Next Article in Journal
Gaining an Understanding of Pneumocystosis in Wales
Next Article in Special Issue
Blood Serum Stimulates the Virulence Potential of Mucorales through Enhancement in Mitochondrial Oxidative Metabolism and Rhizoferrin Production
Previous Article in Journal
Clonal Diversity of Candida auris, Candida blankii, and Kodamaea ohmeri Isolated from Septicemia and Otomycosis in Bangladesh as Determined by Multilocus Sequence Typing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epidemiology, Modern Diagnostics, and the Management of Mucorales Infections

by
David Pham
1,†,
Annaleise R. Howard-Jones
2,3,4,†,
Rebecca Sparks
5,†,
Maurizio Stefani
2,
Varsha Sivalingam
2,
Catriona L. Halliday
2,
Justin Beardsley
1,3,4,6 and
Sharon C.-A. Chen
2,3,4,*
1
Centre for Infectious Diseases & Microbiology, Westmead Hospital, Westmead, NSW 2170, Australia
2
Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology—Institute of Clinical Pathology & Medical Research, Westmead Hospital, Westmead, NSW 2170, Australia
3
Faculty of Medicine & Health, University of Sydney, Camperdown, NSW 2006, Australia
4
Sydney Institute for Infectious Diseases, University of Sydney, Sydney, NSW 2006, Australia
5
Douglass Hanly Moir Pathology, Sydney, NSW 2113, Australia
6
Westmead Institute for Medical Research, Sydney, NSW 2145, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Fungi 2023, 9(6), 659; https://doi.org/10.3390/jof9060659
Submission received: 1 May 2023 / Revised: 8 June 2023 / Accepted: 10 June 2023 / Published: 12 June 2023
(This article belongs to the Special Issue Recent Advances in Mucorales and Mucormycosis, 2nd Edition)

Abstract

:
Mucormycosis is an uncommon, yet deadly invasive fungal infection caused by the Mucorales moulds. These pathogens are a WHO-assigned high-priority pathogen group, as mucormycosis incidence is increasing, and there is unacceptably high mortality with current antifungal therapies. Current diagnostic methods have inadequate sensitivity and specificity and may have issues with accessibility or turnaround time. Patients with diabetes mellitus and immune compromise are predisposed to infection with these environmental fungi, but COVID-19 has established itself as a new risk factor. Mucorales also cause healthcare-associated outbreaks, and clusters associated with natural disasters have also been identified. Robust epidemiological surveillance into burden of disease, at-risk populations, and emerging pathogens is required. Emerging serological and molecular techniques may offer a faster route to diagnosis, while newly developed antifungal agents show promise in preliminary studies. Equitable access to these emerging diagnostic techniques and antifungal therapies will be key in identifying and treating mucormycosis, as delayed initiation of therapy is associated with higher mortality.

1. Introduction

Mucormycosis is an uncommon but aggressive invasive disease caused by fungi belonging to the order Mucorales, with the most common causative genera being Rhizopus, Mucor, and Lichtheimia [1,2,3]. Mucorales are ubiquitous environmental moulds and rarely cause invasive disease in immunocompetent hosts; however, patients with mucormycosis typically have predisposing conditions, such as diabetes mellitus, malignancy, haematopoietic or solid-organ transplantation, and, most recently, concurrent or recent COVID-19 infection [4,5,6]. The patient population at risk for mucormycosis continues to expand due to increasing metabolic disease and the use of immunosuppressive therapies, leading to an increasing incidence of mucormycosis worldwide. Mucorales has also been well-recognised to cause outbreaks within healthcare settings or following natural disasters [7].
Infection is typically acquired through the inhalation of spores into the sinuses or lung of the susceptible host, with subsequent spread to contiguous structures or dissemination via angioinvasion, depending on the host immune response. The most common presentations are rhino-orbital-cerebral mucormycosis (ROCM), typically associated with diabetes mellitus, and pulmonary mucormycosis, which is the most common manifestation in haematology patients; gastrointestinal (GI), cutaneous and soft tissue, and disseminated mucormycosis also occur [1,2]. The diagnosis of mucormycosis is reliant on the identification of fungal angioinvasion on histopathology, as well as culture and culture-independent microbiological methods, with imaging used to support the diagnosis [3,8]. Timely diagnosis is essential for maximising survival through early medical and surgical interventions [3,9]. Even with early recognition and management, the prognosis remains poor, with mortality estimated at 60% for ROCM and 57% for pulmonary mucormycosis in cases published worldwide [10,11].
Mucorales were recently designated as high-priority pathogens by the WHO, receiving this ranking on account of challenges with drug susceptibility and diagnosis [12]. Within this narrative review, we outline the emerging literature in the epidemiology of Mucorales infections, with a focus on newer risks and outbreaks. We also provide an update on the taxonomy of these fungi to provide clarity on causative pathogens; discuss diagnostic methods, including whole-genome sequencing (WGS) approaches; and review key management strategies, including new antifungal agents.

2. Taxonomy of Mucorales

One of the most ancient groups of fungi, the order Mucorales, is characterised by abundant thin-walled aseptate mycelium. Taxonomy was traditionally based upon microscopic morphology and mating experiments, with these organisms classified in the phylum Zygomycota. However, molecular phylogenetic studies of multiple gene regions, including the nuclear ribosomal RNA subunits (18S, 5.8S and 28S), RNA polymerase subunits (RPB1 and RPB2), elongation factor 1-alpha, and α- and β-tubulins, indicated that Zygomycota was polyphyletic [13]. The phylum Zygomycota was abandoned following further phylogenetic analysis in favour of two phyla: Mucoromycota and Zoopagomycota. The Mucoromycota consist of three subphyla: Glomeromycotina, Mortierellomycotina, and Mucoromycotina, of which the latter include the orders Mucorales, Umbelopsidales, and Endogenales [14,15].
Within Mucorales, the structure may be classified at the family level, using four molecular markers: (i) large and (ii) small subunit of the ribosomal DNA, (iii) the partial actin gene, and (iv) the partial gene for elongation factor 1-alpha [16]. Mucorales fungi reproduce asexually by means of single-celled sporangiospores produced endogenously in either sporangia (multi-spored), sporangiola (with 1 or few spores), or merosporangia (spores in rows). Sexual reproduction occurs via the fusion of two opposed suspensors originating from different hyphae to form a zygospore [17]. Additional morphological characteristics are now not considered to be taxonomically relevant [15].
The family structure including the 11 genera (and at least 39 species) of Mucorales described as human pathogens is summarised in Table 1 [15,16]. Rhizopus and Mucor, followed by Lichtheimia and Apophysomyces, are the main pathogenic genera; and Rhizopus arrhizus, Rhizopus microsporus, and Mucor circinelloides the main species [15,18]. Historically, M. circinelloides was divided into four formae, namely f. circinelloides, f. griseocyanus, f. janssenii, and f. lusitanicus, but these now represent the species M. circinelloides sensu stricto, M. griseocyanus, M. janssenii, and M. lusitanicus, respectively; and the additional species M. ramosissimus, M. variicolumellatus, and M. velutinosus make up the M. circinelloides complex known to cause human infections [18]. There is ongoing debate regarding the species concept of R. arrhizus (syn R. oryzae); in this review, two varieties are referred to—R. arrhizus var. arrhizus and R. arrhizus var. delemar [15,19].

3. Epidemiology of Mucormycosis

3.1. Update on Epidemiology: Burden and Causative Pathogens

The WHO reports that the burden of disease caused by Mucorales is currently unknown due to variable reporting strategies and incomplete surveillance data, as mucormycosis is not a reportable disease in most jurisdictions [12]. Other than two systematic reviews of mucormycosis by Roden et al. (929 cases from 1940 to 2003 [1] and by Jeong et al. (851 cases from 2000 to 2017) [2], most data are presented as case series or reports, which typically focus on national or regional epidemiology or are confined to in specific populations [5,21,22,23,24,25,26,27,28,29,30,31,32]. Systematic reviews have also been undertaken for specific clinical manifestations, such as cutaneous, GI, and pulmonary mucormycosis [11,33,34,35].
Despite imperfect epidemiological data, there appeared to be an increasing incidence of mucormycosis worldwide, even prior to the COVID-19 pandemic [1,23,36]. In part, this is attributed to improved recognition and diagnosis, as well as increasing prevalence of risk factors such as diabetes mellitus and immunosuppression [37]. A French nationwide population study revealed an increase from 0.7 to 1.2 cases per million population per year between 1997 and 2006 [36], while the incidence increased 2.5-fold between 2008 and 2018 in an Iranian case series [23].
Globally, the highest reported incidence of mucormycosis comes from India, around 70× higher than elsewhere [38,39]. This has been attributed, in part, to high rates of poorly controlled diabetes mellitus. The overall incidence increased from an estimated 25 cases per year (from 1990–2007) to 89 cases per year (2013–2015) in one tertiary hospital [24], while a series of cross-sectional studies from Chakrabarti et al. showed an increase from 12.9 cases per year (1990–1999) to 50 cases per year at another tertiary hospital (2006–2007) [40,41,42]. A computational model estimated a mean of 171,504 cases (95% CI 147,688–195,777) and 65,500 deaths (38.2% mortality) per year in India prior to COVID-19. Finally, the estimated incidence was 14 cases per 100,000 population per year in India and Pakistan, compared to 0.05–9.5 cases per 100,000 population per year globally [39,43].
Rhizopus (48%), Mucor (14%), and Lichtheimia (13%) spp. are responsible for the majority of mucormycosis cases worldwide, but there is significant regional variation in causative pathogens [2]. These variations are likely attributable to endemicity in the environment, as well as pathophysiological factors such as temperature-dependent growth parameters. For instance, Apophysomyces cases comprise the second most reported Mucorales infections in India, possibly due to the thermotolerant nature of the fungus and its ubiquitous presence the local environment [44,45]. Conversely, Apophysomyces cases have been less commonly described in Africa and Europe, while Lichtheimia is responsible for a greater proportion of European cases [2].
Certain Mucorales have associations with specific clinical manifestations. For instance, Cunninghamella spp. are more common in patients with pulmonary disease (57% versus 20%, p < 0.001) or disseminated disease (33% versus 13%, p = 0.002), and mortality associated with Cunninghamella infections is significantly higher than that caused by other Mucorales (71% versus 44%, p < 0.001) [2]. Apophysomyces spp. (65%), Lichtheimia spp. (45%), and Saksenaea complex (75%) were more commonly isolated in patients with cutaneous disease [2].
Several emerging species are notable. Rhizopus homothallicus was first noted as the cause of two cases of cavitary pulmonary mucormycosis in 2010 [46]. Subsequent case series from India have also reported R. homothallicus causing cutaneous mucormycosis, ROCM [24,47,48,49,50], and CAM [51,52]. Mucor irregularis (previously Rhizomucor variabilis), first described as causing cutaneous mucormycosis in China [53], has since been reported in China and India to be associated with chronic cutaneous mucormycosis in immunocompetent patients [50,54,55]. Mucor velutinosus, first described in 2011, has been implicated in cutaneous and bloodstream disease [56,57,58,59]. Saksenaea erythrospora, first described as causing orbital mucormycosis in 2011 [60], is known to cause cutaneous or orbital disease following trauma or surgery, with descriptions from Asia and the Americas [61,62]. Other emerging Mucorales include Thamnostylum lucknowense [63], Cunninghamella arunalokei [20] and Lichtheimia ornata [64] (all ROCM), and Apophysomyces mexicanus (cutaneous mucormycosis) [65].

3.2. Host Risk Factors

The traditional host risk factors for mucormycosis are well-established and have been detailed in systematic reviews by Roden et al. and Jeong et al. [1,2], as well as other publications within Table 2. These factors both predispose to Mucorales infection and influence the clinical presentation and prognosis of disease [66].

COVID-19-Associated Mucormycosis

Fungal superinfections are well-recognised in patients with severe COVID-19 or those recovering from the disease. In early 2021, increasing cases of CAM were reported from India, with an estimated 2.1-fold rise in mucormycosis during September–December 2020, compared to September–December 2019 [76]. Factors contributing to this include high background prevalence of mucormycosis in India, high incidence of undiagnosed or poorly controlled diabetes mellitus, COVID-19 immune dysregulation, and the immunosuppressive effect of COVID-19 therapies such as steroids [77,78]. Although at comparatively lower rates of incidence, CAM has been reported globally [79,80].
Several systematic reviews have found that CAM occurs predominantly in males (70–79%), patients with underlying diabetes (78–83%), and/or those receiving corticosteroids (76–79%) [6,77,79,81]. Rhizopus spp. were the most implicated pathogens [6]. Most presentations were with ROCM (73–89%), and mortality varied between 30 and 49% [82]. The pathophysiology is thought to involve the interplay of hyperglycaemia, diabetic ketoacidosis, immunosuppression, free iron availability, immune dysregulation, and pulmonary damage from COVID-19 predisposing to tissue invasion [83]. The detailed pathogenesis of CAM is beyond the scope of this review, but the reader is referred to several comprehensive reviews on this topic [84,85,86].
Major challenges with CAM have included difficulties with diagnosis and with access to effective therapies. The imaging changes of mucormycosis may overlap with pneumonitis due to SARS-CoV-2 or be reminiscent of other invasive fungal superinfection with mass lesions, cavitations, and nodules, thus preventing early recognition (Figure 1A) [87]. Access to invasive sampling of respiratory specimens was also reduced due to COVID-19 infection prevention precautions. Additionally, the increased burden of COVID-19 also reduced healthcare resources that may have otherwise been applied to recognise, diagnosis, and treat CAM more effectively [88]. Notably, in India, supplies of effective antifungal agents were rapidly exhausted during their outbreak, exposing weaknesses in supply chains [89]. Moving forward, a high degree of awareness is required to swiftly diagnose and manage patients with CAM.

3.3. Healthcare-Associated Mucormycosis

Although Mucorales cause mostly sporadic infections in the community setting, nosocomial outbreaks are increasingly recognised [7,90,91]. Outbreaks can present with the full range of clinical presentations, including ROCM and disseminated disease. Predisposing factors for disease are similar to sporadic infections (Table 2) [90]. Exposure to mucormycetes can occur throughout the entire healthcare setting, with infections linked to construction [92,93,94]; insufficient ventilation [95,96,97,98]; and contaminated medical devices [99,100,101,102,103,104,105,106], linen [107,108,109,110,111,112], and food [113,114,115,116].
Clusters have been described in several hospital settings, including in intensive care units (ICUs) [117], transplantation units as graft-transmitted mucormycosis in liver- and kidney-transplant recipients [118,119,120], and notably in burn units [34,121,122]. Burn patients have extensive disruptions of the physical skin barrier, along with immune dysfunction. A systematic review of 114 patients across 46 studies showed that the most common pathogen was Mucor spp. (44%) [34]. Cutaneous infection was the most common clinical syndrome (84%), while only 6% of cases were ROCM. Both an increasing percentage of total body surface area burned and infection in two or more locations were associated with higher mortality [34]. Two outbreaks within burn units were due to the contamination of non-sterile bandages [123,124].
Recognition and investigation of healthcare-associated outbreaks can be difficult [125,126], and despite best efforts, epidemiological links can be difficult to establish [127]. Investigations should involve robust hypothesis-testing, the detection of potential healthcare sources, and consideration of environmental testing [125,126]. Molecular methods, including WGS, have emerged to characterise the genomic epidemiology of these outbreaks (see Section 4.8) [128,129,130,131]. “Pseudo-outbreaks” of Mucorales have also been described, and it is important to differentiate true noscomial clusters from environmental contamination or issues resulting from changes in surveillance or testing. For instance, three pseudo-outbreaks of Rhizopus spp. infection originated from contaminated non-sterile wooden sticks or tongue depressors [132,133,134]. In each outbreak, at least one patient was treated with amphotericin for presumptive mucormycosis prior to recognition of the contamination. A third outbreak of M. circinelloides sensu stricto was initially implicated over two Parisian hospitals, with WGS eventually identifying a high degree of genetic diversity amongst the isolates without a point source [124,129].

3.4. Natural Disaster-Associated Mucormycosis

Natural disasters predispose environments to outbreaks of mucormycosis by providing a traumatic portal of entry, whilst disturbed earth or flood waters may carry pathogens, and certain weather patterns have even been hypothesised to induce hypervirulent states [135]. Damage to the health infrastructure during these times results in inadequate sanitation and hygiene and limited access to healthcare services [136]. In the first-described disaster-associated outbreak, eight cases of R. arrhizus infection were reported following a volcanic eruption near Armero, Colombia, in 1985 [137]. These mostly involved severe necrotising fasciitis complicating trauma and carried a mortality of 75%. Notably, R. arrhizus was also isolated from the environmental culture of the disaster mud flow. Three cases of cutaneous mucormycosis from severe trauma (one due to Mucor spp. and two to Apophysomyces elegans) were also reported following the Indian Ocean Tsunami in 2004 [138,139,140].
A cluster of 13 cases of cutaneous mucormycosis in immunocompetent hosts was identified following an EF-5 tornado disaster in Joplin, Missouri, USA, in 2011 [141]. Apophysomyces trapeziformis was isolated from the wounds and other body sites of the patients, five of whom died (38.5%). It has been postulated that the high relative incidence of mucormycosis may be related to induction of hypervirulent phenotypes by the tornadic force [142]. Wurster et al. demonstrated that mechanical stress induced by magnetic stirring induced a hypervirulent phenotype for Mucorales within a Drosophila infection model [142].
Four ROCM infections due to R. arrhizus were reported after widespread flooding in Colorado, USA, in 2013 [143]. Each patient had risk factors for mucormycosis, including haematological malignancy and poorly controlled diabetes. Two patients died despite appropriate surgical and medical care. In another case series of invasive mould infections following floodwater damage from Hurricane Harvey in Houston, Texas, USA, in 2017, 5/20 cases were due to Mucorales infection [144]; however, there was not a selective predominance of causative genera, with the incidence of Mucorales, Aspergillus, and Fusarium invasive infections all increasing. Overall, while data are limited to case series, collectively, there is a plausible link between natural disasters and mucormycosis outbreaks, likely due to environmental disturbance, traumatic injury, suboptimal hygiene, and health infrastructure disruption.

