본문바로가기

Epidemiology, the Impact of COVID-19, and the Mycological Approach in Invasive Mold Infection

Abstract

Invasive fungal infections are a significant health challenge worldwide. Although invasive yeast infections are more prevalent, invasive mold infections (IMIs), such as Aspergillus spp., Mucorales, and Fusarium spp., are rising. The increase in IMIs is attributed to various factors, such as transplantation, aggressive treatments, invasive medical devices, and the emergence of new at-risk populations, such as patients with influenza and SARS-CoV-2-associated aspergillosis. The European Organization for Research and Treatment of Cancer and the Mycosis Study Group initially provided definitions based on host and clinical factors, including radiological findings and mycological evidence. The recent 2020 updates of the definitions state that a case confirmed by microscopy, histopathology, and culture from a sterile site is "proven"; however, regarding mycological criteria, this guideline introduced revised thresholds for the galactomannan assay based on sample types, excluded the 1,3-β-D-glucan detection assay, and acknowledged positive Aspergillus polymerase chain reaction tests from serum, plasma, or bronchoalveolar lavage fluid. The current diagnostic approach improves the accuracy and timeliness of diagnosing fungal infections and facilitates more effective patient management.



Keywords



Aspergillus PCR Conventional methods COVID-19 Galactomannan antigen Invasive mold infections



INTRODUCTION

Invasive fungal infections (IFIs) are systemic infections resulting from the establishment of yeasts or molds in deep-seated tissues, blood, and sterile sites. Nearly 70% of these infections worldwide are attributed to invasive candidiasis, followed by cryptococcosis (20%) and aspergillosis (10%)1. Endemic fungal strains, such as Blastomyces, Histoplasma, ParaCoccidioides, and Coccidioides, also contribute to severe systemic infections. IFIs affect approximately 1.9 million in- dividuals annually, and 3 million people globally suffer from chronic severe fungal infections1,2.

Although invasive mold infections (IMI) are relatively less common than invasive yeast infections, the incidence of mold-related diseases and hospitalizations is continuously increasing1,3. The disease burden of mold infections is in- creasing in Korea4,5. IMI can be caused by Aspergillus spp. and other septated hyaline molds such as Penicillium, Paecilomyces, Acremonium, Scopulariopsis, Fusarium, and Scedosporium, Mucorales, and dematiaceous fungi3,4. Mold infections are seldom controlled at the initial stage but can escalate in severity due to delayed diagnosis, complicating treatment and leading to higher mortality1,3. This increase in mold-related diseases and hospitalizations highlights the urgent need for timely and accurate fungal species detection and identification.

This article reviews the epidemiology and impact of COVID-19 on these infections and current nonculture-based diag- nostic methods, such as serological and molecular methods, with the updated consensus definitions for Invasive Fungal Diseases (IFD) by the Cancer/Mycoses Special Interest Group (EORTC/MSG)6.

EPIDEMIOLOGY AND THE IMPACT OF COVID-19

Invasive mold infection is exacerbated by aggressive medical treatments such as chemotherapy and transplants, invasive devices, such as central venous catheters, and the emergence of new at-risk populations, including individuals with in-fluenza and SARS-CoV-2-associated Aspergillosis7-9. Invasive aspergillosis (IA) and invasive mucormycosis (IM) are the most prevalent IMIs. The mortality rate for candidemia exceeds 25%, while IA results in death in 40% to 90% of immuno- compromised patients. The rate approaches 85% for un- treated IA and nearly 100% for untreated IM, depending on the patient's underlying conditions2,9.

Aspergillus fumigatus is the leading cause of culture-positive IA, followed by A. flavus, A. niger, and A. terreus. Infections from nonfumigatus Aspergillus species are more likely to become disseminated1. Mucormycosis is primarily caused by fungi from the order Mucorales, which frequently results in respiratory infections that can extend to the sinuses, eyes, or central nervous system. IM is commonly caused by Rhizopus spp., while other species, including Mucor spp., Lichtheimia spp., Apophysomyces spp., Cunninghamella spp., and Saksenaea spp., also cause IMI2,4.

