan-fungal PCR assay and Aspergillus antigen detection in the diagnosis of invasive fungal infections in high-risk paediatric cancer patients. Med Mycol. 2006; 44(8):733-9. https://doi.org/10.1080/13693780600939955 PMid:17127630
  36. Chi HW, Yang YS, Shang ST, Chen KH, Yeh KM, Chang FY, et al. Candida albicans versus non-albicans bloodstream infections: the comparison of risk factors and outcome. J Microbiol Immunol Infect. 2011;44(5):369-75. https://doi.org/10.1016/j.jmii.2010.08.010 PMid:21524971
  37. Sayed SA, Hassan EAB, Abdel Hameed MR, Agban MN, Mohammed Saleh MF, Mohammed HH, Abdel-Aal ABM, Elgendy SG. Ketorolac-fluconazole: A New Combination Reverting Resistance in Candida albicans from Acute Myeloid Leukemia Patients on Induction Chemotherapy: In vitro Study. J Blood Med. 2021;12:465-474. https://doi.org/10.2147/JBM.S302158 PMid:34163275 PMCid:PMC8214543
  38. Pagano L, Girmenia C, Mele L, Ricci P, Tosti ME, Nosari A, et al. Infections caused by filamentous fungi in patients with hematologic malignancies. A report of 391 cases by GIMEMA Infection Program. Haematologica. 2001;86(8):862-70.
  39. Neofytos D, Lu K, Hatfield-Seung A, Blackford A, Marr KA, Treadway S, et al. Epidemiology, outcomes, and risk factors of invasive fungal infections in adult patients with acute myelogenous leukemia after induction chemotherapy. Diagn Microbiol Infect Dis. 2013;75(2):144-9. https://doi.org/10.1016/j.diagmicrobio.2012.10.001 PMid:23142166 PMCid:PMC3986043
  40. Chen MJ, HU Rong, Jiang XY, Wu Y, HE Z, Chen J, Zhan L.Dectin-1 rs3901533 and rs7309123 Polymorphisms Increase Susceptibility to Pulmonary Invasive Fungal Disease in Patients with Acute Myeloid Leukemia from a Chinese Han Population. Current Medical Science, 39(6):906-912,2019. https://doi.org/10.1007/s11596-019-2122-3 PMid:31845221
  41. Renshaw M, Rockwell J, Engleman C, et al. Cutting edge: impaired Toll-like receptor expression and function in aging. J Immunol. 2002;169(9):4697-701. https://doi.org/10.4049/jimmunol.169.9.4697 PMid:12391175
  42. Ramzi M, Khalafi-Nezhad A, Iravani Saadi M, Jowkar Z. Association between TLR2 and TLR4 Expression and Response to Induction Therapy in Acute Myeloid Leukemia Patients. Int J Hematol Oncol Stem Cell Res. 2018 Oct 1;12(4):303-312.  https://doi.org/10.18502/ijhoscr.v12i4.109 PMid:30774831 PMCid:PMC6375370
  43. Rahman AH, Taylor DK, Turka LA. The contribution of direct TLR signaling to T cell responses. Immunol Res. 2009;45(1):25-36. doi: 10.1007/s12026-009-8113-x. https://doi.org/10.1007/s12026-009-8113-x PMid:19597998 PMCid:PMC4486050
  44. Bellocchio S, Montagnoli C, Bozza S, Gaziano R, Rossi G, Mambula SS, et al. The contribution of the Toll-like/IL-1 receptor superfamily to innate and adaptive immunity to fungal pathogens in vivo. J Immunol. 2004;172(5):3059-69. https://doi.org/10.4049/jimmunol.172.5.3059 PMid:14978111
  45. Chai LY, Netea MG, Vonk AG, Kullberg BJ. Fungal strategies for overcoming host innate immune response. Med Mycol. 2009;47(3):227-36. https://doi.org/10.1080/13693780802209082 PMid:18654922
  46. Armstrong L, Medford AR, Hunter KJ, Uppington KM, Millar AB. Differential expression of Toll-like receptor (TLR)-2 and TLR-4 on monocytes in human sepsis. Clin Exp Immunol. 2004;136(2):312-9. https://doi.org/10.1111/j.1365-2249.2004.02433.x PMid:15086396 PMCid:PMC1809013
  47. Tsujimoto H, Ono S, Efron PA, Scumpia PO, Moldawer LL, Mochizuki H. Role of Toll-like receptors in the development of sepsis. Shock. 2008;29(3):315-21. https://doi.org/10.1097/SHK.0b013e318157ee55 PMid:18277854
  48. Pamer EG. TLR polymorphisms and the risk of invasive fungal infections. N Engl J Med. 2008;359(17):1836-8. https://doi.org/10.1056/NEJMe0806412 PMid:18946070 PMCid:PMC2630794
  49. Villamón E, Gozalbo D, Roig P, O'Connor JE, Ferrandiz ML, Fradelizi D, et al. Toll-like receptor 2 is dispensable for acquired host immune resistance to Candida albicans in a murine model of disseminated candidiasis. J Microbes. 2004;6(6):542-8. https://doi.org/10.1016/j.micinf.2004.02.015 PMid:15158187
  50. Netea MG, Van Der Graaf CA, Vonk AG, Verschueren I, Van Der Meer JW, Kullberg BJ. The role of toll-like receptor (TLR) 2 and TLR4 in the host defense against disseminated candidiasis. J Infect Dis. 2002;185(10):1483-9. https://doi.org/10.1086/340511 PMid:11992285

[TOP]