4. Diagnostics

The diagnosis of mucormycosis is ever challenging due to the variety of disease manifestations that often mimic other fungal infections, most notably invasive aspergillosis (IA). Diagnostic approaches must incorporate a thorough assessment, with a consistent clinical syndrome, alongside integration of radiological, histopathological, and microbiological findings (Figure 1).

4.1. Radiology

Radiographic findings in mucormycosis vary depending on the site of disease, and imaging often serves to establish clinical suspicion and to guide biopsy for definitive diagnosis [145]. For instance, a reverse halo sign or multiple nodules on chest computed tomographic (CT) imaging are characteristic signs of pulmonary mucormycosis [145]. Cranial CT or magnetic resonance imaging (MRI) findings in ROCM can be non-specific, as the most common finding of sinusitis can also represent disease caused by bacteria or other fungi, and follow-up endoscopic biopsy is strongly recommended [3]. CT or MRI angiography can assist in the assessment of angioinvasion and diagnosis of mycotic aneurysms [146,147,148]. There is also an emerging role of 18-Fluorodeoxyglucose Positron Emission Tomography (FDG PET) in diagnosis, staging of dissemination, and follow-up of mucormycosis [149,150,151].

4.2. Microscopy and Histology

Direct microscopic examination of clinical specimens, using a fluorescing dye such as Calcofluor white, can provide a rapid presumptive diagnosis of mucormycosis [3,152]. Suggestive findings of direct microscopy, even in the absence of a positive culture, should be considered significant [152]. The visualisation of characteristic mucormycete hyphae in tissue biopsy is essential to a definitive diagnosis [8]. Such features include findings of aseptate or pauciseptate ribbon-like hyphae (width 6–25 μm) with irregular branching, visualised by haematoxylin and eosin (H&E), periodic acid-Schiff (PAS), or Grocott–Gomori methenamine silver (GMS) staining. Importantly, histopathology alone cannot always distinguish between mucormycosis and other fungal infections [153]. While Mucorales are historically described as having right-angle branching as opposed to the acute-angle branching angles of septate moulds such as Aspergillus, this morphological feature can be masked by pressure effects on the fungus and the distorting effects induced by tissue processing [3].
Immunohistochemical (IHC) targets, such as α-1,6-mannan, have been investigated with limited in vitro success, as cross-reactivity with Aspergillus, Fusarium and Candida spp. limits their specificity for mucormycosis [154]. Monoclonal antibodies against specific Mucorales demonstrate high sensitivity and specificity, making them useful to differentiate mucormycosis against other invasive fungal infections, although they are not widely available [155,156,157].

4.3. Culture-Based Diagnostics

The culture of appropriate specimens from patients with suspected mucormycosis is essential to yield an isolate for accurate speciation and antimicrobial susceptibility testing [3]. However, sensitivity of culture in microscopically positive samples is only up to 50% [158]. It is critical that tissue samples are handled without homogenisation or grinding, which disrupts the hyphae and culture viability [159]. Morphological identification of Mucorales is detailed elsewhere [152,160]. Whilst morphological examination is key, other identification methods, such as proteomics or sequencing, are increasingly used.
The case-series data on matrix-assisted laser desorption/ionization–time of flight (MALDI-TOF) mass spectrometry for Mucorales identification are promising [161,162], although molecular approaches remain the definitive method for species identification. The internal transcribed spacer regions (ITS1 and ITS2) of rDNA or the 18S or 28S rRNA genes themselves are the most frequently used targets, with the ITS region being the preferred DNA barcoding marker for fungal speciation [163,164]. To determine species identification, sequences are typically compared against the Westerdijk Fungal Biodiversity Institute (http://www.wi.knaw.nl/, accessed on 1 May 2023) and the International Society for Human and Animal Mycology (ISHAM) (http://its.mycologylab.org/, accessed on 1 May 2023) databases. However, as for many other fungal pathogens, a single target has not been identified that performs consistently well for speciation of all Mucorales [159].

4.4. Molecular Diagnostics

Molecular-based assays are pivotal in mucormycosis diagnosis, allowing for expedited results from clinical specimens and/or detection of Mucorales DNA in culture-negative specimens, as well as playing an emerging role in molecular biomarker monitoring. Polymerase chain reaction (PCR) diagnostics from tissue and bronchoalveolar lavage (BAL) fluid are well established, and fresh tissues in which fungal elements have been visualised carry higher diagnostic utility [165,166].
The spore-coating protein homolog encoding CotH genes has emerged as a promising PCR target, as the genes are uniquely and universally present among Mucorales [167,168]. Recent studies on a single-target CotH PCR assay demonstrated a sensitivity of 90% and a specificity of 100% in a mouse model of mucormycosis, with no CotH amplification seen in BAL fluid, plasma, and urine samples from uninfected and Aspergillus-infected mice [169]. Interestingly, CotH positivity was observed in urine, plasma, and BAL fluid from 24 h after infection, suggesting that this target may be useful for early diagnosis [169].
The diagnostic yield of PCR on serum or whole blood is limited; hence, the reported efficacy of Mucorales-specific PCR assays on these specimens varies widely. Assays incorporating multiple targets to encompass Mucor, Rhizopus, Rhizomucor, and Lichtheimia species have been used to diagnose early infection and to prognosticate based on treatment response [170]. In one study including 232 patients with suspected invasive mould disease, the PCR on whole blood was helpful in detecting mucormycosis a median of four days prior to culture positivity, and PCR negativity after amphotericin initiation was associated with an 85% reduction in 30-day mortality (adjusted hazard ratio, 0.14; 95% CI, 0.03–0.72; p = 0.02) [171]. An interlaboratory evaluation of this assay demonstrated high reproducibility and performance [172].
The commercial assay, MucorGenius® (PathoNostics, Maastricht, The Netherlands), a semi-quantitative PCR assay targeting the 28S rRNA target, has shown modest sensitivity (75%) based on retrospective testing of 106 stored residual blood samples from 16 patients with culture-confirmed mucormycosis [173]. However, a positive MucorGenius® PCR result preceded culture positivity by a median of eight days, again underlining the potential for the use of this assay in enabling early diagnosis [173]. A retrospective French study investigating pulmonary mucormycosis also demonstrated high sensitivity and specificity for both the MucorGenius® and an in-house PCR [174]. By contrast, a Mucorales-specific semi-nested PCR on serum was not effective in the diagnosis of confirmed Mucorales-associated sinusitis in immunocompromised patients where none of the five patients was identified [175].
Isothermal amplification methods are not in routine use for the diagnosis of mucormycosis. That being said, one report using 42 reference strains of Mucorales suggested the utility of this approach as a sensitive and specific diagnostic tool for rapid diagnosis of R. microsporus, R. arrhizus var. arrhizus, R. arrhizus var. delemar, M. irregularis, M. circinelloides, L. ramosa, and L. corymbifera infections [176]. A high-resolution melting analysis (HRMA) of PCR products from BAL fluid also showed promise in differentiating some Mucorales species with a sensitivity of 100% and specificity of 93% [177].

4.5. Serological Diagnosis

Antigen-based diagnostic assays in mainstream use for invasive fungal infections—namely the galactomannan and beta-d-glucan assays—are not useful in the diagnosis of mucormycosis due to the absence of these antigens in the cell walls of Mucorales [178]. The possibility of dual infections must be considered, particularly in immunocompromised patients, so a positive galactomannan or beta-d-glucan result does not necessarily exclude concurrent mucormycosis. Early studies of novel antigen targets have shown promise, such as the use of the Rhizopus-specific antigen for diagnosis of R. arrhizus infection in murine models [179].
Several serological assays have been evaluated for the diagnosis of mucormycosis, using ELISA-based techniques, Western immunoblots, and immunodiffusion assays. ELISA-based assays have displayed sensitivities of 81% and specificities of 97% but demonstrated non-specific cross-reactivity with Candida and Aspergillus spp., using serum samples [180]. None of these approaches has accumulated sufficient clinical data to support clinical implementation [155,179,181]. Lateral flow immunoassays (LFIAs), unlike for IA [182], are not yet of utility in the diagnosis of mucormycosis; however, initial reports of an LFIA targeting fucomannan, a Mucorales cell-wall carbohydrate, have shown promising results on serum, urine, and BAL fluid in a murine model [183].
One novel approach under investigation for the diagnosis of mucormycosis is an enzyme-linked immunospot (ELISpot) or immunocytofluorimetric assay, which detects Mucorales-specific T-cell activity via quantitation of interferon-gamma release from activated T cells [184,185,186]. The preliminary observational data are promising, but clinical data are limited to very small case series.

4.6. Emerging Diagnostic Approaches

Due to the clinical severity of invasive mucormycosis and association of delayed diagnosis with compromised clinical outcomes [187], emerging diagnostic techniques are constantly under evaluation to improve the timeliness and accuracy of diagnosis. Research is ongoing into the utility of metabolomic signatures such as breath tests for volatile metabolites [188], biosensor-based approaches in clinical specimens [189], fluorescence in situ hybridisation techniques for diagnosis from formalin-fixed paraffin-embedded tissues [190], and mass spectrometry of serum disaccharides [191]. To date, these methodologies remain under active exploration within the research sphere only.

4.7. Susceptibility Testing

The European Committee on Antimicrobial Susceptibility Testing (EUCAST) and the Clinical and Laboratory Standards Institute (CLSI) have developed standardised methodologies for antifungal susceptibility testing of Mucorales [192,193]. While the methods differ, there is good agreement, although EUCAST minimum inhibitory concentrations (MICs) tend to be 1- to 3-fold dilutions higher than those of CLSI [194,195]. Spectrophotometric readings show reasonable agreement with visual inspection of microdilution plates [196,197,198]. Epidemiological cutoff values (ECVs or ECOFFs) have been determined for some species, but clinical breakpoints (CBPs) have not been defined. Due to species-specific variations in antifungal susceptibility, in vitro antifungal susceptibility testing is recommended to establish epidemiologic knowledge to aid in the establishment of ECVs/ECOFFs and CBPs (Table 3), as well as to ultimately optimise treatment [199].
In vitro MICs show that amphotericin B is the most active antifungal against most Mucorales [201,202,203,204,205]. In one small series of 10 mucormycosis cases, there was a significantly better 6-week response when the pathogen MIC was ≤0.5 mg/L versus >0.5 mg/L for amphotericin B (83% versus 0%; p = 0.05) [206]. The ≥95% ECV for amphotericin B against L. corymbifera, M. circinelloides, R. arrhizus, and R. microsporus was calculated to be 1, 1, 2, and 2 mg/L, respectively (Table 3) [200]. Mucorales, which exhibit higher MICs for amphotericin B, include Apophysomyces spp. and Cunninghamella bertholletiae [200,202,203,204,207].
Posaconazole and isavuconazole have good in vitro activity against many Mucorales. The 95% ECV of posaconazole for L. corymbifera, R. arrhizus, and R. microsporus is 1 mg/L, while the ECV for M. circinelloides is 4 mg/L (Table 3) [200]. While posaconazole has overall lower MICs than isavuconazole, its blood concentrations are lower, and the clinical significance of these differences are unclear [195,204,208]. Rhizopus spp. tend to have higher posaconazole MICs, while M. circinelloides tends to have higher isavuconazole MICs [195,203,205,209]. Itraconazole activity is variable and more species specific (Table 3). Other triazoles, including voriconazole, and echinocandins display high MICs against Mucorales, and are considered intrinsically resistant [202,203,210,211]. While terbinafine is active in vitro against some Mucorales (geometric mean MIC < 1 mg/L for L. corymbifera, Cunninghamella spp., R. microsporus, and R. pusillus), its efficacy is poor in animal models [199,203].
There have been a small number of studies looking at the synergistic activity of antifungal combinations using chequerboard microdilution methods [212]. Certain drug combinations have synergistic activity against some strains and species, but this is not readily predictable [213,214,215,216]. Dannaoui et al. studied 35 Mucorales isolates and found combination amphotericin B/rifampicin to be synergistic or additive, combination amphotericin B/terbinafine to be synergistic for 20% of isolates, and combination terbinafine/voriconazole to be synergistic for 44% of isolates [213]. On the other hand, there was neither synergism nor antagonism with combination amphotericin B/posaconazole amongst 11 R. arrhizus isolates [215]. Schwarz et al. reviewed combination antifungal therapy for mucormycosis, outlining existing studies for combination susceptibility testing for both antifungal and non-antifungal drugs and found that while antagonism is rare, synergy rates were highly variable [212].
A limited number of studies have compared the reference broth microdilution method against alternative methods such as disc diffusion or gradient strip testing. Disc diffusion appears feasible, with a low rate of major and very major errors demonstrated for posaconazole and amphotericin B when using cutoffs of 1 and 4 mg/L to pragmatically define sensitivity and resistance, respectively [207]. There have been conflicting studies with gradient strip testing, with some studies showing reasonable agreement of MICs and others demonstrating conflicting results, especially for amphotericin B and posaconazole [197,217,218,219].
Molecular determinants of antifungal resistance are not well studied or described. Research into R. arrhizus has demonstrated that CYP51A is also responsible for its intrinsic voriconazole and fluconazole resistance [220,221]. Modelling studies suggest that the differential activity of the azoles against Rhizopus and Mucor spp. may be explained by a key F129 residue of the CYP51 F5 gene in Mucorales spp. which confers decreased binding of short-side-chain (fluconazole, voriconazole) but not long-side-chain azoles (posaconazole) [222].

4.8. Whole-Genome Sequencing

Earlier investigations of mucormycosis case clusters have detailed epidemiological linkages without data on the genetic relatedness between strains, partly as there are no established genotyping makers for Mucorales [105,132]. The advent of WGS approaches for phylogenetic analyses has yielded new information, as well as challenges in delineating genetic diversity within different species [128,129,130,131,141,223]. Firstly, a WGS analysis of Mucorales is problematic due to the large size of their genomes (45.3 Mbp for R. arrhizus), as well as the presence of whole-genome duplication events [221]. Most assembled genomes of R. microsporus var. rhizopodiformis strains were ≅25 Mbp, but some isolates had larger genomes (43–51 Mbp), which are indicative of genome expansion [130]. The genomes of R. pusillis (≅25 Mbp), Apophysomyces (≅32 Mbp), and Lichtheimia spp. (≅58 Mbp) are larger still [128,224]. Genome size influences pre-analytic and analytic factors such as capacity, throughput, selection of sequencing coverage, and read depths.
Secondly, reference genomes for Mucorales are less rigorously standardized in comparison with reference bacterial genomes. Nonetheless, there are publicly available reference genomes that have been assembled by research groups, such as M. circinelloides 1006 PhL [114], M. indicus B7402 [225,226], R. microsporus (GenBank: GCA_006680115.1) [130], and R. pusillus (GenBank: GCA_900175165.2) [128]. Variability of comparative sequence results may be evident with the use of different reference genomes. Thirdly, deriving the genetic threshold or pairwise distance cutoff to determine if two or more strains within a species are clonal can be problematic. This threshold will vary with species, recombination events, and the bioinformatics pipeline used. In addition, the genome variability of Mucorales within the hospital environment and in the community is uncertain in comparison to baseline genomic epidemiology, which is fundamental to contextualising the relatedness of strains [125].
Garcia-Hermoso et al. were amongst the first to use WGS analysis to investigate a possible nosocomial cluster of six patients with Mucor circinelloides sensu stricto wound infection [129]. Twenty-one isolates (fourteen outbreak and seven unrelated isolates) were studied against the reference genome M. circinelloides 1006PhL. This study set the premise that the genome of M. circinelloides was stable, established parameters of the sequencing process, and assigned a genetic threshold (by estimating evolutionary distance and number of single nucleotide polymorphisms (SNPs) between pairs of isolates) to determine the genetic proximity between genomes from identical or clonal strains. Unexpectedly, approximately 5296 SNPs were identified as a cutoff below which two isolates could be designated as being of the same strain. Four clades were identified, where each clade was separated by >290,000 SNPs, while isolates within each clade varied by ≅20,000 SNPs [223,227]. Contrary to the initial notion that single-strain clonal transmission caused the outbreak, the cluster was due to multiple unrelated strains present in the environment. These conclusions are similar to those of the Joplin outbreak investigation [141,224] where there was no linkage between genetic groups of A. trapeziformis and exposure [224].
In a WGS-directed investigation of three cases of mucormycosis in heart and lung transplant patients over 6 months, genome assembly of two R. pusillus strains yielded > 5900 core SNPs between them; the third isolate was Lichtheimia ramosa [128]. This report reiterated the need to interpret core SNPs between isolates with caution in the absence of defined cutoffs for clonal vs. non-clonal strains. For R. microsporus var. rhizopodiformis, however, 21 of 24 isolates associated with a cluster of surgical infections were of a single clade, with 17 of the 21 forming an inner clade consisting of only 1235 SNPs (range 60–912, mean 430). Although this is indicative of a likely common geographic origin, the genomic diversity was reported to be inconsistent with a point source outbreak [130].
Similarly, Nguyen et al. investigated a cluster of four transplant-associated cases of mucormycosis (three Rhizopus spp. and one Lichtheimia sp.) over a 10-month period and performed WGS on 72 clinical and environmental Mucorales including the outbreak isolates [131]. Both core and pan-genome analyses were performed. Notably, the core genome analysis had insufficient resolution to identify unique isolates and falsely clustered them. The pangenome analysis indicated extensive genetic diversity among clinical and environmental isolates.
Thus, while WGS approaches can resolve genetic diversity between strains, there remain challenges in demonstrating definitive associations with clinical and environmental strains, or to pinpoint a source of infection. Defining thresholds of SNP variation between isolates to determine relatedness remains challenging.