Other hyaline molds, Fusarium spp., Scedosporium spp., Penicillium spp., and Acremonium spp., are also implicated in IMIs. Fusarium spp. typically disseminates in patients with a hematologic malignancy and hematopoietic stem cell trans- plant (HSCT) recipients but appears localized in solid organ transplant (SOT) recipients, leading to better outcomes than in neutropenic patients10. Scedosporium spp. can cause condi-tions ranging from mycetoma to lethal disseminated infections, especially in SOT recipients from nearly drowned donors11. Dermatiaceous fungi such as Alternaria spp., Exophiala spp., and Curvularia spp. can cause diseases ranging from skin lesions to disseminated infection in high-risk patients10. SOT recipients frequently exhibit skin conditions, while HSCT re- cipients are prone to lung complications, which are particularly problematic in these patients9,11. Indeed, Aspergillus spp., Fusarium spp., and Mucorales are known to cause healthcare-associated infections in chronic disease patients12.

COVID-19 has impacted the incidence of fungal infections. COVID-19-associated pulmonary aspergillosis (CAPA) was first identified in China in early 2020, affecting about 10% of invasively ventilated COVID-19 patients and about 20.1% of those with acute respiratory failure requiring ventilation. A. fumigatus is the predominant pathogen causing CAPA linked to azole-resistant Aspergillus strains8. The patients most commonly affected are those with acute respiratory failure due to COVID-19, particularly patients treated with systemic corticosteroids or tocilizumab13. Unlike influenza-associated pulmonary aspergillosis, CAPA is usually diagnosed about 8 days after ICU admission and has a mortality that exceeds 40%. During India's second COVID-19 wave in early 2021, over 47,500 COVID-19-associated mucormycosis (CAM) cases were reported. The prevalence of CAM among hospitalized COVID-19 patients was 0.27% in India and about 1~2% in Europe8,14. CAM commonly leads to rhino-orbital-cerebral and pulmonary diseases in COVID-19 patients with high mortality and extensive necrosis, often necessitating disfiguring surgical debridement. Major risk factors include uncontrolled diabetes and systemic corticosteroid overuse13.

DEFINITION OF INVASIVE MOLD INFECTION

Several criteria, such as those from EORTC/MSG, Bulpa, Blot, and the modified Aspergillosis in ICU (AspICU), have been recommended for the timely diagnosis and management of IFIs in various clinical settings6,7,15. Definitions by the EORTC/ MSG for immunocompromised individuals were first proposed in 2002 and later updated in 2008 and 2020, incorporating host factors, clinical criteria, and mycological criteria6,15,16. However, the AspICU criteria proposed for ICU patients do not include microbial biomarkers such as the galactomannan (GM) antigen and Aspergillus polymerase chain reaction (PCR)7.

According to the EORTC/MSG definition, a case confirmed by microscopy, histopathology, and culture from a sterile site is "proven". The diagnosis is "probable" if three criteria are met: the presence of at least 1 host factor, a clinical feature, and mycologic evidence. Conversely, a case that meets the appropriate host factors and clinical evidence but lacks mycological support is considered "possible"16. Due to increasing data on Aspergillus GM thresholds and the new PCR-based diagnostic tests, the 2020 update to the EORTC/MSG criteria includes modifications in mycological criteria and additional host factors, such as SOT and hematologic malignancies. This document underscores the importance of laboratory diagnosis in identifying IFD, highlighting the roles of GM testing and PCR assays for Aspergillus6 (Table 1). Adopting the 2020 EORTC/MSG criteria led to a significant 27.3% reduction in the diagnosis of probable IPA, contributing to a more uniform patient classification of treatment response, prognosis, and mortality. This consistency enhances the reliability of mortality predictions compared to the 2008 criteria17.

Criteria

Fungus/
disease

Mycologic feature

Conventional method

Serology*

PCR based assay

Proven invasive mold disease

Mold

Histopathologic, cytopathologic, or direct microscopy by needle aspiration or biopsy

Not applicable




Amplification of fungal DNA
by PCR combined with DNA sequencing when molds are
seen in formalin-fixed
paraffin-embedded tissue

Recovery by culture of a
sterile specimen (excluding BAL fluid, a paranasal or mastoid sinus cavity
specimen, and urine)

 

 

Probable invasive pulmonary mold disease

Mold

Aspergillus, Fusarium, Scedosporium species, or Mucorales recovered by culture from sputum, BAL, bronchial brush, or aspirate

 

 

Microscopical detection of fungal elements in sputum, BAL, bronchial brush, or aspirate indicating a mold

 

 

Tracheobronchitis

Aspergillus recovered by
culture of BAL or
bronchial brush

 

 

Microscopic detection of fungal elements in BAL or bronchial brush

 

 

Sinonasal diseases

Mold recovered by culture
of sinus aspirate samples

 

 

Microscopic detection of fungal elements in sinus aspirate samples

 

 

Aspergillus spp.,

only

Recovered by culture from sputum, BAL, bronchial
brush, or aspirate








Galactomannan Ag:
Any 1 of the following

 

Single serum or plasma:
1.0, BAL 1.0

Single CSF: 1.0

Single serum or plasma:
0.7 and BAL 0.8



Aspergillus PCR

: Any 1 of the following

 

PCR positive as

2 consecutive plasma,
serum, or whole blood

2 duplicated BAL fluid
PCR positive

1 PCR positive in plasma,
serum, or whole blood and
1 PCR test positive in BAL fluid

*1,3-β-D glucan was not considered to provide mycological evidence of any invasive mold disease

Abbreviations: BAL, bronchoalveolar lavage; CSF, cerebrospinal fluid; PCR, polymerase chain reaction

Table 1. Mycological approaches based on the updated 2020 European Organization for Research and Treatment of Cancer-Invasive Fungal Infections Cooperative Group/National Institute of Allergy and Infectious Diseases Mycosis Study Group (EORTC/MSG) guideline for defining proven, and probable invasive mold disease
MYCOLOGICAL APPROACH

1. The conventional method: microscopy, histopathology, and culture-based tests

Histopathologic demonstration of tissue invasion by hyphae or mold recovery from a culture of a specimen obtained from a clinically sterile site is a "proven" IMD16. This category can apply to any patient, regardless of their immunocompromised status. However, these conventional methods have inherent limitations, including a prolonged turnaround time, the inva- sive nature of the samples, and lower sensitivity. The propor- tion of proven cases was relatively low, from 1.8% to 27.0. Blood culture sensitivity for aspergillosis was markedly low at 1~5%, in contrast to 50~95% for candidemia1,19. Recovery of Aspergillus spp. from blood cultures rarely indicates endo- vascular disease and is almost always contamination6. A nationwide study in Korea found that only 8% of IPA cases were proven with these methods20.

2. Galactomannan (GM) antigen assay

GM is a polysaccharide cell wall component released by Aspergillus spp. during growth. It can be detected in serum, bronchoalveolar lavage (BAL) fluid, cerebrospinal fluid (CSF), and others15,21. The 2002 EORTC/MSG definition included Aspergillus antigen detection, such as GM, as a criterion for probable IA16. However, the Platelia Aspergillus EIA (Bio-Rad, Marnes-la-Coquette, France), an immune-enzymatic sandwich microplate assay using a rat antigalactofuranose monoclonal antibody (EB-A2) was first approved in 2003 and remains the only FDA-approved GM assay, despite the availability of multiple commercial assays. Initially, the cut-off for GM was between 1.0 and 1.5 to minimize false positives, but in 2008, this threshold was lowered to 0.5 to improve performance in adult hematology patients21,22. The 2020 EORTC/MSG definition raised the cut-off back to 1.0 for individual plasma, serum, BAL fluid, or CSF samples, setting specific criteria for simultaneous positive results in serum and BAL fluid: ≥0.7 for serum and ≥0.8 for BAL6.