5. Management and New Antifungal Agents

Optimal management of Mucorales infection remains challenging, as there is a scarcity of randomised clinical trials to direct best practice treatment, and management guidelines are based on open-label studies, small clinical case series, and expert opinion [3]. The European Confederation for Medical Mycology (ECMM), in conjunction with the American Society for Microbiology (ASM) and International Society for Human and Animal Mycology (ISHAM), recommends a multimodal approach including (i) aggressive surgical debridement or resection of the affected region(s), (ii) antifungal therapy, and (iii) strict management of predisposing factors such as uncontrolled diabetes mellitus and neutropenia [3]. More recently, analogous recommendations have been made for the treatment of CAM [153,228]. The current treatment recommendations are summarised in Figure 2.
The preferred agent for empiric antifungal therapy, as well as targeted treatment in both adult and paediatric patients, is high-dose amphotericin B, with improved treatment success achieved with lipid amphotericin B formulations (either liposomal or lipid complex), while the deoxycholate formulation is recommended only in cases where other endorsed amphotericin B formulations are unavailable. Isavuconazole and posaconazole (delayed release tablets and intravenous formulations only) are both reasonable first-line alternatives when amphotericin is contraindicated, or they can alternatively be used as salvage therapy. Both are preferred over posaconazole oral suspension, as the latter is associated with higher rates of treatment failure [3,229]. When posaconazole is used, routine therapeutic drug monitoring (TDM) is strongly recommended, aiming for a trough level of >1 mg/L, while there is no current evidence to suggest that TDM is required for isavuconazole [3].
Combination antifungal therapy has been studied. For treatment refractory cases, the evidence suggests improved outcomes with combined amphotericin and echinocandin, or amphotericin and posaconazole; however, other retrospective evaluations report treatment equivalence, particularly in first line therapy [212]. Thus, this strategy is usually reserved for salvage therapy and in severely immunocompromised individuals [230]. Hyperbaric oxygen exposure in individuals with diabetes has been recommended as adjunctive therapy, with success described in several case reports and series in this population [230].
The treatment arsenal for Mucorales infection remains limited, and, even with current best-practice therapy, mortality rates over 60% are frequently described. Furthermore, current antifungal agents have limitations in terms of substantive toxicity and/or variable in vitro activity [231]. Thus, alternative therapeutic options are needed, and they are the subject of investigation. New antifungal agents, some with novel modes of action, are under varying stages of development and clinical evaluation for treatment of Mucorales infection. Those that have gained more recent attention include oteseconazole, manogepix (and its prodrug fosmanogepix), and jawsamycin.
Oteseconazole (VT-1161; Viamet Pharmaceuticals Inc., Durham, NC, UK), a long side-chain azole, is a novel metalloenzyme inhibitor which prevents ergosterol synthesis through selective inhibition of fungal CYP51 (lanosterol 14α-demethylase) [232]. Early studies noted potency in vitro against R. arrhizus var. arrhizus; however, the potency against R. arrhizus var. delemar was poor. In vivo R. arrhizus var. arrhizus infection studies in immunocompromised mice demonstrated a ≅1 log decrease in lung and brain fungal burdens in treated mice compared to placebo controls [233,234]. Whilst clinical trials of oteseconazole are in progress for other fungal infections, such as vulvovaginal candidiasis, further data on its efficacy in humans for either prophylaxis or treatment of Mucorales infections are needed.
Fosmanogepix (prodrug of manogepix) prevents the maturation of glycerolphosphatidyl inositol (GPI)-anchored proteins by inhibiting the enzymatic action of inositol acyltransferases (Gwt1), compromising fungal growth. In a murine model, combined fosmanogepix with liposomal amphotericin B was superior to amphotericin B alone in the treatment of invasive mucormycosis [235]. Potent in vitro profiles were exhibited in both R. arrhzius var arrhizus and R. arrhizus var. delemar isolates, and efficacy has been demonstrated when used as a single agent to treat R. arrhizus pulmonary and disseminated infection in immunosuppressed mice [236]. Clinical-outcome data in humans are currently absent; a phase II clinical trial (NTC04240886) was terminated prematurely due to enrolment issues. A phase III study has been planned.
Jawsamycin (FR-900848) inhibits the Spt14/Gpi3-mediated biosynthesis of glycosylphosphatidylinositol, which is essential to maintain the integrity of the fungal cell wall. Low MICs were recorded when ATCC isolates of six common species of Mucorales were exposed to jawsamycin in vitro, including R. arrhizus, L. corymbifera, and M. circinelloides. Additionally, enhanced survival and a decreased fungal burden were demonstrated in murine models with pulmonary infection due to R. arrzhius var. delemar [237].
Calcineurin and mTOR inhibitors are more novel treatment options which have both demonstrated synergistic in vitro activity against Mucorales isolates when combined with posaconazole, isavuconazole, or amphotericin B [238,239,240]. In vivo efficacy has been shown with rapamycin alone (in an insect model) and tacrolimus in combination with posaconazole (in insect and murine models) [241,242].
There are several other novel antifungal agents in early stages of evaluation against Mucorales, with some promising in vitro and in vivo activity, particularly for Rhizopus spp. These agents are detailed in Table 4. At the time of writing, there are no proposed clinical trials or clinical evaluations in humans.

6. Conclusions

In this paper, we described significant advances in diagnostic methods and promising signals for new antifungal therapy options, but significant challenges still exist in regard to controlling the burden of disease caused by this WHO high-priority pathogen group. First, an accurate estimate of the burden is required, along with a better understanding of the epidemiology of disease—especially as at-risk populations continue to emerge and expand. This requires systematic surveillance across countries, using common data points and ensuring that laboratory data are married to clinical meta-data.
Research and development efforts must focus on developing accurate, cheap, rapid diagnostic tests—ideally available in a point-of-care format. Advanced molecular epidemiology is complicated and challenging but holds significant promise; however, robust pathology stewardship will be needed.
Research must continue to focus on the development of new antifungal agents and adjunctive therapies. Mortality is unacceptably high with the current armamentarium, and newer agents, while promising, still require further investigation before being introduced to routine clinical care. Finally, there must be equitable access to available treatments and diagnostics. Improved supply chains, fair pricing, and capacity building will contribute to this goal.

Author Contributions

Conceptualization, S.C.-A.C.; investigation, D.P., A.R.H.-J., R.S., M.S., V.S., C.L.H. and S.C.-A.C.; resources, S.C.-A.C.; writing—original draft preparation, D.P., A.R.H.-J., R.S., M.S., V.S., C.L.H. and S.C.-A.C.; writing—review and editing, D.P., A.R.H.-J., R.S., C.L.H., J.B. and S.C.-A.C.; visualization, D.P., R.S., V.S. and C.L.H.; supervision, C.L.H., J.B. and S.C.-A.C.; project administration, S.C.-A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMB:Amphotericin B
ATCC:American Type Culture Collection
BAL:Bronchoalveolar lavage
c-AmB:Amphotericin B deoxycholate
CAM:COVID-19-associated mucormycosis
CBP:Clinical breakpoint
CI:Confidence interval
CLSI:Clinical and Laboratory Standards Institute
CT:Computed tomography
COVID-19:Coronavirus disease 2019
DNA:Deoxyribonucleic acid
ECV/ECOFF:Epidemiological cutoff value
EUCAST:European Committee on Antimicrobial Susceptibility Testing
FDG-PET:Fluorodeoxyglucose positron emission tomography
FFPE:Formalin fixed paraffin-embedded
GI:Gastrointestinal
GMS:Grocott–Gomori methenamine silver
H&E:Haematoxylin and eosin
HRMA:High-resolution melting analysis
IA:Invasive aspergillosis
ICU:Intensive care unit
IHC:Immunohistochemistry
ITS:Internal transcribed spacer
L-AmB:Liposomal Amphotericin B
LFIA:Lateral flow immunoassay
MALDI TOFMatrix assisted laser desorption ionisation-time of flight
MIC:Minimum inhibitory concentration
MRI:Magnetic Resonance Imaging
PAS:Periodic acid-Schiff
PCR:Polymerase chain reaction
POS:Posaconazole
RNA:Ribonucleic acid
ROCM:Rhino-orbital-cerebral mucormycosis
SNP:Single nucleotide polymorphism
TDM:Therapeutic drug monitoring
ITR:Itraconazole
WGS:Whole genome sequencing

References

  1. Roden, M.M.; Zaoutis, T.E.; Buchanan, W.L.; Knudsen, T.A.; Sarkisova, T.A.; Schaufele, R.L.; Sein, M.; Sein, T.; Chiou, C.C.; Chu, J.H.; et al. Epidemiology and Outcome of Zygomycosis: A Review of 929 Reported Cases. Clin. Infect. Dis. 2005, 41, 634–653. [Google Scholar] [CrossRef] [Green Version]
  2. Jeong, W.; Keighley, C.; Wolfe, R.; Lee, W.L.; Slavin, M.A.; Kong, D.C.M.; Chen, S.C.-A. The Epidemiology and Clinical Manifestations of Mucormycosis: A Systematic Review and Meta-Analysis of Case Reports. Clin. Microbiol. Infect. 2019, 25, 26–34. [Google Scholar] [CrossRef] [Green Version]
  3. Cornely, O.A.; Alastruey-Izquierdo, A.; Arenz, D.; Chen, S.C.A.; Dannaoui, E.; Hochhegger, B.; Hoenigl, M.; Jensen, H.E.; Lagrou, K.; Lewis, R.E.; et al. Global Guideline for the Diagnosis and Management of Mucormycosis: An Initiative of the European Confederation of Medical Mycology in Cooperation with the Mycoses Study Group Education and Research Consortium. Lancet Infect. Dis. 2019, 19, e405–e421. [Google Scholar] [CrossRef] [PubMed]
  4. Cuenca-Estrella, M.; Bernal-Martinez, L.; Isla, G.; Gomez-Lopez, A.; Alcazar-Fuoli, L.; Buitrago, M.J. Incidence of Zygomycosis in Transplant Recipients. Clin. Microbiol. Infect. 2009, 15, 37–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Corzo-León, D.E.; Chora-Hernández, L.D.; Rodríguez-Zulueta, A.P.; Walsh, T.J. Diabetes Mellitus as the Major Risk Factor for Mucormycosis in Mexico: Epidemiology, Diagnosis, and Outcomes of Reported Cases. Med. Mycol. 2018, 56, 29–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hoenigl, M.; Seidel, D.; Carvalho, A.; Rudramurthy, S.M.; Arastehfar, A.; Gangneux, J.-P.; Nasir, N.; Bonifaz, A.; Araiza, J.; Klimko, N.; et al. The Emergence of COVID-19 Associated Mucormycosis: A Review of Cases from 18 Countries. Lancet Microbe 2022, 3, e543–e552. [Google Scholar] [CrossRef]
  7. Walther, G.; Wagner, L.; Kurzai, O. Outbreaks of Mucorales and the Species Involved. Mycopathologia 2019, 185, 765–781. [Google Scholar] [CrossRef]
  8. Donnelly, J.P.; Chen, S.C.; Kauffman, C.A.; Steinbach, W.J.; Baddley, J.W.; Verweij, P.E.; Clancy, C.J.; Wingard, J.R.; Lockhart, S.R.; Groll, A.H.; 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] [CrossRef] [Green Version]
  9. Jeong, W.; Keighley, C.; Wolfe, R.; Lee, W.L.; Slavin, M.A.; Chen, S.C.-A.; Kong, D.C.M. Contemporary Management and Clinical Outcomes of Mucormycosis: A Systematic Review and Meta-Analysis of Case Reports. Int. J. Antimicrob. Agents 2019, 53, 589–597. [Google Scholar] [CrossRef]
  10. Vaughan, C.; Bartolo, A.; Vallabh, N.; Leong, S.C. A Meta-Analysis of Survival Factors in Rhino-Orbital-Cerebral Mucormycosis-Has Anything Changed in the Past 20 Years? Clin. Otolaryngol. 2018, 43, 1454–1464. [Google Scholar] [CrossRef]
  11. Muthu, V.; Agarwal, R.; Dhooria, S.; Sehgal, I.S.; Prasad, K.T.; Aggarwal, A.N.; Chakrabarti, A. Has the Mortality from Pulmonary Mucormycosis Changed over Time? A Systematic Review and Meta-Analysis. Clin. Microbiol. Infect. 2021, 27, 538–549. [Google Scholar] [CrossRef] [PubMed]
  12. World Health Organization. WHO Fungal Priority Pathogens List to Guide Research, Development and Public Health Action; World Health Organization: Geneva, Switzerland, 2022. [Google Scholar]
  13. Hibbett, D.S.; Binder, M.; Bischoff, J.F.; Blackwell, M.; Cannon, P.F.; Eriksson, O.E.; Huhndorf, S.; James, T.; Kirk, P.M.; Lücking, R.; et al. A Higher-Level Phylogenetic Classification of the Fungi. Mycol. Res. 2007, 111, 509–547. [Google Scholar] [CrossRef] [PubMed]
  14. Spatafora, J.W.; Chang, Y.; Benny, G.L.; Lazarus, K.; Smith, M.E.; Berbee, M.L.; Bonito, G.; Corradi, N.; Grigoriev, I.; Gryganskyi, A.; et al. A Phylum-Level Phylogenetic Classification of Zygomycete Fungi Based on Genome-Scale Data. Mycologia 2016, 108, 1028–1046. [Google Scholar] [CrossRef] [Green Version]
  15. Walther, G.; Wagner, L.; Kurzai, O. Updates on the Taxonomy of Mucorales with an Emphasis on Clinically Important Taxa. J. Fungi 2019, 5, 106. [Google Scholar] [CrossRef] [Green Version]
  16. Hoffmann, K.; Pawłowska, J.; Walther, G.; Wrzosek, M.; de Hoog, G.S.; Benny, G.L.; Kirk, P.M.; Voigt, K. The Family Structure of the Mucorales: A Synoptic Revision Based on Comprehensive Multigene-Genealogies. Persoonia-Mol. Phylogeny Evol. Fungi 2013, 30, 57–76. [Google Scholar] [CrossRef] [Green Version]
  17. Kwon-Chung, K.J. Taxonomy of Fungi Causing Mucormycosis and Entomophthoramycosis (Zygomycosis) and Nomenclature of the Disease: Molecular Mycologic Perspectives. Clin. Infect. Dis. 2012, 54, S8–S15. [Google Scholar] [CrossRef] [Green Version]
  18. Wagner, L.; Stielow, J.B.; de Hoog, G.S.; Bensch, K.; Schwartze, V.U.; Voigt, K.; Alastruey-Izquierdo, A.; Kurzai, O.; Walther, G. A New Species Concept for the Clinically Relevant Mucor circinelloides Complex. Persoonia-Mol. Phylogeny Evol. Fungi 2020, 44, 67–97. [Google Scholar] [CrossRef] [PubMed]
  19. Dolatabadi, S.; de Hoog, G.S.; Meis, J.F.; Walther, G. Species Boundaries and Nomenclature of Rhizopus arrhizus (syn. R. oryzae). Mycoses 2014, 57, 108–127. [Google Scholar] [CrossRef]
  20. Hallur, V.; Prakash, H.; Sable, M.; Preetam, C.; Purushotham, P.; Senapati, R.; Shankarnarayan, S.A.; Bag, N.D.; Rudramurthy, S.M. Cunninghamella arunalokei a New Species of Cunninghamella from India Causing Disease in an Immunocompetent Individual. J. Fungi 2021, 7, 670. [Google Scholar] [CrossRef]
  21. Kennedy, K.J.; Daveson, K.; Slavin, M.A.; van Hal, S.J.; Sorrell, T.C.; Lee, A.; Marriott, D.J.; Chapman, B.; Halliday, C.L.; Hajkowicz, K.; et al. Mucormycosis in Australia: Contemporary Epidemiology and Outcomes. Clin. Microbiol. Infect. 2016, 22, 775–781. [Google Scholar] [CrossRef] [Green Version]
  22. Stemler, J.; Hamed, K.; Salmanton-García, J.; Rezaei-Matehkolaei, A.; Gräfe, S.K.; Sal, E.; Zarrouk, M.; Seidel, D.; Abdelaziz Khedr, R.; Ben-Ami, R.; et al. Mucormycosis in the Middle East and North Africa: Analysis of the FungiScope® Registry and Cases from the Literature. Mycoses 2020, 63, 1060–1068. [Google Scholar] [CrossRef] [PubMed]
  23. Dolatabadi, S.; Ahmadi, B.; Rezaei-Matehkolaei, A.; Zarrinfar, H.; Skiada, A.; Mirhendi, H.; Nashibi, R.; Niknejad, F.; Nazeri, M.; Rafiei, A.; et al. Mucormycosis in Iran: A Six-Year Retrospective Experience. J. Mycol. Med. 2018, 28, 269–273. [Google Scholar] [CrossRef] [PubMed]
  24. Prakash, H.; Ghosh, A.K.; Rudramurthy, S.M.; Singh, P.; Xess, I.; Savio, J.; Pamidimukkala, U.; Jillwin, J.; Varma, S.; Das, A.; et al. A Prospective Multicenter Study on Mucormycosis in India: Epidemiology, Diagnosis, and Treatment. Med. Mycol. 2019, 57, 395–402. [Google Scholar] [CrossRef]
  25. Nucci, M.; Engelhardt, M.; Hamed, K. Mucormycosis in South America: A Review of 143 Reported Cases. Mycoses 2019, 62, 730–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kontoyiannis, D.P.; Yang, H.; Song, J.; Kelkar, S.S.; Yang, X.; Azie, N.; Harrington, R.; Fan, A.; Lee, E.; Spalding, J.R. Prevalence, Clinical and Economic Burden of Mucormycosis-Related Hospitalizations in the United States: A Retrospective Study. BMC Infect. Dis. 2016, 16, 730. [Google Scholar] [CrossRef] [Green Version]
  27. Skiada, A.; Pagano, L.; Groll, A.; Zimmerli, S.; Dupont, B.; Lagrou, K.; Lass-Florl, C.; Bouza, E.; Klimko, N.; Gaustad, P.; et al. Zygomycosis in Europe: Analysis of 230 Cases Accrued by the Registry of the European Confederation of Medical Mycology (ECMM) Working Group on Zygomycosis between 2005 and 2007. Clin. Microbiol. Infect. 2011, 17, 1859–1867. [Google Scholar] [CrossRef] [Green Version]
  28. Kontoyiannis, D.P.; Azie, N.; Franks, B.; Horn, D.L. Prospective Antifungal Therapy (PATH) Alliance®: Focus on Mucormycosis. Mycoses 2014, 57, 240–246. [Google Scholar] [CrossRef]
  29. Patel, A.; Kaur, H.; Xess, I.; Michael, J.S.; Savio, J.; Rudramurthy, S.; Singh, R.; Shastri, P.; Umabala, P.; Sardana, R.; et al. A Multicentre Observational Study on the Epidemiology, Risk Factors, Management and Outcomes of Mucormycosis in India. Clin. Microbiol. Infect. 2020, 26, 944.e9–944.e15. [Google Scholar] [CrossRef]
  30. Lanternier, F.; Dannaoui, E.; Morizot, G.; Elie, C.; Garcia-Hermoso, D.; Huerre, M.; Bitar, D.; Dromer, F.; Lortholary, O. A Global Analysis of Mucormycosis in France: The RetroZygo Study (2005–2007). Clin. Infect. Dis. 2012, 54, S35–S43. [Google Scholar] [CrossRef] [Green Version]
  31. Wei, L.-W.; Zhu, P.-Q.; Chen, X.-Q.; Yu, J. Mucormycosis in Mainland China: A Systematic Review of Case Reports. Mycopathologia 2022, 187, 1–14. [Google Scholar] [CrossRef]
  32. Bretagne, S.; Sitbon, K.; Desnos-Ollivier, M.; Garcia-Hermoso, D.; Letscher-Bru, V.; Cassaing, S.; Millon, L.; Morio, F.; Gangneux, J.-P.; Hasseine, L.; et al. Active Surveillance Program to Increase Awareness on Invasive Fungal Diseases: The French RESSIF Network (2012 to 2018). mBio 2022, 13, e00920-22. [Google Scholar] [CrossRef] [PubMed]
  33. Skiada, A.; Drogari-Apiranthitou, M.; Pavleas, I.; Daikou, E.; Petrikkos, G. Global Cutaneous Mucormycosis: A Systematic Review. J. Fungi 2022, 8, 194. [Google Scholar] [CrossRef]
  34. Dang, J.; Goel, P.; Choi, K.J.; Massenzio, E.; Landau, M.J.; Pham, C.H.; Huang, S.; Yenikomshian, H.A.; Spellberg, B.; Gillenwater, T.J. Mucormycosis Following Burn Injuries: A Systematic Review. Burns 2023, 49, 15–25. [Google Scholar] [CrossRef] [PubMed]
  35. Didehdar, M.; Chegini, Z.; Moradabadi, A.; Anoushirvani, A.A.; Tabaeian, S.P.; Yousefimashouf, M.; Shariati, A. Gastrointestinal Mucormycosis: A Periodic Systematic Review of Case Reports from 2015 to 2021. Microb. Pathog. 2022, 163, 105388. [Google Scholar] [CrossRef]
  36. Bitar, D.; Van Cauteren, D.; Lanternier, F.; Dannaoui, E.; Che, D.; Dromer, F.; Desenclos, J.-C.; Lortholary, O. Increasing Incidence of Zygomycosis (Mucormycosis), France, 1997–2006. Emerg. Infect. Dis. 2009, 15, 1395–1401. [Google Scholar] [CrossRef]
  37. Skiada, A.; Pavleas, I.; Drogari-Apiranthitou, M. Epidemiology and Diagnosis of Mucormycosis: An Update. J. Fungi 2020, 6, 265. [Google Scholar] [CrossRef]
  38. Prakash, H.; Chakrabarti, A. Epidemiology of Mucormycosis in India. Microorganisms 2021, 9, 523. [Google Scholar] [CrossRef]
  39. Prakash, H.; Chakrabarti, A. Global Epidemiology of Mucormycosis. J. Fungi 2019, 5, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Chakrabarti, A.; Chatterjee, S.S.; Das, A.; Panda, N.; Shivaprakash, M.R.; Kaur, A.; Varma, S.C.; Singhi, S.; Bhansali, A.; Sakhuja, V. Invasive Zygomycosis in India: Experience in a Tertiary Care Hospital. Postgrad. Med. J. 2009, 85, 573–581. [Google Scholar] [CrossRef]
  41. Chakrabarti, A.; Das, A.; Mandal, J.; Shivaprakash, M.R.; George, V.K.; Tarai, B.; Rao, P.; Panda, N.; Verma, S.C.; Sakhuja, V. The Rising Trend of Invasive Zygomycosis in Patients with Uncontrolled Diabetes Mellitus. Med. Mycol. 2006, 44, 335–342. [Google Scholar] [CrossRef] [Green Version]
  42. Chakrabarti, A.; Das, A.; Sharma, A.; Panda, N.; Das, S.; Gupta, K.L.; Sakhuja, V. Ten Years’ Experience in Zygomycosis at a Tertiary Care Centre in India. J. Infect. 2001, 42, 261–266. [Google Scholar] [CrossRef]
  43. Jabeen, K.; Farooqi, J.; Mirza, S.; Denning, D.; Zafar, A. Serious Fungal Infections in Pakistan. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 949–956. [Google Scholar] [CrossRef] [PubMed]
  44. Samaddar, A.; Shrimali, T.; Sharma, A. Mucormycosis Caused by Apophysomyces Species: An Experience from a Tertiary Care Hospital in Western India and Systematic Review of Global Cases. Mycoses 2023, 66, 181–195. [Google Scholar] [CrossRef] [PubMed]
  45. Prakash, H.; Ghosh, A.K.; Rudramurthy, S.M.; Paul, R.A.; Gupta, S.; Negi, V.; Chakrabarti, A. The Environmental Source of Emerging Apophysomyces variabilis Infection in India. Med. Mycol. 2016, 54, 567–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Chakrabarti, A.; Marak, R.S.K.; Shivaprakash, M.R.; Gupta, S.; Garg, R.; Sakhuja, V.; Singhal, S.; Baghela, A.; Dixit, A.; Garg, M.K.; et al. Cavitary Pulmonary Zygomycosis Caused by Rhizopus homothallicus. J. Clin. Microbiol. 2010, 48, 1965–1969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kokkayil, P.; Pandey, M.; Agarwal, R.; Kale, P.; Singh, G.; Xess, I. Rhizopus homothallicus Causing Invasive Infections: Series of Three Cases from a Single Centre in North India. Mycopathologia 2017, 182, 921–926. [Google Scholar] [CrossRef]
  48. Compain, F.; Aït-Ammar, N.; Botterel, F.; Gibault, L.; Le Pimpec Barthes, F.; Dannaoui, E. Fatal Pulmonary Mucormycosis Due to Rhizopus homothallicus. Mycopathologia 2017, 182, 907–913. [Google Scholar] [CrossRef]
  49. Taneja, J.; Agrawal, V.K.; Jamal, D.; Abbas, Z. A Rare Case of Pulmonary Mucormycosis Caused By Rhizopus homothallicus in Post Heart Transplant Patient. J. Intensive Crit. Care 2020, 6, 23. [Google Scholar]
  50. Chander, J.; Kaur, M.; Singla, N.; Punia, R.; Singhal, S.; Attri, A.; Alastruey-Izquierdo, A.; Stchigel, A.; Cano-Lira, J.; Guarro, J. Mucormycosis: Battle with the Deadly Enemy over a Five-Year Period in India. J. Fungi 2018, 4, 46. [Google Scholar] [CrossRef] [Green Version]
  51. Kaur, H.; Kanaujia, R.; Rudramurthy, S.M. Rhizopus homothallicus: An Emerging Pathogen in Era of COVID-19 Associated Mucormycosis. Indian. J. Med. Microbiol. 2021, 39, 473–474. [Google Scholar] [CrossRef]
  52. Gupta, M.K.; Selvaraj, S.; Tilak, R.; Kumar, N.; Kumar, R.; Chakravarty, J. Rhizopus homothallicus Rhino-orbital-cerebral Mucormycosis: Six Cases from a Tertiary Care Centre, North India. Trop. Med. Int. Health 2023, 28, 144–150. [Google Scholar] [CrossRef] [PubMed]
  53. Lu, X.; Liu, Z.; Shen, Y.; She, X.; Lu, G.; Zhan, P.; Fu, M.; Zhang, X.; Ge, Y.; Liu, W. Primary Cutaneous Zygomycosis Caused by Rhizomucor variabilis: A New Endemic Zygomycosis? A Case Report and Review of 6 Cases Reported from China. Clin. Infect. Dis. 2009, 49, e39–e43. [Google Scholar] [CrossRef] [Green Version]
  54. Hemashettar, B.M.; Patil, R.N.; O’Donnell, K.; Chaturvedi, V.; Ren, P.; Padhye, A.A. Chronic Rhinofacial Mucormycosis Caused by Mucor irregularis (Rhizomucor variabilis) in India. J. Clin. Microbiol. 2011, 49, 2372–2375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Tang, X.; Guo, P.; Wong, H.; Xie, J.; Han, J.; Xu, Y.; Zhou, H. Vacuum-Assisted Closure and Skin Grafting Combined with Amphotericin B for Successful Treatment of an Immunocompromised Patient with Cutaneous Mucormycosis Caused by Mucor irregularis: A Case Report and Literature Review. Mycopathologia 2021, 186, 449–459. [Google Scholar] [CrossRef] [PubMed]
  56. Joichi, Y.; Chijimatsu, I.; Yarita, K.; Kamei, K.; Miki, M.; Onodera, M.; Harada, M.; Yokozaki, M.; Kobayashi, M.; Ohge, H. Detection of Mucor velutinosus in a Blood Culture after Autologous Peripheral Blood Stem Cell Transplantation: A Pediatric Case Report. Med. Mycol. J. 2014, 55, E43–E48. [Google Scholar] [CrossRef] [Green Version]
  57. Sugui, J.A.; Christensen, J.A.; Bennett, J.E.; Zelazny, A.M.; Kwon-Chung, K.J. Hematogenously Disseminated Skin Disease Caused by Mucor velutinosus in a Patient with Acute Myeloid Leukemia. J. Clin. Microbiol. 2011, 49, 2728–2732. [Google Scholar] [CrossRef] [Green Version]