The GM assay has high sensitivity and moderate accuracy in diagnosing IA in immunocompromised patients. In patients with hematologic malignancies, the GM assay has higher sensitivity and specificity in BAL fluid than serum15,21. Sensitivity is low in SOT recipients due to less frequent angioinvasion. False-positive results are usually reported in pediatric patients without clinical signs of IA. Galactofuranose in various fungi and certain substances has led to reports of false-positive results21. These false-positive results can arise from colonization by fungi, such as the genera Fusarium, Histo- plasma, Penicillium, Alternaria, Paecilomyces, or Geotrichum, the administration of piperacillin-tazobactam or β-lactams, patients receiving total parenteral nutrition, such as Plasma-Lyte (Baxter, Deerfield, IL, USA), NP2 Enfant AP-HP parenteral nutrition (Fresenius, Kabi, Sevres, France), and intravenous gluconate solution23,24.

Additionally, the cross-reactivity of BAL fluid samples with Mycoplasma pneumoniae or anesthetic drugs has been re- ported24,25. Patients with mucositis or compromised intestinal barriers have false-positive GM results due to dietary factors. When using BAL fluid for testing, lung transplant recipients have a significantly higher rate of false-positive results due to extensive airway colonization25,26. These inaccuracies lead to unnecessary diagnostic procedures, treatments, and radiation exposure from CT scans aimed at detecting IPA, adverse reactions from antifungal medications, and increased health- care costs due to unnecessary tests and treatments. False negatives or reduced detection has also been reported in patients with chronic granulomatous disease or hyperimmuno- globulin E (hyper-IgE) syndrome (formerly Job syndrome) despite their increased risk of IA27.

3. (1,3)-β-D-glucan (BDG) assay

(1,3)-β-D-glucan (BDG), a polysaccharide component of fungal cell walls, is a pan-fungal antigen detected in various species, including Candida spp., Pneumocystis jivoveci, Asper- gillus spp., Acremonium spp., and Fusarium spp. with ex- ceptions such as Cryptococcus spp., Mucorales, and the yeast phase of Blastomyces dematitidis22. The commercial BDG assay, the Fungitell Assay (Associates of Cape Cod Inc., East Falmouth, MA, USA) has received FDA approval and numerous other BDG assays are available, each with various cut-off values: Fungitell assay, cut-off value 60~80 pg/mL; the Wako β-glucan test, cut-off value 11 pg/mL (Wako Pure Chemical, Osaka, Japan); and the Fungitec-G, cut-off value 20 pg/mL (Seikagaku, Tokyo, Japan)28,29.

The revised 2020 EORTC/MSG criteria exclude BDG detec- tion as a microbiologic criterion for IFD since BDG detection is not specific to a single IMD6. False positives can occur in concurrent bacteremia such as Pseudomonas aeruginosa and Streptococcus species, certain β-lactam antibiotics, specific antitumor chemotherapy agents, various fractionated blood components, and hemodialysis membranes. However, BDG detection is considered suitable for diagnosing probable IFD in specific clinical situations, such as patients with hematologic malignancies, post-hematopoietic stem cell transplantation neutropenia, and selection of high-risk ICU patients with complications from gastrointestinal surgery suspected of having an infection29,30. Additionally, due to its relatively high NPV, BDG testing is considered useful for excluding IFD in patients28.

4. Polymerase chain reaction (PCR) assay

The PCR assay is recommended for Aspergillus spp. and Pneumocystis jirovecii infections, and serological testing will likely enhance the diagnosis. Molecular diagnostics represent the only nonclassical mycological approach for IM4,31. The International Fungal PCR Initiative (FPCRI) standardized DNA extraction protocols from serum, plasma, and whole blood to improve diagnostic accuracy32. BAL fluid is preferred for detecting respiratory pathogens, while debrided tissue is favored for skin and soft tissue infections. In formalin-fixed paraffin-embedded (FFPE) tissue, amplification of fungal DNA by PCR combined with DNA sequencing can be used when molds are seen. However, caution is needed with FFPE tissue due to a low DNA yield and potential amplification interference from colonizers31.

The 2020 updates to the EORTC/MSG definitions now include Aspergillus-specific plasma, serum, whole blood, and BAL fluid PCR assays as a mycological criterion for probable IA6. The European Aspergillus PCR Initiative (EAPCRI) Working Group of the International Society of Human and Animal Mycology has validated Aspergillus PCR assays in plasma, serum, and whole blood, except BAL fluid19,32. Aspergillus PCR assays have moderate accuracy for diagnosing IPA in immunocompromised patients. They require two positive results for confirmation to achieve higher specificity. In high-risk hematology patients, these assays in blood show a high NPV of 94% and a PPV of 70%, reducing the need for empir- ical antifungal therapy33. Aspergillus PCR assays in patients not previously treated with antifungals show a sensitivity of 71.4% and a specificity of 92.3%, with a PPV of 62.5% and an NPV of 98.3%; however, in patients with Aspergillus-active prophylaxis, the PPV drops to 5.4% while the NPV increases to 100%, indicating that PCR screening is more effective in patients not on primary antimold prophylaxis34.

PCR assays for Aspergillus DNA in specimens such as BAL fluid, CSF, and tissue are useful for diagnosing IA in SOT recipients or patients under long-term immunosuppressive therapy6. However, BAL fluid assays may not differentiate between colonization and active infection, with a PPV of about 72% in nonhematology patients14,35. While Candida PCR assays lack standardization, Aspergillus PCR assays are well-standardized, aiding in result comparison and detecting azole resistance mutations. Still, the absence of an FDA-endorsed Aspergillus PCR assay limits its global use31. Despite the lack of significant differences in mortality between the redefined possible and probable IA groups, elevated DNA levels (greater than 150 copies/mL) in quantitative PCR have been associated with higher 90-day mortality rates6,33,36. The false-positive results in Aspergillus PCR assays are often due to environmental contamination, cross-reactions with other fungi, such as Penicillium spp., or respiratory tract colon- ization37.

The absence of serological tests to confirm the diagnosis of IM highlights the need for a molecular assay. The increasing use of Mucorales-specific PCR assays enhances accuracy in detecting DNA in blood and BAL fluid, using dual loci (ITS and TEF1a) for DNA barcoding31,38. Mucorales DNA has been detected in blood 3 to 68 days before conventional methods, eight days earlier than histology or culture, and three days before imaging31. Quantitative PCR in serum samples has been proven to be of high prognostic value. Patients with decreasing fungal loads have better survival rates. However, more comprehensive studies are needed to clarify its sensitivity and role in patients treated with antifungals and those with rhino-orbital-cerebral mucormycosis31,39.

CONCLUSION

IMIs are exacerbated by medical interventions, such as chemotherapy and transplants, invasive devices, and the emergence of new at-risk populations, including those with influenza and SARS-CoV-2-associated aspergillosis. Diag- nostic mycological approaches have evolved beyond trad- itional microscopy and culture to include serology-based and molecular approaches, offering rapid, sensitive, and specific detection. This fact is particularly vital for timely and accurate diagnosis, allowing the initiation of appropriate treatment. The 2020 EORTC/MSG criteria have integrated these newer diagnostic tools, reflecting advancements in nonculture-based methods that improve diagnostic accuracy and patient man- agement in fungal infections. These molecular diagnostics are pivotal in differentiating invasive infections from colo- nization, especially in immunocompromised patients, and have significantly refined the approach to invasive mold infection detection and management.



References


1. Bongomin F, Gago S, Oladele RO, Denning DW. Global and multi-national prevalence of fungal diseases-estimate precision. J Fungi (Basel) 2017;3:57
Google Scholar 

2. Lass-Flörl C, Steixner S. The changing epidemiology of fungal infections. Mol Asp Med 2023;94:101215


3. Drgona L, Khachatryan A, Stephens J, Charbonneau C, Kantecki M, Haider S, et al. Clinical and economic burden of invasive fungal diseases in Europe: focus on pre-emptive and empirical treatment of Aspergillus and Candida species. Eur J Clin Microbiol Infect Dis 2014;33: 7-21
Google Scholar 

4. Lee SO. Diagnosis and Treatment of invasive mold dis- eases. Infect Chemother 2023;55:10-21
Google Scholar 

5. Huh K, Ha YE, Denning DW, Peck KR. Serious fungal infections in Korea. Eur J Clin Microbiol Infect Dis 2017; 36:957-963
Google Scholar 

6. Donnelly JP, Chen SC, Kauffman CA, Steinbach WJ, Baddley JW, Verweij PE, 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. Clin Infect Dis 2020;71:1367-1376
Google Scholar 