  58. Kumar, M.; Jantausch, B.; Torres, C.; Campos, J.; Zelazny, A. Central Line–Associated Mucor velutinosus Bloodstream Infection in an Immunocompetent Pediatric Patient. J. Pediatr. Infect. Dis. Soc. 2018, 7, e55–e57. [Google Scholar] [CrossRef] [Green Version]
  59. Álvarez, E.; Cano, J.; Stchigel, A.M.; Sutton, D.A.; Fothergill, A.W.; Salas, V.; Rinaldi, M.G.; Guarro, J. Two New Species of Mucor from Clinical Samples. Med. Mycol. 2011, 49, 62–72. [Google Scholar] [CrossRef] [Green Version]
  60. Hospenthal, D.R.; Chung, K.K.; Lairet, K.; Thompson, E.H.; Guarro, J.; Renz, E.M.; Sutton, D.A. Saksenaea erythrospora Infection Following Combat Trauma. J. Clin. Microbiol. 2011, 49, 3707–3709. [Google Scholar] [CrossRef] [Green Version]
  61. Troiano, G.; Nante, N. Emerging Fungal Infections: Focus on Saksenaea erythrospora. J. Prev. Med. Hyg. 2021, 62, E382–E385. [Google Scholar] [CrossRef]
  62. Chander, J.; Singla, N.; Kaur, M.; Punia, R.S.; Attri, A.; Alastruey-Izquierdo, A.; Cano-Lira, J.F.; Stchigel, A.M.; Guarro, J. Saksenaea erythrospora, an Emerging Mucoralean Fungus Causing Severe Necrotizing Skin and Soft Tissue Infections—A Study from a Tertiary Care Hospital in North India. Infect. Dis. 2017, 49, 170–177. [Google Scholar] [CrossRef] [PubMed]
  63. Xess, I.; Mohapatra, S.; Shivaprakash, M.R.; Chakrabarti, A.; Benny, G.L.; O’Donnell, K.; Padhye, A.A. Evidence Implicating Thamnostylum lucknowense as an Etiological Agent of Rhino-Orbital Mucormycosis. J. Clin. Microbiol. 2012, 50, 1491–1494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Pan, J.; Tsui, C.; Li, M.; Xiao, K.; de Hoog, G.S.; Verweij, P.E.; Cao, Y.; Lu, H.; Jiang, Y. First Case of Rhinocerebral Mucormycosis Caused by Lichtheimia ornata, with a Review of Lichtheimia Infections. Mycopathologia 2020, 185, 555–567. [Google Scholar] [CrossRef] [PubMed]
  65. Bonifaz, A.; Stchigel, A.M.; Guarro, J.; Guevara, E.; Pintos, L.; Sanchis, M.; Cano-Lira, J.F. Primary Cutaneous Mucormycosis Produced by the New Species Apophysomyces mexicanus. J. Clin. Microbiol. 2014, 52, 4428–4431. [Google Scholar] [CrossRef] [Green Version]
  66. Petrikkos, G.; Skiada, A.; Lortholary, O.; Roilides, E.; Walsh, T.J.; Kontoyiannis, D.P. Epidemiology and Clinical Manifestations of Mucormycosis. Clin. Infect. Dis. 2012, 54, S23–S34. [Google Scholar] [CrossRef]
  67. Rammaert, B.; Lanternier, F.; Poirée, S.; Kania, R.; Lortholary, O. Diabetes and Mucormycosis: A Complex Interplay. Diabetes Metab. 2012, 38, 193–204. [Google Scholar] [CrossRef]
  68. Kontoyiannis, D.P.; Wessel, V.C.; Bodey, G.P.; Rolston, K.V.I. Zygomycosis in the 1990s in a Tertiary-Care Cancer Center. Clin. Infect. Dis. 2000, 30, 851–856. [Google Scholar] [CrossRef] [Green Version]
  69. Maertens, J.; Demuynck, H.; Verbeken, E.; Zachée, P.; Verhoef, G.; Vandenberghe, P.; Boogaerts, M. Mucormycosis in Allogeneic Bone Marrow Transplant Recipients: Report of Five Cases and Review of the Role of Iron Overload in the Pathogenesis. Bone Marrow Transpl. Transplant. 1999, 24, 307–312. [Google Scholar] [CrossRef] [Green Version]
  70. Singh, N.; Aguado, J.M.; Bonatti, H.; Forrest, G.; Gupta, K.L.; Safdar, N.; John, G.T.; Pursell, K.J.; Muñoz, P.; Patel, R.; et al. Zygomycosis in Solid Organ Transplant Recipients: A Prospective, Matched Case-Control Study to Assess Risks for Disease and Outcome. J. Infect. Dis. 2009, 200, 1002–1011. [Google Scholar] [CrossRef]
  71. Moreira, J.; Varon, A.; Galhardo, M.C.; Santos, F.; Lyra, M.; Castro, R.; Oliveira, R.; Lamas, C.C. The Burden of Mucormycosis in HIV-Infected Patients: A Systematic Review. J. Infect. 2016, 73, 181–188. [Google Scholar] [CrossRef] [Green Version]
  72. Boelaert, J.R.; Fenves, A.Z.; Coburn, J.W. Deferoxamine Therapy and Mucormycosis in Dialysis Patients: Report of an International Registry. Am. J. Kidney Dis. 1991, 18, 660–667. [Google Scholar] [CrossRef]
  73. McNab, A.A. Iron Overload Is a Risk Factor for Zygomycosis. Arch. Ophthalmol. 1997, 115, 919. [Google Scholar] [CrossRef]
  74. Meyerowitz, E.A.; Sanchez, S.; Mansour, M.K.; Triant, V.A.; Goldberg, M.B. Isolated Cerebral Mucormycosis in Immunocompetent Adults Who Inject Drugs: Case Reports and Systematic Review of the Literature. Open. Forum Infect. Dis. 2020, 7, ofaa552. [Google Scholar] [CrossRef]
  75. Lelievre, L.; Garcia-Hermoso, D.; Abdoul, H.; Hivelin, M.; Chouaki, T.; Toubas, D.; Mamez, A.-C.; Lantieri, L.; Lortholary, O.; Lanternier, F. Posttraumatic Mucormycosis. Medicine 2014, 93, 395–404. [Google Scholar] [CrossRef] [PubMed]
  76. Patel, A.; Agarwal, R.; Rudramurthy, S.M.; Shevkani, M.; Xess, I.; Sharma, R.; Savio, J.; Sethuraman, N.; Madan, S.; Shastri, P.; et al. Multicenter Epidemiologic Study of Coronavirus Disease–Associated Mucormycosis, India. Emerg. Infect. Dis. 2021, 27, 2349–2359. [Google Scholar] [CrossRef] [PubMed]
  77. Singh, A.K.; Singh, R.; Joshi, S.R.; Misra, A. Mucormycosis in COVID-19: A Systematic Review of Cases Reported Worldwide and in India. Diabetes Metab. Syndr. Clin. Res. Rev. 2021, 15, 102146. [Google Scholar] [CrossRef] [PubMed]
  78. Chakrabarti, A.; Singh, R. Mucormycosis in India: Unique Features. Mycoses 2014, 57, 85–90. [Google Scholar] [CrossRef] [PubMed]
  79. Özbek, L.; Topçu, U.; Manay, M.; Esen, B.H.; Bektas, S.N.; Aydın, S.; Özdemir, B.; Khostelidi, S.N.; Klimko, N.; Cornely, O.; et al. COVID-19-Associated Mucormycosis: A Systematic Review and Meta-Analysis of 958 Cases. Clin. Microbiol. Infect. 2023, 29, 722–731. [Google Scholar] [CrossRef]
  80. Hoenigl, M.; Seidel, D.; Sprute, R.; Cunha, C.; Oliverio, M.; Goldman, G.H.; Ibrahim, A.S.; Carvalho, A. COVID-19-Associated Fungal Infections. Nat. Microbiol. 2022, 7, 1127–1140. [Google Scholar] [CrossRef]
  81. Joshi, S.; Telang, R.; Tambe, M.; Havaldar, R.; Sane, M.; Shaikh, A.; Roy, C.; Yathati, K.; Sonawale, S.; Borkar, R.; et al. Outbreak of Mucormycosis in Coronavirus Disease Patients, Pune, India. Emerg. Infect. Dis. 2022, 28, 1–8. [Google Scholar] [CrossRef]
  82. Almyroudi, M.P.; Akinosoglou, K.; Rello, J.; Blot, S.; Dimopoulos, G. Clinical Phenotypes of COVID-19 Associated Mucormycosis (CAM): A Comprehensive Review. Diagnostics 2022, 12, 3092. [Google Scholar] [CrossRef] [PubMed]
  83. García-Carnero, L.C.; Mora-Montes, H.M. Mucormycosis and COVID-19-Associated Mucormycosis: Insights of a Deadly but Neglected Mycosis. J. Fungi 2022, 8, 445. [Google Scholar] [CrossRef] [PubMed]
  84. Sharma, R.; Kumar, P.; Rauf, A.; Chaudhary, A.; Prajapati, P.K.; Emran, T.B.; Gonçalves Lima, C.M.; Conte-Junior, C.A. Mucormycosis in the COVID-19 Environment: A Multifaceted Complication. Front. Cell Infect. Microbiol. 2022, 12, 964. [Google Scholar] [CrossRef]
  85. Radotra, B.; Challa, S. Pathogenesis and Pathology of COVID-Associated Mucormycosis: What Is New and Why. Curr. Fungal Infect. Rep. 2022, 16, 206–220. [Google Scholar] [CrossRef] [PubMed]
  86. Dogra, S.; Arora, A.; Aggarwal, A.; Passi, G.; Sharma, A.; Singh, G.; Barnwal, R.P. Mucormycosis Amid COVID-19 Crisis: Pathogenesis, Diagnosis, and Novel Treatment Strategies to Combat the Spread. Front. Microbiol. 2022, 12, 4005. [Google Scholar] [CrossRef]
  87. Krishna, V.; Bansal, N.; Morjaria, J.; Kaul, S. COVID-19-Associated Pulmonary Mucormycosis. J. Fungi 2022, 8, 711. [Google Scholar] [CrossRef]
  88. Narayanan, S.; Chua, J.V.; Baddley, J.W. Coronavirus Disease 2019–Associated Mucormycosis: Risk Factors and Mechanisms of Disease. Clin. Infect. Dis. 2022, 74, 1279–1283. [Google Scholar] [CrossRef]
  89. Ghazi, B.K.; Rackimuthu, S.; Wara, U.U.; Mohan, A.; Khawaja, U.A.; Ahmad, S.; Ahmad, S.; Hasan, M.M.; Dos Santos Costa, A.C.; Ahmad, S.; et al. Rampant Increase in Cases of Mucormycosis in India and Pakistan: A Serious Cause for Concern during the Ongoing COVID-19 Pandemic. Am. J. Trop. Med. Hyg. 2021, 105, 1144–1147. [Google Scholar] [CrossRef]
  90. Rammaert, B.; Lanternier, F.; Zahar, J.-R.; Dannaoui, E.; Bougnoux, M.-E.; Lecuit, M.; Lortholary, O. Healthcare-Associated Mucormycosis. Clin. Infect. Dis. 2012, 54, S44–S54. [Google Scholar] [CrossRef] [Green Version]
  91. Antoniadou, A. Outbreaks of Zygomycosis in Hospitals. Clin. Microbiol. Infect. 2009, 15, 55–59. [Google Scholar] [CrossRef] [Green Version]
  92. Kanamori, H.; Rutala, W.A.; Sickbert-Bennett, E.E.; Weber, D.J. Review of Fungal Outbreaks and Infection Prevention in Healthcare Settings during Construction and Renovation. Clin. Infect. Dis. 2015, 61, 433–444. [Google Scholar] [CrossRef]
  93. Krasinski, K.; Holzman, R.S.; Hanna, B.; Greco, M.A.; Graff, M.; Bhogal, M. Nosocomial Fungal Infection during Hospital Renovation. Infect. Control 1985, 6, 278–282. [Google Scholar] [CrossRef] [Green Version]
  94. Rickerts, V.; Böhme, A.; Viertel, A.; Behrendt, G.; Jacobi, V.; Tintelnot, K.; Just-Nübling, G. Cluster of Pulmonary Infections Caused by Cunninghamella Bertholletiae in Immunocompromised Patients. Clin. Infect. Dis. 2000, 31, 910–913. [Google Scholar] [CrossRef] [Green Version]
  95. Abzug, M.J.; Gardner, S.; Glode, M.P.; Cymanski, M.; Roe, M.H.; Odom, L.F. Heliport-Associated Nosocomial Mucormycoses. Infect. Control Hosp. Epidemiol. 1992, 13, 325–326. [Google Scholar] [CrossRef] [PubMed]
  96. El-Mahallawy, H.A.; Khedr, R.; Taha, H.; Shalaby, L.; Mostafa, A. Investigation and Management of a Rhizomucor Outbreak in a Pediatric Cancer Hospital in Egypt. Pediatr. Blood Cancer 2016, 63, 171–173. [Google Scholar] [CrossRef] [PubMed]
  97. Levy, V.; Rio, B.; Bazarbachi, A.; Hunault, M.; Delmer, A.; Zittoun, R.; Blanc, V.; Wolff, M. Two Cases of Epidemic Mucormycosis Infection in Patients with Acute Lymphoblastic Leukemia. Am. J. Hematol. 1996, 52, 64–65. [Google Scholar] [CrossRef]
  98. del Palacio Hernanz, A.; Fereres, J.; Garraus, S.L.; Rodriguez-Noriega, A.; Sanz, F.S. Nosocomial Infection by Rhizomucor pusillus in a Clinical Haematology Unit. J. Hosp. Infect. 1983, 4, 45–49. [Google Scholar] [CrossRef]
  99. Hammond, D.E. Cutaneous Phycomycosis. Report of Three Cases with Identification of Rhizopus. Arch. Dermatol. 1979, 115, 990–992. [Google Scholar] [CrossRef]
  100. Sheldon, D.L. Cutaneous Mucormycosis. Two Documented Cases of Suspected Nosocomial Cause. JAMA J. Am. Med. Assoc. 1979, 241, 1032–1034. [Google Scholar] [CrossRef]
  101. Gartenberg, G.; Bottone, E.J.; Keusch, G.T.; Weitzman, I. Hospital-Acquired Mucormycosis (Rhizopus rhizopodiformis) of Skin and Subcutaneous Tissue. N. Engl. J. Med. 1978, 299, 1115–1118. [Google Scholar] [CrossRef]
  102. Centers for Disease Control and Prevention Nosocomial Outbreak of Rhizopus Infections Associated with Elastoplast Wound Dressings-Minnesota. Morb. Mortal. Wkly. Rep. Report. 1978, 27, 33–34.
  103. Mitchell, S.; Gray, J.; Morgan, M.; Hocking, M.; Durbin, G. Nosocomial Infection with Rhizopus microsporus in Preterm Infants: Association with Wooden Tongue Depressors. Lancet 1996, 348, 441–443. [Google Scholar] [CrossRef] [PubMed]
  104. Maraví-Poma, E.; Rodríguez-Tudela, J.L.; de Jalón, J.G.; Manrique-Larralde, A.; Torroba, L.; Urtasun, J.; Salvador, B.; Montes, M.; Mellado, E.; Rodríguez-Albarrán, F.; et al. Outbreak of Gastric Mucormycosis Associated with the Use of Wooden Tongue Depressors in Critically Ill Patients. Intensive Care Med. 2004, 30, 724–728. [Google Scholar] [CrossRef]
  105. LeMaile-Williams, M.; Burwell, L.A.; Salisbury, D.; Noble-Wang, J.; Arduino, M.; Lott, T.; Brandt, M.E.; Iiames, S.; Srinivasan, A.; Fridkin, S.K. Outbreak of Cutaneous Rhizopus arrhizus Infection Associated with Karaya Ostomy Bags. Clin. Infect. Dis. 2006, 43, e83–e88. [Google Scholar] [CrossRef] [Green Version]
  106. Gamarra, S.; Chaves, M.S.; Cabeza, M.S.; Macedo, D.; Leonardelli, F.; Franco, D.; Boleas, M.; Garcia-Effron, G. Mucormycosis Outbreak Due to Rhizopus microsporus after Arthroscopic Anterior Cruciate Ligament Reconstruction Surgery Evaluated by RAPD and MALDI-TOF Mass Spectrometry. J. Mycol. Med. 2018, 28, 617–622. [Google Scholar] [CrossRef] [PubMed]
  107. Sundermann, A.J.; Clancy, C.J.; Pasculle, A.W.; Liu, G.; Cheng, S.; Cumbie, R.B.; Driscoll, E.; Ayres, A.; Donahue, L.; Buck, M.; et al. Remediation of Mucorales-Contaminated Healthcare Linens at a Laundry Facility Following an Investigation of a Case Cluster of Hospital-Acquired Mucormycosis. Clin. Infect. Dis. 2022, 74, 1401–1407. [Google Scholar] [CrossRef]
  108. Duffy, J.; Harris, J.; Gade, L.; Sehulster, L.; Newhouse, E.; O’Connell, H.; Noble-Wang, J.; Rao, C.; Balajee, S.A.; Chiller, T. Mucormycosis Outbreak Associated with Hospital Linens. Pediatr. Infect. Dis. J. 2014, 33, 472–476. [Google Scholar] [CrossRef]
  109. Cheng, V.C.C.; Chen, J.H.K.; Wong, S.C.Y.; Leung, S.S.M.; So, S.Y.C.; Lung, D.C.; Lee, W.-M.; Trendell-Smith, N.J.; Chan, W.-M.; Ng, D.; et al. Hospital Outbreak of Pulmonary and Cutaneous Zygomycosis Due to Contaminated Linen Items from Substandard Laundry. Clin. Infect. Dis. 2016, 62, 714–721. [Google Scholar] [CrossRef] [Green Version]
  110. Sundermann, A.J.; Clancy, C.J.; Pasculle, A.W.; Liu, G.; Cumbie, R.B.; Driscoll, E.; Ayres, A.; Donahue, L.; Pergam, S.A.; Abbo, L.; et al. How Clean Is the Linen at My Hospital? The Mucorales on Unclean Linen Discovery Study of Large United States Transplant and Cancer Centers. Clin. Infect. Dis. 2019, 68, 850–853. [Google Scholar] [CrossRef]
  111. Jordan, A.; James, A.E.; Gold, J.A.W.; Wu, K.; Glowicz, J.; Wolfe, F.; Vyas, K.; Litvintseva, A.; Gade, L.; Liverett, H.; et al. Investigation of a Prolonged and Large Outbreak of Healthcare-Associated Mucormycosis Cases in an Acute Care Hospital—Arkansas, June 2019–May 2021. Open. Forum Infect. Dis. 2022, 9, ofac510. [Google Scholar] [CrossRef]
  112. Garner, D.; Machin, K. Investigation and Management of an Outbreak of Mucormycosis in a Paediatric Oncology Unit. J. Hosp. Infect. 2008, 70, 53–59. [Google Scholar] [CrossRef] [PubMed]
  113. Vallabhaneni, S.; Walker, T.A.; Lockhart, S.R.; Ng, D.; Chiller, T.; Melchreit, R.; Brandt, M.E.; Smith, R.M. Centers for Disease Control and Prevention (CDC) Notes from the Field: Fatal Gastrointestinal Mucormycosis in a Premature Infant Associated with a Contaminated Dietary Supplement—Connecticut, 2014. MMWR Morb. Mortal. Wkly. Rep. 2015, 64, 155–156. [Google Scholar] [PubMed]
  114. Lee, S.C.; Billmyre, R.B.; Li, A.; Carson, S.; Sykes, S.M.; Huh, E.Y.; Mieczkowski, P.; Ko, D.C.; Cuomo, C.A.; Heitman, J. Analysis of a Food-Borne Fungal Pathogen Outbreak: Virulence and Genome of a Mucor Circinelloides Isolate from Yogurt. mBio 2014, 5, e01390-14. [Google Scholar] [CrossRef] [Green Version]
  115. Bouakline, A.; Lacroix, C.; Roux, N.; Gangneux, J.P.; Derouin, F. Fungal Contamination of Food in Hematology Units. J. Clin. Microbiol. 2000, 38, 4272–4273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Cheng, V.C.C.; Chan, J.F.W.; Ngan, A.H.Y.; To, K.K.W.; Leung, S.Y.; Tsoi, H.W.; Yam, W.C.; Tai, J.W.M.; Wong, S.S.Y.; Tse, H.; et al. Outbreak of Intestinal Infection Due to Rhizopus microsporus. J. Clin. Microbiol. 2009, 47, 2834–2843. [Google Scholar] [CrossRef] [Green Version]
  117. Poirier, P.; Nourrisson, C.; Gibold, L.; Chalus, E.; Guelon, D.; Descamp, S.; Traore, O.; Cambon, M.; Aumeran, C. Three Cases of Cutaneous Mucormycosis with Lichtheimia spp. (Ex Absidia/Mycocladus) in ICU. Possible Cross-Transmission in an Intensive Care Unit between 2 Cases. J. Mycol. Med. 2013, 23, 265–269. [Google Scholar] [CrossRef] [PubMed]