7. Schauwvlieghe A, Rijnders BJA, Philips N, Verwijs R, Vanderbeke L, Van Tienen C, et al. Invasive aspergillosis in patients admitted to the intensive care unit with severe influenza: a retrospective cohort study. Lancet Respir Med 2018;6:782-792
Google Scholar 

8. Patel A, Agarwal R, Rudramurthy SM, Shevkani M, Xess I, Sharma R, et al. Multicenter epidemiologic study of coronavirus disease-associated mucormycosis. India. Emerg Infect Dis 2021;27:2349-2359
Google Scholar 

9. Sung AH, Martin S, Phan B, Benigno M, Stephens J, Chambers R, et al. Patient characteristics and risk factors in invasive mold infections: comparison from a systematic review and database analysis. Clinicoecon Outcomes Res 2021;13:593-602
Google Scholar 

10. Naggie S, Perfect JR. Molds: hyalohyphomycosis, phaeo- hyphomycosis, and zygomycosis. Clin Chest Med 2009; 30:337-353
Google Scholar 

11. Ramirez-Garcia A, Pellon A, Rementeria A, Buldain I, Barreto-Bergter E, Rollin-Pinheiro R, et al. Scedosporium and Lomentospora: an updated overview of underrated opportunists. Med Mycol 2018;56:S102-S125
Google Scholar 

12. Lee R, Cho SY, Lee DG. Fundamentals of mycology for infection control and prevention. Korean J Healthc Assoc Infect Control Prev 2020;25:86-99
Google Scholar 

13. Bartoletti M, Pascale R, Cricca M, Rinaldi M, Maccaro A, Bussini L, et al. Epidemiology of invasive pulmonary asper- gillosis among intubated patients with COVID-19: a prospective study. Clin Infect Dis 2021;73:e3606-e3614
Google Scholar 

14. Hoenigl M, Seidel D, Sprute R, Cunha C, Oliverio M, Goldman GH, et al. COVID-19-associated fungal infec- tions. Nat Microbiol 2022;7:1127-1140
Google Scholar 

15. De Pauw 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) Consensus Group. Clin Infect Dis 2008;46:1813-1821
Google Scholar 

16. 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;34:7-14
Google Scholar 

17. Acet-Öztürk NA, Ömer-Topçu D, Vurat-Acar K, Aydın-Güçlü Ö, Pınar İE, Demirdöğen E, et al. Impact of revised EORTC/MSGERC 2020 criteria on diagnosis and pro- gnosis of invasive pulmonary aspergillosis in patients with hematological malignancies undergoing bronchoscopy. J Mycol Med 2022;32:101304


18. Marr KA, Schlamm HT, Herbrecht R, Rottinghaus ST, Bow EJ, Cornely OA, et al. Combination antifungal therapy for invasive aspergillosis: a randomized trial. Ann Intern Med 2015;162:81-89
Google Scholar 

19. Fang W, Wu J, Cheng M, Zhu X, Du M, Chen C, et al. Diagnosis of invasive fungal infections: challenges and recent developments. J Biomed Sci 2023;30:42
Google Scholar 

20. Kim SH, Moon SM, Han SH, Chung JW, Moon SY, Lee MS, et al. Epidemiology and clinical outcomes of invasive pulmonary aspergillosis: a nationwide multicenter study in Korea. Infect Chemother 2012;44:282-288
Google Scholar 

21. Pfeiffer CD, Fine JP, Safdar N. Diagnosis of invasive asper- gillosis using a galactomannan assay: a meta-analysis. Clin Infect Dis 2006;42:1417-1427
Google Scholar 

22. Sanglard D. Emerging threats in antifungal-resistant fungal pathogens. Front Med 2016;3:11
Google Scholar 

23. Sulahian A, Touratier S, Ribaud P. False positive test for Aspergillus antigenemia related to concomitant admin- istration of piperacillin and tazobactam. N Engl J Med 2003;349:2366-2367
Google Scholar 

24. Chong E, Ko JH, Kim DR, Lee YH, Yang J, Kim H, et al. False-positive Aspergillus galactomannan immunoassay in the glucose component of total parenteral nutrition products. Microbiol Spectr 2023;11:e01673-23
Google Scholar 

25. Shin DW, Cho J, Choi KS, Lee J, Choi Y, Choi SJ, et al. False-positive results of galactomannan assays in patients administered glucose-containing solutions. Sci Rep 2024; 14:2552
Google Scholar 

26. Clancy CJ, Jaber RA, Leather HL, Wingard JR, Staley B, Wheat LJ, et al. Bronchoalveolar lavage galactomannan in diagnosis of invasive pulmonary aspergillosis among solid-organ transplant recipients. J Clin Microbiol 2007; 45:1759-1765
Google Scholar 

27. Verweij PE, Poulain D, Obayashi T, Patterson TF, Denning DW, Ponton J. Current trends in the detection of anti- genaemia, metabolites and cell wall markers for the diagnosis and therapeutic monitoring of fungal infections. Med Mycol 1998;36:146-155
Google Scholar 

28. Lamoth F, Akan H, Andes D, Cruciani M, Marchetti O, Ostrosky-Zeichner L, et al. Assessment of the role of 1,3-β-D-glucan testing for the diagnosis of invasive fungal infections in adults. Clin Infect Dis 2021;72:S102-S108
Google Scholar 

29. Yoo IY, Park YJ. Culture-independent diagnostic appro- aches for invasive aspergillosis in solid organ transplant recipients. Korean J Transplant 2023;37:155-164
Google Scholar 

30. Theel ES, Doern CD. β-D-glucan testing is important for diagnosis of invasive fungal infections. J Clin Microbiol 2013;51:3478-3483
Google Scholar 

31. Gudisa R, Harchand R, Rudramurthy SM. Nucleic-acid-based molecular fungal diagnostics: a way to a better future. Diagnostics (Basel) 2024;14:520
Google Scholar 

32. White PL, Wingard JR, Bretagne S, Löffler J, Patterson TF, Slavin MA, et al. Aspergillus polymerase chain reaction: systematic review of evidence for clinical use in com- parison with antigen testing. Clin Infect Dis 2015;61: 1293-1303
Google Scholar 

33. Cruciani M, Mengoli C, Barnes R, Donnelly JP, Loeffler J, Jones BL, et al. Polymerase chain reaction blood tests for the diagnosis of invasive aspergillosis in immuno- compromised people. Cochrane Database Syst Rev 2019; 9:CD009551.
Google Scholar 

34. Springer J, Lackner M, Ensinger C, Risslegger B, Morton CO, Nachbaur D, et al. Clinical evaluation of a Mucorales-specific real-time PCR assay in tissue and serum samples. J Med Microbiol 2016;65:1414-1421
Google Scholar 

35. Arastehfar A, Carvalho A, van de Veerdonk FL, Jenks JD, Koehler P, Krause R, et al. COVID-19 associated pulmo- nary aspergillosis (CAPA)-from immunology to treatment. J Fungi 2020;6:91
Google Scholar 

36. Imbert S, Gauthier L, Joly I, Brossas JY, Uzunov M, Touafek F, et al. Aspergillus PCR in serum for the diagnosis, follow-up and prognosis of invasive aspergillosis in neutropenic and nonneutropenic patients. Clin Microbiol Infect 2016; 22:562.e1-e8
Google Scholar 

37. Barnes RA, White PL, Morton CO, Rogers TR, Cruciani M, Loeffler J, et al. Diagnosis of aspergillosis by PCR: Clinical considerations and technical tips. Med Mycol 2018;56: 60-72
Google Scholar 

38. Kidd SE, Chen SC, Meyer W, Halliday CL. A new age in molecular diagnostics for invasive fungal disease: Are we ready? Front Microbiol 2019;10:2903
Google Scholar 

39. White PL, Alanio A, Brown L, Cruciani M, Hagen F, Gorton R, et al. An overview of using fungal DNA for the diag- nosis of invasive mycoses. Expert Rev Mol Diagn 2022; 22:169-184
Google Scholar 

Congratulatory MessageClick here!

Download this article