  118. Thatipelli, S.; Santoiemma, P.; Echenique, I.A.; Green, R.; Ison, M.G.; Ladner, D.; Kanwar, Y.S.; Stosor, V. Donor-derived Renal Allograft Mucormycosis in a Combined Liver and Kidney Transplantation: Case Report and Review of the Literature. Transpl. Transplant. Infect. Dis. 2021, 23, e13534. [Google Scholar] [CrossRef]
  119. Zhan, H.X.; Lv, Y.; Zhang, Y.; Liu, C.; Wang, B.; Jiang, Y.Y.; Liu, X.M. Hepatic and Renal Artery Rupture Due to Aspergillus and Mucor Mixed Infection after Combined Liver and Kidney Transplantation: A Case Report. Transpl. Transplant. Proc. 2008, 40, 1771–1773. [Google Scholar] [CrossRef]
  120. Davari, H.R.; Malekhossini, S.A.; Salahi, H.; Bahador, A.; Saberifirozi, M.; Geramizadeh, B.; Lahsaee, S.M.; Khosravi, M.B.; Imanieh, M.H.; Bagheri, M.H. Outcome of Mucormycosis in Liver Transplantation: Four Cases and a Review of Literature. Exp. Clin. Transpl. Transplant. 2003, 1, 147–152. [Google Scholar]
  121. Devauchelle, P.; Jeanne, M.; Fréalle, E. Mucormycosis in Burn Patients. J. Fungi 2019, 5, 25. [Google Scholar] [CrossRef] [Green Version]
  122. Littlehales, E.; Teague, R.; Andrew, D.; Yassaie, E. Mucormycosis in Burns: A Review. J. Burn. Care Res. 2022, 43, 353–360. [Google Scholar] [CrossRef] [PubMed]
  123. Christiaens, G.; Hayette, M.P.; Jacquemin, D.; Melin, P.; Mutsers, J.; De Mol, P. An Outbreak of Absidia corymbifera Infection Associated with Bandage Contamination in a Burns Unit. J. Hosp. Infect. 2005, 61, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fréalle, E.; Rocchi, S.; Bacus, M.; Bachelet, H.; Pasquesoone, L.; Tavernier, B.; Mathieu, D.; Millon, L.; Jeanne, M. Real-Time Polymerase Chain Reaction Detection of Lichtheimia Species in Bandages Associated with Cutaneous Mucormycosis in Burn Patients. J. Hosp. Infect. 2018, 99, 68–74. [Google Scholar] [CrossRef] [PubMed]
  125. Hartnett, K.P.; Jackson, B.R.; Perkins, K.M.; Glowicz, J.; Kerins, J.L.; Black, S.R.; Lockhart, S.R.; Christensen, B.E.; Beer, K.D. A Guide to Investigating Suspected Outbreaks of Mucormycosis in Healthcare. J. Fungi 2019, 5, 69. [Google Scholar] [CrossRef] [Green Version]
  126. Davoudi, S.; Graviss, L.S.; Kontoyiannis, D.P. Healthcare-Associated Outbreaks Due to Mucorales and Other Uncommon Fungi. Eur. J. Clin. Investig. 2015, 45, 767–773. [Google Scholar] [CrossRef]
  127. Foster, C.E.; Revell, P.A.; Campbell, J.R.; Marquez, L. Healthcare-Associated Pediatric Cutaneous Mucormycosis at Texas Children’s Hospital, 2012–2019. Pediatr. Infect. Dis. J. 2021, 40, 746–748. [Google Scholar] [CrossRef]
  128. Marek, C.; Croxen, M.A.; Dingle, T.C.; Bharat, A.; Schwartz, I.S.; Wiens, R.; Smith, S. The Use of Genome Sequencing to Investigate an Outbreak of Hospital-acquired Mucormycosis in Transplant Patients. Transpl. Transplant. Infect. Dis. 2019, 21, e13163. [Google Scholar] [CrossRef] [PubMed]
  129. Garcia-Hermoso, D.; Criscuolo, A.; Lee, S.C.; Legrand, M.; Chaouat, M.; Denis, B.; Lafaurie, M.; Rouveau, M.; Soler, C.; Schaal, J.-V.; et al. Outbreak of Invasive Wound Mucormycosis in a Burn Unit Due to Multiple Strains of Mucor circinelloides f. circinelloides Resolved by Whole-Genome Sequencing. mBio 2018, 9, e00573-18. [Google Scholar] [CrossRef] [Green Version]
  130. Bowers, J.R.; Monroy-Nieto, J.; Gade, L.; Travis, J.; Refojo, N.; Abrantes, R.; Santander, J.; French, C.; Dignani, M.C.; Hevia, A.I.; et al. Rhizopus microsporus Infections Associated with Surgical Procedures, Argentina, 2006–2014. Emerg. Infect. Dis. 2020, 26, 937–944. [Google Scholar] [CrossRef]
  131. Nguyen, M.H.; Kaul, D.; Muto, C.; Cheng, S.J.; Richter, R.A.; Bruno, V.M.; Liu, G.; Beyhan, S.; Sundermann, A.J.; Mounaud, S.; et al. Genetic Diversity of Clinical and Environmental Mucorales Isolates Obtained from an Investigation of Mucormycosis Cases among Solid Organ Transplant Recipients. Microb. Genom. 2020, 6, mgen000473. [Google Scholar] [CrossRef]
  132. Holzel, H.; Macqueen, S.; MacDonald, A.; Alexander, S.; Campbell, C.K.; Johnson, E.M.; Warnock, D.W. Rhizopus microsporus in Wooden Tongue Depressors: A Major Threat or Minor Inconvenience? J. Hosp. Infect. 1998, 38, 113–118. [Google Scholar] [CrossRef]
  133. Verweij, P.E.; Voss, A.; Donnelly, J.P.; de Pauw, B.E.; Meis, J.F. Wooden Sticks as the Source of a Pseudoepidemic of Infection with Rhizopus microsporus var. rhizopodiformis among Immunocompromised Patients. J. Clin. Microbiol. 1997, 35, 2422–2423. [Google Scholar] [CrossRef] [Green Version]
  134. Harper, J.; Coulter, C.; Lye, G.; Nimmo, G. Rhizopus and Tongue Depressors. Lancet 1996, 348, 1250. [Google Scholar] [CrossRef]
  135. Benedict, K.; Park, B.J. Invasive Fungal Infections after Natural Disasters. Emerg. Infect. Dis. 2014, 20, 349–355. [Google Scholar] [CrossRef] [PubMed]
  136. Floret, N.; Viel, J.-F.; Mauny, F.; Hoen, B.; Piarroux, R. Negligible Risk for Epidemics after Geophysical Disasters. Emerg. Infect. Dis. 2006, 12, 543–548. [Google Scholar] [CrossRef] [PubMed]
  137. Patiño, J.F.; Castro, D.; Valencia, A.; Morales, P. Necrotizing Soft Tissue Lesions after a Volcanic Cataclysm. World J. Surg. 1991, 15, 240–247. [Google Scholar] [CrossRef]
  138. Snell, B.J.; Tavakoli, K. Necrotizing Fasciitis Caused by Apophysomyces elegans Complicating Soft-Tissue and Pelvic Injuries in a Tsunami Survivor from Thailand. Plast. Reconstr. Surg. 2007, 119, 448–449. [Google Scholar] [CrossRef]
  139. Andresen, D.; Donaldson, A.; Choo, L.; Knox, A.; Klaassen, M.; Ursic, C.; Vonthethoff, L.; Krilis, S.; Konecny, P. Multifocal Cutaneous Mucormycosis Complicating Polymicrobial Wound Infections in a Tsunami Survivor from Sri Lanka. Lancet 2005, 365, 876–878. [Google Scholar] [CrossRef]
  140. Maegele, M.; Gregor, S.; Yuecel, N.; Simanski, C.; Paffrath, T.; Rixen, D.; Heiss, M.; Rudroff, C.; Saad, S.; Perbix, W.; et al. One Year Ago Not Business as Usual: Wound Management, Infection and Psychoemotional Control during Tertiary Medical Care Following the 2004 Tsunami Disaster in Southeast Asia. Crit. Care 2006, 10, R50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Neblett Fanfair, R.; Benedict, K.; Bos, J.; Bennett, S.D.; Lo, Y.-C.; Adebanjo, T.; Etienne, K.; Deak, E.; Derado, G.; Shieh, W.-J.; et al. Necrotizing Cutaneous Mucormycosis after a Tornado in Joplin, Missouri, in 2011. N. Engl. J. Med. 2012, 367, 2214–2225. [Google Scholar] [CrossRef] [Green Version]
  142. Wurster, S.; Tatara, A.M.; Albert, N.D.; Ibrahim, A.S.; Heitman, J.; Lee, S.C.; Shetty, A.C.; McCracken, C.; Graf, K.T.; Mikos, A.G.; et al. Tornadic Shear Stress Induces a Transient, Calcineurin-Dependent Hypervirulent Phenotype in Mucorales Molds. mBio 2020, 11, e01414-20. [Google Scholar] [CrossRef] [PubMed]
  143. Davies, B.W.; Smith, J.M.; Hink, E.M.; Durairaj, V.D. Increased Incidence of Rhino-Orbital-Cerebral Mucormycosis after Colorado Flooding. Ophthalmic Plast. Reconstr. Surg. 2017, 33, S148–S151. [Google Scholar] [CrossRef] [PubMed]
  144. Wurster, S.; Paraskevopoulos, T.; Toda, M.; Jiang, Y.; Tarrand, J.J.; Williams, S.; Chiller, T.M.; Jackson, B.R.; Kontoyiannis, D.P. Invasive Mould Infections in Patients from Floodwater-Damaged Areas after Hurricane Harvey—A Closer Look at an Immunocompromised Cancer Patient Population. J. Infect. 2022, 84, 701–709. [Google Scholar] [CrossRef]
  145. Jung, J.; Kim, M.Y.; Lee, H.J.; Park, Y.S.; Lee, S.-O.; Choi, S.-H.; Kim, Y.S.; Woo, J.H.; Kim, S.-H. Comparison of Computed Tomographic Findings in Pulmonary Mucormycosis and Invasive Pulmonary Aspergillosis. Clin. Microbiol. Infect. 2015, 21, 684.e11–684.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Mazzai, L.; Anglani, M.; Giraudo, C.; Martucci, M.; Cester, G.; Causin, F. Imaging Features of Rhinocerebral Mucormycosis: From Onset to Vascular Complications. Acta Radiol. 2022, 63, 232–244. [Google Scholar] [CrossRef]
  147. Pruthi, H.; Muthu, V.; Bhujade, H.; Sharma, A.; Baloji, A.; Ratnakara, R.G.; Bal, A.; Singh, H.; Sandhu, M.S.; Negi, S.; et al. Pulmonary Artery Pseudoaneurysm in COVID-19-Associated Pulmonary Mucormycosis: Case Series and Systematic Review of the Literature. Mycopathologia 2022, 187, 31–37. [Google Scholar] [CrossRef]
  148. Stanzani, M.; Sassi, C.; Lewis, R.E.; Tolomelli, G.; Bazzocchi, A.; Cavo, M.; Vianelli, N.; Battista, G. High Resolution Computed Tomography Angiography Improves the Radiographic Diagnosis of Invasive Mold Disease in Patients with Hematological Malignancies. Clin. Infect. Dis. 2015, 60, 1603–1610. [Google Scholar] [CrossRef] [Green Version]
  149. Douglas, A.; Lau, E.; Thursky, K.; Slavin, M. What, Where and Why: Exploring Fluorodeoxyglucose-PET’s Ability to Localise and Differentiate Infection from Cancer. Curr. Opin. Infect. Dis. 2017, 30, 552–564. [Google Scholar] [CrossRef]
  150. Eibschutz, L.S.; Rabiee, B.; Asadollahi, S.; Gupta, A.; Assadi, M.; Alavi, A.; Gholamrezanezhad, A. FDG-PET/CT of COVID-19 and Other Lung Infections. Semin. Nucl. Med. 2022, 52, 61–70. [Google Scholar] [CrossRef] [PubMed]
  151. Longhitano, A.; Alipour, R.; Khot, A.; Bajel, A.; Antippa, P.; Slavin, M.; Thursky, K. The Role of 18F-Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography (FDG PET/CT) in Assessment of Complex Invasive Fungal Disease and Opportunistic Co-infections in Patients with Acute Leukemia Prior to Allogeneic Hematopoietic Cell Transplant. Transpl. Transplant. Infect. Dis. 2021, 23, e13547. [Google Scholar] [CrossRef]
  152. Lass-Flörl, C. Zygomycosis: Conventional Laboratory Diagnosis. Clin. Microbiol. Infect. 2009, 15, 60–65. [Google Scholar] [CrossRef] [Green Version]
  153. Mansoor, S.; Ahmed, T.I.; Happa, K.; Sultan, M.; Manhas, S.; Shamas, S. Spectrum of Mucormycosis before and during COVID-19: Epidemiology, Diagnosis, and Current Therapeutic Interventions. Curr. Fungal Infect. Rep. 2022, 16, 131–142. [Google Scholar] [CrossRef] [PubMed]
  154. Burnham-Marusich, A.R.; Hubbard, B.; Kvam, A.J.; Gates-Hollingsworth, M.; Green, H.R.; Soukup, E.; Limper, A.H.; Kozel, T.R. Conservation of Mannan Synthesis in Fungi of the Zygomycota and Ascomycota Reveals a Broad Diagnostic Target. mSphere 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  155. Jensen, H.E.; Salonen, J.; Ekfors, T.O. The Use of Immunohistochemistry to Improve Sensitivity and Specificity in the Diagnosis of Systemic Mycoses in Patients with Haematological Malignancies. J. Pathol. 1997, 181, 100–105. [Google Scholar] [CrossRef]
  156. Jung, J.; Park, Y.S.; Sung, H.; Song, J.S.; Lee, S.-O.; Choi, S.-H.; Kim, Y.S.; Woo, J.H.; Kim, S.-H. Assessment of the Accuracy of Histomorphologic Diagnosis of Aspergillosis and Mucormycosis by Immunohistochemical Tests. Clin. Infect. Dis. 2015, 61, civ660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Choi, S.; Song, J.S.; Kim, J.Y.; Cha, H.H.; Yun, J.H.; Park, J.W.; Jung, K.H.; Jo, K.M.; Jung, J.; Kim, M.J.; et al. Diagnostic Performance of Immunohistochemistry for the Aspergillosis and Mucormycosis. Mycoses 2019, 62, 1006–1014. [Google Scholar] [CrossRef]
  158. Walsh, T.J.; Gamaletsou, M.N.; McGinnis, M.R.; Hayden, R.T.; Kontoyiannis, D.P. Early Clinical and Laboratory Diagnosis of Invasive Pulmonary, Extrapulmonary, and Disseminated Mucormycosis (Zygomycosis). Clin. Infect. Dis. 2012, 54, S55–S60. [Google Scholar] [CrossRef] [PubMed]
  159. Lackner, M.; Caramalho, R.; Lass-Flörl, C. Laboratory Diagnosis of Mucormycosis: Current Status and Future Perspectives. Future Microbiol. 2014, 9, 683–695. [Google Scholar] [CrossRef]
  160. Kidd, S.; Halliday, C.; Ellis, D. Descriptions of Medical Fungi, 4th ed.; CAB International: Wallingford, UK, 2023. [Google Scholar]
  161. Becker, P.T.; de Bel, A.; Martiny, D.; Ranque, S.; Piarroux, R.; Cassagne, C.; Detandt, M.; Hendrickx, M. Identification of Filamentous Fungi Isolates by MALDI-TOF Mass Spectrometry: Clinical Evaluation of an Extended Reference Spectra Library. Med. Mycol. 2014, 52, 826–834. [Google Scholar] [CrossRef] [Green Version]
  162. De Carolis, E.; Posteraro, B.; Lass-Flörl, C.; Vella, A.; Florio, A.R.; Torelli, R.; Girmenia, C.; Colozza, C.; Tortorano, A.M.; Sanguinetti, M.; et al. Species Identification of Aspergillus, Fusarium and Mucorales with Direct Surface Analysis by Matrix-Assisted Laser Desorption Ionization Time-of-Flight Mass Spectrometry. Clin. Microbiol. Infect. 2012, 18, 475–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Bellemain, E.; Carlsen, T.; Brochmann, C.; Coissac, E.; Taberlet, P.; Kauserud, H. ITS as an Environmental DNA Barcode for Fungi: An in Silico Approach Reveals Potential PCR Biases. BMC Microbiol. 2010, 10, 189. [Google Scholar] [CrossRef] [Green Version]
  164. Nilsson, R.H.; Ryberg, M.; Abarenkov, K.; Sjökvist, E.; Kristiansson, E. The ITS Region as a Target for Characterization of Fungal Communities Using Emerging Sequencing Technologies. FEMS Microbiol. Lett. 2009, 296, 97–101. [Google Scholar] [CrossRef] [PubMed]
  165. Sparks, R.; Halliday, C.; Chen, S.C.-A. Panfungal PCR on Formalin Fixed Paraffin Embedded Tissue—To Proceed or Not Proceed? Pathology 2023, 55, S46. [Google Scholar] [CrossRef]
  166. Trubiano, J.; Dennison, A.; Morrissey, C.; Chua, K.; Halliday, C.; Chen, S.-A.; Spelman, D. Clinical Utility of Panfungal Polymerase Chain Reaction for the Diagnosis of Invasive Fungal Disease: A Single Center Experience. Med. Mycol. 2016, 54, 138–146. [Google Scholar] [CrossRef] [Green Version]
  167. Chibucos, M.C.; Soliman, S.; Gebremariam, T.; Lee, H.; Daugherty, S.; Orvis, J.; Shetty, A.C.; Crabtree, J.; Hazen, T.H.; Etienne, K.A.; et al. An Integrated Genomic and Transcriptomic Survey of Mucormycosis-Causing Fungi. Nat. Commun. 2016, 7, 12218. [Google Scholar] [CrossRef] [Green Version]
  168. Gebremariam, T.; Liu, M.; Luo, G.; Bruno, V.; Phan, Q.T.; Waring, A.J.; Edwards, J.E.; Filler, S.G.; Yeaman, M.R.; Ibrahim, A.S. CotH3 Mediates Fungal Invasion of Host Cells during Mucormycosis. J. Clin. Investig. 2014, 124, 237–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Baldin, C.; Soliman, S.S.M.; Jeon, H.H.; Alkhazraji, S.; Gebremariam, T.; Gu, Y.; Bruno, V.M.; Cornely, O.A.; Leather, H.L.; Sugrue, M.W.; et al. PCR-Based Approach Targeting Mucorales-Specific Gene Family for Diagnosis of Mucormycosis. J. Clin. Microbiol. 2018, 56, e00746-18. [Google Scholar] [CrossRef] [Green Version]
  170. Millon, L.; Herbrecht, R.; Grenouillet, F.; Morio, F.; Alanio, A.; Letscher-Bru, V.; Cassaing, S.; Chouaki, T.; Kauffmann-Lacroix, C.; Poirier, P.; et al. Early Diagnosis and Monitoring of Mucormycosis by Detection of Circulating DNA in Serum: Retrospective Analysis of 44 Cases Collected through the French Surveillance Network of Invasive Fungal Infections (RESSIF). Clin. Microbiol. Infect. 2016, 22, 810.e1–810.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Millon, L.; Caillot, D.; Berceanu, A.; Bretagne, S.; Lanternier, F.; Morio, F.; Letscher-Bru, V.; Dalle, F.; Denis, B.; Alanio, A.; et al. Evaluation of Serum Mucorales Polymerase Chain Reaction (PCR) for the Diagnosis of Mucormycoses: The MODIMUCOR Prospective Trial. Clin. Infect. Dis. 2022, 75, 777–785. [Google Scholar] [CrossRef]
  172. Rocchi, S.; Scherer, E.; Mengoli, C.; Alanio, A.; Botterel, F.; Bougnoux, M.E.; Bretagne, S.; Cogliati, M.; Cornu, M.; Dalle, F.; et al. Interlaboratory Evaluation of Mucorales PCR Assays for Testing Serum Specimens: A Study by the Fungal PCR Initiative and the Modimucor Study Group. Med. Mycol. 2021, 59, 126–138. [Google Scholar] [CrossRef]
  173. Mercier, T.; Reynders, M.; Beuselinck, K.; Guldentops, E.; Maertens, J.; Lagrou, K. Serial Detection of Circulating Mucorales DNA in Invasive Mucormycosis: A Retrospective Multicenter Evaluation. J. Fungi 2019, 5, 113. [Google Scholar] [CrossRef] [Green Version]
  174. Guegan, H.; Iriart, X.; Bougnoux, M.-E.; Berry, A.; Robert-Gangneux, F.; Gangneux, J.-P. Evaluation of MucorGenius® Mucorales PCR Assay for the Diagnosis of Pulmonary Mucormycosis. J. Infect. 2020, 81, 311–317. [Google Scholar] [CrossRef] [PubMed]
  175. Shokouhi, S.; Mirzaei, J.; Mohseni Sajadi, M.; Javadi, A. Comparison of Serum PCR Assay and Histopathology for the Diagnosis of Invasive Aspergillosis and Mucormycosis in Immunocompromised Patients with Sinus Involvement. Curr. Med. Mycol. 2016, 2, 46–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Dolatabadi, S.; Najafzadeh, M.J.; de Hoog, G.S. Rapid Screening for Human-Pathogenic Mucorales Using Rolling Circle Amplification. Mycoses 2014, 57, 67–72. [Google Scholar] [CrossRef] [Green Version]
  177. Lengerova, M.; Racil, Z.; Hrncirova, K.; Kocmanova, I.; Volfova, P.; Ricna, D.; Bejdak, P.; Moulis, M.; Pavlovsky, Z.; Weinbergerova, B.; et al. Rapid Detection and Identification of Mucormycetes in Bronchoalveolar Lavage Samples from Immunocompromised Patients with Pulmonary Infiltrates by Use of High-Resolution Melt Analysis. J. Clin. Microbiol. 2014, 52, 2824–2828. [Google Scholar] [CrossRef] [Green Version]
  178. Huppler, A.R.; Fisher, B.T.; Lehrnbecher, T.; Walsh, T.J.; Steinbach, W.J. Role of Molecular Biomarkers in the Diagnosis of Invasive Fungal Diseases in Children. J. Pediatr. Infect. Dis. Soc. 2017, 6, S32–S44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Sato, K.; Oinuma, K.-I.; Niki, M.; Yamagoe, S.; Miyazaki, Y.; Asai, K.; Yamada, K.; Hirata, K.; Kaneko, Y.; Kakeya, H. Identification of a Novel Rhizopus-Specific Antigen by Screening with a Signal Sequence Trap and Evaluation as a Possible Diagnostic Marker of Mucormycosis. Med. Mycol. 2017, 55, 713–719. [Google Scholar] [CrossRef] [Green Version]
  180. Kaufman, L.; Turner, L.F.; McLaughlin, D.W. Indirect Enzyme-Linked Immunosorbent Assay for Zygomycosis. J. Clin. Microbiol. 1989, 27, 1979–1982. [Google Scholar] [CrossRef] [Green Version]
  181. Wysong, D.R.; Waldorf, A.R. Electrophoretic and Immunoblot Analyses of Rhizopus arrhizus Antigens. J. Clin. Microbiol. 1987, 25, 358–363. [Google Scholar] [CrossRef] [Green Version]
  182. Heldt, S.; Hoenigl, M. Lateral Flow Assays for the Diagnosis of Invasive Aspergillosis: Current Status. Curr. Fungal Infect. Rep. 2017, 11, 45–51. [Google Scholar] [CrossRef] [Green Version]
  183. Orne, C.; Burnham-Marusich, A.; Baldin, C.; Gebremariam, T.; Ibrahim, A.; Kvam, A.; Kozel, T. Cell Wall Fucomannan Is a Biomarker for Diagnosis of Invasive Murine Mucormycosis. In Proceedings of the 28th ECCMID, Madrid, Spain, 21–24 April 2018; pp. 21–24. [Google Scholar]
  184. Potenza, L.; Vallerini, D.; Barozzi, P.; Riva, G.; Forghieri, F.; Zanetti, E.; Quadrelli, C.; Candoni, A.; Maertens, J.; Rossi, G.; et al. Mucorales-Specific T Cells Emerge in the Course of Invasive Mucormycosis and May Be Used as a Surrogate Diagnostic Marker in High-Risk Patients. Blood 2011, 118, 5416–5419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Bacher, P.; Steinbach, A.; Kniemeyer, O.; Hamprecht, A.; Assenmacher, M.; Vehreschild, M.J.G.T.; Vehreschild, J.J.; Brakhage, A.A.; Cornely, O.A.; Scheffold, A. Fungus-Specific CD4+ T Cells for Rapid Identification of Invasive Pulmonary Mold Infection. Am. J. Respir. Crit. Care Med. 2015, 191, 348–352. [Google Scholar] [CrossRef]
  186. Potenza, L.; Vallerini, D.; Barozzi, P.; Riva, G.; Gilioli, A.; Forghieri, F.; Candoni, A.; Cesaro, S.; Quadrelli, C.; Maertens, J.; et al. Mucorales-Specific T Cells in Patients with Hematologic Malignancies. PLoS ONE 2016, 11, e0149108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Chamilos, G.; Lewis, R.E.; Kontoyiannis, D.P. Delaying Amphotericin B–Based Frontline Therapy Significantly Increases Mortality among Patients with Hematologic Malignancy Who Have Zygomycosis. Clin. Infect. Dis. 2008, 47, 503–509. [Google Scholar] [CrossRef]
  188. Koshy, S.; Ismail, N.; Astudillo, C.L.; Haeger, C.M.; Aloum, O.; Acharige, M.T.; Farmakiotis, D.; Baden, L.R.; Marty, F.M.; Kontoyiannis, D.P.; et al. Breath-Based Diagnosis of Invasive Mucormycosis (IM). Open. Forum Infect. Dis. 2017, 4, S53–S54. [Google Scholar] [CrossRef]
  189. Hussain, K.K.; Malavia, D.; MJohnson, E.; Littlechild, J.; Winlove, C.P.; Vollmer, F.; Gow, N.A. Biosensors and Diagnostics for Fungal Detection. J. Fungi 2020, 6, 349. [Google Scholar] [CrossRef]
  190. Liu, X.; Song, Y.; Li, R. The Use of Combined PCR, Fluorescence in Situ Hybridisation and Immunohistochemical Staining to Diagnose Mucormycosis from Formalin-fixed Paraffin-embedded Tissues. Mycoses 2021, 64, 1460–1470. [Google Scholar] [CrossRef]
  191. Mery, A.; Sendid, B.; François, N.; Cornu, M.; Poissy, J.; Guerardel, Y.; Poulain, D. Application of Mass Spectrometry Technology to Early Diagnosis of Invasive Fungal Infections. J. Clin. Microbiol. 2016, 54, 2786–2797. [Google Scholar] [CrossRef] [Green Version]
  192. Guinea, J.; Meletiadis, J.; Arikan-Akdagli, S.; Muehlethaler, K.; Kahlmeter, G.; Arendrup, M.; Subcommittee on Antifungal Susceptibility Testing (AFST) of the ESCMID; European Committee for Antimicrobial Susceptibility Testing (EUCAST). EUCAST Definitive Document E.DEF 9.4. Method for the Determination of Broth Dilution Minimum Inhibitory Concentrations of Antifungal Agents for Conidia Forming Moulds. Available online: https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/AFST/Files/EUCAST_EDef_9.4_method_for_susceptibility_testing_of_moulds.pdf (accessed on 16 April 2023).
  193. Alexander, B.; Procop, G.; Dufresne, P.; Espinel-Ingroff, A.; Fuller, J.; Ghannoum, M.; Hanson, K.; Holliday, D.; Ostrosky-Zeichner, L.; Schuetz, A.; et al. M38—Reference Method for Broth Dilution Antifungal Susceptibility Testing of Filamentous Fungi, 3rd ed.; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2017. [Google Scholar]
  194. Chowdhary, A.; Singh, P.K.; Kathuria, S.; Hagen, F.; Meis, J.F. Comparison of the EUCAST and CLSI Broth Microdilution Methods for Testing Isavuconazole, Posaconazole, and Amphotericin B against Molecularly Identified Mucorales Species. Antimicrob. Agents Chemother. 2015, 59, 7882–7887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Arendrup, M.C.; Jensen, R.H.; Meletiadis, J. In Vitro Activity of Isavuconazole and Comparators against Clinical Isolates of the Mucorales Order. Antimicrob. Agents Chemother. 2015, 59, 7735–7742. [Google Scholar] [CrossRef] [Green Version]
  196. Dannaoui, E. In Vitro Susceptibilities of Zygomycetes to Conventional and New Antifungals. J. Antimicrob. Chemother. 2003, 51, 45–52. [Google Scholar] [CrossRef]
  197. Vidal, P.; Schwarz, P.; Dannaoui, E. Evaluation of the Gradient Concentration Strip Method for Antifungal Susceptibility Testing of Isavuconazole and Comparators for Mucorales Species. Antimicrob. Agents Chemother. 2019, 63, e00838-19. [Google Scholar] [CrossRef] [Green Version]
  198. Escribano, P.; Mesquida, A.; López-Montesinos, S.; Reigadas, E.; Muñoz, P.; Guinea, J. Amphotericin B, Itraconazole, Posaconazole, and Isavuconazole MICs against Clinical Mucorales Isolates Obtained by Visual Inspection and Spectrophotometric Reading According to the EUCAST 9.4 Procedure. Med. Mycol. 2023, 61, myad045. [Google Scholar] [CrossRef]
  199. Wagner, L.; de Hoog, S.; Alastruey-Izquierdo, A.; Voigt, K.; Kurzai, O.; Walther, G. A Revised Species Concept for Opportunistic Mucor Species Reveals Species-Specific Antifungal Susceptibility Profiles. Antimicrob. Agents Chemother. 2019, 63, e00653-19. [Google Scholar] [CrossRef] [Green Version]
  200. Espinel-Ingroff, A.; Chakrabarti, A.; Chowdhary, A.; Cordoba, S.; Dannaoui, E.; Dufresne, P.; Fothergill, A.; Ghannoum, M.; Gonzalez, G.M.; Guarro, J.; et al. Multicenter Evaluation of MIC Distributions for Epidemiologic Cutoff Value Definition To Detect Amphotericin B, Posaconazole, and Itraconazole Resistance among the Most Clinically Relevant Species of Mucorales. Antimicrob. Agents Chemother. 2015, 59, 1745–1750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Vitale, R.G.; de Hoog, G.S.; Schwarz, P.; Dannaoui, E.; Deng, S.; Machouart, M.; Voigt, K.; van de Sande, W.W.J.; Dolatabadi, S.; Meis, J.F.; et al. Antifungal Susceptibility and Phylogeny of Opportunistic Members of the Order Mucorales. J. Clin. Microbiol. 2012, 50, 66–75. [Google Scholar] [CrossRef] [Green Version]
  202. Almyroudis, N.G.; Sutton, D.A.; Fothergill, A.W.; Rinaldi, M.G.; Kusne, S. In Vitro Susceptibilities of 217 Clinical Isolates of Zygomycetes to Conventional and New Antifungal Agents. Antimicrob. Agents Chemother. 2007, 51, 2587–2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Alastruey-Izquierdo, A.; Castelli, M.V.; Cuesta, I.; Monzon, A.; Cuenca-Estrella, M.; Rodriguez-Tudela, J.L. Activity of Posaconazole and Other Antifungal Agents against Mucorales Strains Identified by Sequencing of Internal Transcribed Spacers. Antimicrob. Agents Chemother. 2009, 53, 1686–1689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Badali, H.; Cañete-Gibas, C.; McCarthy, D.; Patterson, H.; Sanders, C.; David, M.P.; Mele, J.; Fan, H.; Wiederhold, N.P. Epidemiology and Antifungal Susceptibilities of Mucoralean Fungi in Clinical Samples from the United States. J. Clin. Microbiol. 2021, 59, e0123021. [Google Scholar] [CrossRef]
  205. Borman, A.M.; Fraser, M.; Patterson, Z.; Palmer, M.D.; Johnson, E.M. In Vitro Antifungal Drug Resistance Profiles of Clinically Relevant Members of the Mucorales (Mucoromycota) Especially with the Newer Triazoles. J. Fungi 2021, 7, 271. [Google Scholar] [CrossRef]
  206. Lamoth, F.; Damonti, L.; Alexander, B.D. Role of Antifungal Susceptibility Testing in Non-Aspergillus Invasive Mold Infections. J. Clin. Microbiol. 2016, 54, 1638–1640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Espinel-Ingroff, A.; Arthington-Skaggs, B.; Iqbal, N.; Ellis, D.; Pfaller, M.A.; Messer, S.; Rinaldi, M.; Fothergill, A.; Gibbs, D.L.; Wang, A. Multicenter Evaluation of a New Disk Agar Diffusion Method for Susceptibility Testing of Filamentous Fungi with Voriconazole, Posaconazole, Itraconazole, Amphotericin B, and Caspofungin. J. Clin. Microbiol. 2007, 45, 1811–1820. [Google Scholar] [CrossRef] [Green Version]
  208. Kaindl, T.; Andes, D.; Engelhardt, M.; Saulay, M.; Larger, P.; Groll, A.H. Variability and Exposure–Response Relationships of Isavuconazole Plasma Concentrations in the Phase 3 SECURE Trial of Patients with Invasive Mould Diseases. J. Antimicrob. Chemother. 2019, 74, 761–767. [Google Scholar] [CrossRef] [Green Version]
  209. Pfaller, M.A.; Rhomberg, P.R.; Wiederhold, N.P.; Gibas, C.; Sanders, C.; Fan, H.; Mele, J.; Kovanda, L.L.; Castanheira, M. In Vitro Activity of Isavuconazole against Opportunistic Fungal Pathogens from Two Mycology Reference Laboratories. Antimicrob. Agents Chemother. 2018, 62, e01230-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Sun, Q.N.; Fothergill, A.W.; McCarthy, D.I.; Rinaldi, M.G.; Graybill, J.R. In Vitro Activities of Posaconazole, Itraconazole, Voriconazole, Amphotericin B, and Fluconazole against 37 Clinical Isolates of Zygomycetes. Antimicrob. Agents Chemother. 2002, 46, 1581–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Carvalhaes, C.G.; Rhomberg, P.R.; Huband, M.D.; Pfaller, M.A.; Castanheira, M. Antifungal Activity of Isavuconazole and Comparator Agents against Contemporaneous Mucorales Isolates from USA, Europe, and Asia-Pacific. J. Fungi 2023, 9, 241. [Google Scholar] [CrossRef] [PubMed]
  212. Schwarz, P.; Cornely, O.A.; Dannaoui, E. Antifungal Combinations in Mucorales: A Microbiological Perspective. Mycoses 2019, 62, myc.12909. [Google Scholar] [CrossRef]
  213. Dannaoui, E.; Afeltra, J.; Meis, J.F.G.M.; Verweij, P.E. In Vitro Susceptibilities of Zygomycetes to Combinations of Antimicrobial Agents. Antimicrob. Agents Chemother. 2002, 46, 2708–2711. [Google Scholar] [CrossRef] [Green Version]
  214. Biswas, C.; Sorrell, T.C.; Djordjevic, J.T.; Zuo, X.; Jolliffe, K.A.; Chen, S.C.-A. In Vitro Activity of Miltefosine as a Single Agent and in Combination with Voriconazole or Posaconazole against Uncommon Filamentous Fungal Pathogens. J. Antimicrob. Chemother. 2013, 68, 2842–2846. [Google Scholar] [CrossRef]
  215. Arikan, S.; Sancak, B.; Alp, S.; Hascelik, G.; Mcnicholas, P. Comparative in vitro Activities of Posaconazole, Voriconazole, Itraconazole, and Amphotericin B against Aspergillus and Rhizopus, and Synergy Testing for Rhizopus. Med. Mycol. 2008, 46, 567–573. [Google Scholar] [CrossRef] [Green Version]
  216. Zhang, S.; Li, R.; Yu, J. Drug Combinations against Mucor irregularis in vitro. Antimicrob. Agents Chemother. 2013, 57, 3395–3397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Torres-Narbona, M.; Guinea, J.; Martínez-Alarcón, J.; Peláez, T.; Bouza, E. In Vitro Activities of Amphotericin B, Caspofungin, Itraconazole, Posaconazole, and Voriconazole against 45 Clinical Isolates of Zygomycetes: Comparison of CLSI M38-A, Sensititre YeastOne, and the Etest. Antimicrob. Agents Chemother. 2007, 51, 1126–1129. [Google Scholar] [CrossRef] [Green Version]
  218. Lamoth, F.; Alexander, B.D. Comparing Etest and Broth Microdilution for Antifungal Susceptibility Testing of the Most-Relevant Pathogenic Molds. J. Clin. Microbiol. 2015, 53, 3176–3181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Caramalho, R.; Maurer, E.; Binder, U.; Araújo, R.; Dolatabadi, S.; Lass-Flörl, C.; Lackner, M. Etest Cannot Be Recommended for in vitro Susceptibility Testing of Mucorales. Antimicrob. Agents Chemother. 2015, 59, 3663–3665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Macedo, D.; Leonardelli, F.; Dudiuk, C.; Theill, L.; Cabeza, M.S.; Gamarra, S.; Garcia-Effron, G. Molecular Confirmation of the Linkage between the Rhizopus oryzae CYP51A Gene Coding Region and Its Intrinsic Voriconazole and Fluconazole Resistance. Antimicrob. Agents Chemother. 2018, 62, e00224-18. [Google Scholar] [CrossRef] [Green Version]
  221. Ma, L.-J.; Ibrahim, A.S.; Skory, C.; Grabherr, M.G.; Burger, G.; Butler, M.; Elias, M.; Idnurm, A.; Lang, B.F.; Sone, T.; et al. Genomic Analysis of the Basal Lineage Fungus Rhizopus oryzae Reveals a Whole-Genome Duplication. PLoS Genet. 2009, 5, e1000549. [Google Scholar] [CrossRef]
  222. Caramalho, R.; Tyndall, J.D.A.; Monk, B.C.; Larentis, T.; Lass-Flörl, C.; Lackner, M. Intrinsic Short-Tailed Azole Resistance in Mucormycetes Is Due to an Evolutionary Conserved Aminoacid Substitution of the Lanosterol 14α-Demethylase. Sci. Rep. 2017, 7, 15898. [Google Scholar] [CrossRef] [Green Version]
  223. Garcia-Hermoso, D.; Hoinard, D.; Gantier, J.-C.; Grenouillet, F.; Dromer, F.; Dannaoui, E. Molecular and Phenotypic Evaluation of Lichtheimia corymbifera (Formerly Absidia corymbifera) Complex Isolates Associated with Human Mucormycosis: Rehabilitation of L. ramosa. J. Clin. Microbiol. 2009, 47, 3862–3870. [Google Scholar] [CrossRef] [Green Version]
  224. Etienne, K.A.; Gillece, J.; Hilsabeck, R.; Schupp, J.M.; Colman, R.; Lockhart, S.R.; Gade, L.; Thompson, E.H.; Sutton, D.A.; Neblett-Fanfair, R.; et al. Whole Genome Sequence Typing to Investigate the Apophysomyces Outbreak Following a Tornado in Joplin, Missouri, 2011. PLoS ONE 2012, 7, e49989. [Google Scholar] [CrossRef] [Green Version]
  225. Findley, K.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M.; Kong, H.H.; et al. Topographic Diversity of Fungal and Bacterial Communities in Human Skin. Nature 2013, 498, 367–370. [Google Scholar] [CrossRef] [Green Version]
  226. Shelburne, S.A.; Ajami, N.J.; Chibucos, M.C.; Beird, H.C.; Tarrand, J.; Galloway-Peña, J.; Albert, N.; Chemaly, R.F.; Ghantoji, S.S.; Marsh, L.; et al. Implementation of a Pan-Genomic Approach to Investigate Holobiont-Infecting Microbe Interaction: A Case Report of a Leukemic Patient with Invasive Mucormycosis. PLoS ONE 2015, 10, e0139851. [Google Scholar] [CrossRef] [PubMed]
  227. Stiller, J.W.; Hall, B.D. The Origin of Red Algae: Implications for Plastid Evolution. Proc. Natl. Acad. Sci. USA 1997, 94, 4520–4525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Rudramurthy, S.M.; Hoenigl, M.; Meis, J.F.; Cornely, O.A.; Muthu, V.; Gangneux, J.P.; Perfect, J.; Chakrabarti, A. ECMM/ISHAM Recommendations for Clinical Management of COVID-19 Associated Mucormycosis in Low- and Middle-income Countries. Mycoses 2021, 64, 1028–1037. [Google Scholar] [CrossRef] [PubMed]
  229. Bupha-Intr, O.; Butters, C.; Reynolds, G.; Kennedy, K.; Meyer, W.; Patil, S.; Bryant, P.; Morrissey, C.O.; Slavin, M.A.; Thursky, K.A.; et al. Consensus Guidelines for the Diagnosis and Management of Invasive Fungal Disease Due to Moulds Other than Aspergillus in the Haematology/Oncology Setting, 2021. Intern. Med. J. 2021, 51, 177–219. [Google Scholar] [CrossRef] [PubMed]
  230. Smith, C.; Lee, S.C. Current Treatments against Mucormycosis and Future Directions. PLoS Pathog. 2022, 18, e1010858. [Google Scholar] [CrossRef]
  231. Ganesan, P.; Ganapathy, D.; Sekaran, S.; Murthykumar, K.; Sundramoorthy, A.K.; Pitchiah, S.; Shanmugam, R. Molecular Mechanisms of Antifungal Resistance in Mucormycosis. Biomed. Res. Int. 2022, 2022, 6722245. [Google Scholar] [CrossRef]
  232. Hoekstra, W.J.; Garvey, E.P.; Moore, W.R.; Rafferty, S.W.; Yates, C.M.; Schotzinger, R.J. Design and Optimization of Highly-Selective Fungal CYP51 Inhibitors. Bioorganic Med. Chem. Lett. 2014, 24, 3455–3458. [Google Scholar] [CrossRef]
  233. Gebremariam, T.; Wiederhold, N.P.; Fothergill, A.W.; Garvey, E.P.; Hoekstra, W.J.; Schotzinger, R.J.; Patterson, T.F.; Filler, S.G.; Ibrahim, A.S. VT-1161 Protects Immunosuppressed Mice from Rhizopus arrhizus var. arrhizus Infection. Antimicrob. Agents Chemother. 2015, 59, 7815–7817. [Google Scholar] [CrossRef] [Green Version]
  234. Gebremariam, T.; Alkhazraji, S.; Lin, L.; Wiederhold, N.P.; Garvey, E.P.; Hoekstra, W.J.; Schotzinger, R.J.; Patterson, T.F.; Filler, S.G.; Ibrahim, A.S. Prophylactic Treatment with VT-1161 Protects Immunosuppressed Mice from Rhizopus arrhizus var. arrhizus Infection. Antimicrob. Agents Chemother. 2017, 61, e00390-17. [Google Scholar] [CrossRef] [Green Version]
  235. Gebremariam, T.; Gu, Y.; Alkhazraji, S.; Youssef, E.; Shaw, K.J.; Ibrahim, A.S. The Combination Treatment of Fosmanogepix and Liposomal Amphotericin B Is Superior to Monotherapy in Treating Experimental Invasive Mold Infections. Antimicrob. Agents Chemother. 2022, 66, e0038022. [Google Scholar] [CrossRef]
  236. Gebremariam, T.; Alkhazraji, S.; Alqarihi, A.; Wiederhold, N.P.; Shaw, K.J.; Patterson, T.F.; Filler, S.G.; Ibrahim, A.S. Fosmanogepix (APX001) Is Effective in the Treatment of Pulmonary Murine Mucormycosis Due to Rhizopus arrhizus. Antimicrob. Agents Chemother. 2020, 64, e00178-20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Fu, Y.; Estoppey, D.; Roggo, S.; Pistorius, D.; Fuchs, F.; Studer, C.; Ibrahim, A.S.; Aust, T.; Grandjean, F.; Mihalic, M.; et al. Jawsamycin Exhibits in vivo Antifungal Properties by Inhibiting Spt14/Gpi3-Mediated Biosynthesis of Glycosylphosphatidylinositol. Nat. Commun. 2020, 11, 3387. [Google Scholar] [CrossRef] [PubMed]
  238. Dannaoui, E.; Schwarz, P.; Lortholary, O. In Vitro Interactions between Antifungals and Immunosuppressive Drugs against Zygomycetes. Antimicrob. Agents Chemother. 2009, 53, 3549–3551. [Google Scholar] [CrossRef] [Green Version]
  239. Shirazi, F.; Kontoyiannis, D.P. The Calcineurin Pathway Inhibitor Tacrolimus Enhances the in vitro Activity of Azoles against Mucorales via Apoptosis. Eukaryot. Cell 2013, 12, 1225–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Schwarz, P.; Schwarz, P.V.; Felske-Zech, H.; Dannaoui, E. In Vitro Interactions between Isavuconazole and Tacrolimus, Cyclosporin A or Sirolimus against Mucorales. J. Antimicrob. Chemother. 2019, 74, 1921–1927. [Google Scholar] [CrossRef]
  241. Bastidas, R.J.; Shertz, C.A.; Lee, S.C.; Heitman, J.; Cardenas, M.E. Rapamycin Exerts Antifungal Activity in vitro and in vivo against Mucor circinelloides via FKBP12-Dependent Inhibition of Tor. Eukaryot. Cell 2012, 11, 270–281. [Google Scholar] [CrossRef] [Green Version]
  242. Lewis, R.E.; Ben-Ami, R.; Best, L.; Albert, N.; Walsh, T.J.; Kontoyiannis, D.P. Tacrolimus Enhances the Potency of Posaconazole Against Rhizopus oryzae in vitro and in an Experimental Model of Mucormycosis. J. Infect. Dis. 2013, 207, 834–841. [Google Scholar] [CrossRef] [Green Version]
  243. Colley, T.; Alanio, A.; Kelly, S.L.; Sehra, G.; Kizawa, Y.; Warrilow, A.G.S.; Parker, J.E.; Kelly, D.E.; Kimura, G.; Anderson-Dring, L.; et al. In vitro and in vivo Antifungal Profile of a Novel and Long-Acting Inhaled Azole, PC945, on Aspergillus fumigatus Infection. Antimicrob. Agents Chemother. 2017, 61, e02280-16. [Google Scholar] [CrossRef] [Green Version]
  244. Koselny, K.; Green, J.; DiDone, L.; Halterman, J.P.; Fothergill, A.W.; Wiederhold, N.P.; Patterson, T.F.; Cushion, M.T.; Rappelye, C.; Wellington, M.; et al. The Celecoxib Derivative AR-12 Has Broad-Spectrum Antifungal Activity in vitro and Improves the Activity of Fluconazole in a Murine Model of Cryptococcosis. Antimicrob. Agents Chemother. 2016, 60, 7115–7127. [Google Scholar] [CrossRef] [Green Version]
  245. Pfaller, M.A.; Messer, S.A.; Georgopapadakou, N.; Martell, L.A.; Besterman, J.M.; Diekema, D.J. Activity of MGCD290, a Hos2 Histone Deacetylase Inhibitor, in Combination with Azole Antifungals against Opportunistic Fungal Pathogens. J. Clin. Microbiol. 2009, 47, 3797–3804. [Google Scholar] [CrossRef] [Green Version]
  246. Galgóczy, L.; Lukács, G.; Nyilasi, I.; Papp, T.; Vágvölgyi, C. Antifungal Activity of Statins and Their Interaction with Amphotericin B against Clinically Important Zygomycetes. Acta Biol. Hung. 2010, 61, 356–365. [Google Scholar] [CrossRef] [PubMed]
  247. Nyilasi, I.; Kocsubé, S.; Krizsán, K.; Galgóczy, L.; Pesti, M.; Papp, T.; Vágvölgyi, C. In Vitro Synergistic Interactions of the Effects of Various Statins and Azoles against Some Clinically Important Fungi. FEMS Microbiol. Lett. 2010, 307, 175–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Gebremariam, T.; Alkhazraji, S.; Soliman, S.S.M.; Gu, Y.; Jeon, H.H.; Zhang, L.; French, S.W.; Stevens, D.A.; Edwards, J.E.; Filler, S.G.; et al. Anti-CotH3 Antibodies Protect Mice from Mucormycosis by Prevention of Invasion and Augmenting Opsonophagocytosis. Sci. Adv. 2019, 5, eaaw1327. [Google Scholar] [CrossRef] [Green Version]
  249. Goel, P.; Jain, V.; Sengar, M.; Mohta, A.; Das, P.; Bansal, P. Gastrointestinal Mucormycosis: A Success Story and Appraisal of Concepts. J. Infect. Public Health 2013, 6, 58–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Ben-Ami, R.; Lewis, R.E.; Tarrand, J.; Leventakos, K.; Kontoyiannis, D.P. Antifungal Activity of Colistin against Mucorales Species in vitro and in a Murine Model of Rhizopus oryzae Pulmonary Infection. Antimicrob. Agents Chemother. 2010, 54, 484–490. [Google Scholar] [CrossRef] [Green Version]
  251. Chatterjee, K.; Taneja, J.; Khullar, S.; Pandey, A.K. Antifungal Activity of Silver Nanoparticles on Fungal Isolates from Patients of Suspected Mucormycosis. Int. Microbiol. 2022, 26, 143–147. [Google Scholar] [CrossRef]
  252. Elfiky, A.A. Dual Targeting of RdRps of SARS-CoV-2 and the Mucormycosis-Causing Fungus: An in silico Perspective. Future Microbiol. 2022, 17, 755–762. [Google Scholar] [CrossRef]
Figure 1. (A) Chest computed tomography in a patient with COVID-19 demonstrating a large mass in the right lower lobe of the lung with the beginnings of cavitation subsequently diagnosed as mucormycosis. (B) Histopathology demonstrating irregular broad aseptate hyphae (Rhizopus spp.) visualized by Periodic Acid Schiff staining on lung biopsy of the mass in panel A.
Figure 1. (A) Chest computed tomography in a patient with COVID-19 demonstrating a large mass in the right lower lobe of the lung with the beginnings of cavitation subsequently diagnosed as mucormycosis. (B) Histopathology demonstrating irregular broad aseptate hyphae (Rhizopus spp.) visualized by Periodic Acid Schiff staining on lung biopsy of the mass in panel A.
Jof 09 00659 g001
Figure 2. Algorithm for Management of Mucormycosis (adapted from [3,153,228,229]). L-AmB, Liposomal Amphotericin B; c-AmB, Amphotericin B deoxycholate.
Figure 2. Algorithm for Management of Mucormycosis (adapted from [3,153,228,229]). L-AmB, Liposomal Amphotericin B; c-AmB, Amphotericin B deoxycholate.
Jof 09 00659 g002
Table 1. Clinically relevant species of Mucorales with their modern assignments in the context of their family structure (as published by Hoffman et al. [16] and adapted from Walther et al. [15]).
Table 1. Clinically relevant species of Mucorales with their modern assignments in the context of their family structure (as published by Hoffman et al. [16] and adapted from Walther et al. [15]).
FamilyGenusClinically Relevant Species
MucoraceaeActinomucorA. elegans
SaksenaeaceaeApophysomycesA. mexicanus
A. ossiformis
A. trapeziformis
A. variabilis
MucoraceaeCokeromycesC. recurvatus
CunninghamellaceaeCunninghamellaC. arunalokei [20]
C. bertholletiae
C. blakesleeana
C. echinulata
C. elegans
LichtheimiaceaeLichtheimiaL. corymbifera
L. ornata
L. ramosa
MucoraceaeMucorM. amphibiorum
M. circinelloides *
M. griseocyanus *
M. indicus
M. irregularis
M. janssenii *
M. lusitanicus *
M. plumbeus
M. racemosus
M. ramosissimus *
M. variicolumellatus *
M. velutinosus *
LichtheimiaceaeRhizomucorR. miehei
R. pusillus
RhizopodaceaeRhizopusR. arrhizus (including var. arrhizus and var. delemar)
R. homothallicus
R. microsporus
R. schipperae
SaksenaeaceaeSaksenaeaS. erythrospora
S. loutrophoriformis
S. trapezispora
S. vasiformis
SyncephalastraceaeSyncephalastrumS. racemosum
LichtheimiaceaeThamnostylumT. lucknowense
* Member of the Mucor circinelloides complex [18].
Table 2. Classical host factors with associated clinical syndromes of mucormycosis.
Table 2. Classical host factors with associated clinical syndromes of mucormycosis.
Host FactorAssociated Clinical SyndromeReferences
Diabetes mellitus, particularly with ketoacidosisROCM[67]
Corticosteroid useROCM[68]
Haematologic malignanciesPulmonary or disseminated infection[68]
COVID-19ROCM[6]
Haematopoietic cell transplantationPulmonary[69]
Solid organ transplantationDisseminated infection[70]
HIV/AIDSDisseminated infection[71]
Treatment with deferoxamineROCM[72]
Iron overloadROCM[73]
Injection drug useIsolated cerebral[74]
Major traumaCutaneous[75]
BurnsCutaneous[34]
Table 3. Epidemiologic cutoff values (ECVs) of minimum inhibitory concentrations (MICs) of amphotericin B (AMB), posaconazole (POS), and itraconazole (ITR) and for four Mucorales species (adapted from [200]).
Table 3. Epidemiologic cutoff values (ECVs) of minimum inhibitory concentrations (MICs) of amphotericin B (AMB), posaconazole (POS), and itraconazole (ITR) and for four Mucorales species (adapted from [200]).
SpeciesAntifungalMIC (mg/L)Calculated ECV
RangeMode≥95%≥97.5%
L. corymbiferaAMB0.06–160.512
POS0.06–40.512
ITR0.06–80.25Not determined
M. circinelloidesAMB0.03–40.2512
POS0.06–16144
ITR0.25–164Not determined
R. arrhizusAMB0.03–4124
POS0.03–320.512
ITR0.06–160.522
R. microsporusAMB0.06–40.522
POS0.06–160.512
ITR0.25–321Not determined
Table 4. New and novel antifungal agents with in vitro and/or in vivo activity against Mucorales.
Table 4. New and novel antifungal agents with in vitro and/or in vivo activity against Mucorales.
AgentMechanism of ActionEfficacyReferences
Opelconazole (inhaled)Inhibition of formation of ergosterol in fungal cell membraneIn vitro activity against R. arrhizus (syn. oryzae) (ATCC 11145) (MIC 2 mg/L); poor activity against R. pusillus, M. circinelloides, and L. corymbifera[243]
AR-12Celecoxib derivative which inhibits fungal acetyl coenzyme A (acetyl-CoA) synthetaseIn vitro activity against R. arrhizus (MIC 4 mg/L)[244]
MGCD290Inhibits fungal histone deacetylase 2In vitro synergy with triazoles[245]
Statins (fluvastatin, rosuvastatin or atorvastatin)Affect the synthesis of ergosterol by inhibiting 3-hydroxy-3-methylglutaryl-CoA (HMGCoA) reductaseIn vitro synergy with either amphotericin B or various azoles in combination [246,247]
Anti-CotH3 antibodiesMonoclonal or polyclonal antibodies that inhibit CotH3, a fungal cell protein which binds to glucose regulated protein 78 on endothelial cellsIn vivo activity in murine models with improved outcomes.[248]
Colistin Polymyxin antibioticIn vitro and in vivo activity in murine models
Single case report of GI mucormycosis demonstrated treatment success when used in combination with standard of care
[249,250]
Silver nanoparticlesSmall particles with known antimicrobial activityPotent in vitro activity against some R. arrhizus isolates (MIC < 8–64 mg/L)[251]
SofosbuvirAntiviral targeting RNA-dependent RNA polymeraseIn silico experiments demonstrating inhibition of R. arrhizus[252]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pham, D.; Howard-Jones, A.R.; Sparks, R.; Stefani, M.; Sivalingam, V.; Halliday, C.L.; Beardsley, J.; Chen, S.C.-A. Epidemiology, Modern Diagnostics, and the Management of Mucorales Infections. J. Fungi 2023, 9, 659. https://doi.org/10.3390/jof9060659

AMA Style

Pham D, Howard-Jones AR, Sparks R, Stefani M, Sivalingam V, Halliday CL, Beardsley J, Chen SC-A. Epidemiology, Modern Diagnostics, and the Management of Mucorales Infections. Journal of Fungi. 2023; 9(6):659. https://doi.org/10.3390/jof9060659

Chicago/Turabian Style

Pham, David, Annaleise R. Howard-Jones, Rebecca Sparks, Maurizio Stefani, Varsha Sivalingam, Catriona L. Halliday, Justin Beardsley, and Sharon C.-A. Chen. 2023. "Epidemiology, Modern Diagnostics, and the Management of Mucorales Infections" Journal of Fungi 9, no. 6: 659. https://doi.org/10.3390/jof9060659

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop