REVIEW article

Mucormycosis in 2023: an update on pathogenesis and management.

Abdullah Alqarihi

  • 1 Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
  • 2 Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States
  • 3 Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States

Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus , Mucor , and Lichtheimia , accounting for > 90% of all cases. MCR is seen in patients with severe immunosuppression such as those with hematologic malignancy or transplantation, Diabetes Mellitus (DM) and diabetic ketoacidosis (DKA) and immunocompetent patients with severe wounds. The recent SARS COV2 epidemy in India has resulted in a tremendous increase in MCR cases, typically seen in the setting of uncontrolled DM and corticosteroid use. In addition to the diversity of affected hosts, MCR has pleiotropic clinical presentations, with rhino-orbital/rhino-cerebral, sino-pulmonary and necrotizing cutaneous forms being the predominant manifestations. Major insights in MCR pathogenesis have brought into focus the host receptors (GRP78) and signaling pathways (EGFR activation cascade) as well as the adhesins used by Mucorales for invasion. Furthermore, studies have expanded on the importance of iron availability and the complex regulation of iron homeostasis, as well as the pivotal role of mycotoxins as key factors for tissue invasion. The molecular toolbox to study Mucorales pathogenesis remains underdeveloped, but promise is brought by RNAi and CRISPR/Cas9 approaches. Important recent advancements have been made in early, culture-independent molecular diagnosis of MCR. However, development of new potent antifungals against Mucorales remains an unmet need. Therapy of MCR is multidisciplinary and requires a high index of suspicion for initiation of early Mucorales-active antifungals. Reversal of underlying immunosuppression, if feasible, rapid DKA correction and in selected patients, surgical debulking are crucial for improved outcomes.

Introduction

Fungi are ubiquitously found in many environments, and have important roles in the ecosystem and biodiversity as they are essential in nutrient cycling and recycling of waste ( Frąc et al., 2018 ). It is estimated that there are 1.5 million different types of fungi, from which only 300 are known to cause illness (Centers for Disease Control and Prevention, https://www.cdc.gov/fungal/diseases/index.html ). In this review, we focus on Mucorales, a group of commercially and increasingly medically significant molds. Specifically, we provide an overview of the pathogenesis, along with epidemiology (in view of the recent major outbreak of COVID-19 associated mucormycosis in India), pathology and molecular diagnosis, and current therapeutic advances in mucormycosis (MCR).

The importance of Mucorales fungi has been established as multifaceted because of their capacity to release a range of commercially used lytic enzymes including amylases, lipases, and proteases, as well as production of essential medical and pharmacological substances such as steroids and terpenoids as well ( Morin-Sardin et al., 2017 ). However, since its first description by Paltauf in 1885 MCR, the invasive disease caused by a variety of Mucorales has come to central stage in modern mycology and infectious diseases. MCR is a severe and frequently lethal infection which can affect a pleiad of different immunosuppressed patients such as those who have received organ transplants and immunocompetent hosts who are trauma victims of natural disasters ( Spellberg et al., 2005a ; Warkentien et al., 2012 ; Tribble et al., 2013 ; Warkentien et al., 2015 ; Weintrob et al., 2015 ; Spellberg and Ibrahim, 2018 ) ( Figure 1A ).

www.frontiersin.org

Figure 1 Frequency of mucormycosis manifestation in susceptible hosts and the etiologic agents of the disease. (A) Frequency of mucormycosis by underlaying predisposing host condition. (B) Etiological agents of mucormycosis. (C) Frequency of different types of mucormycosis reported. * Data adapted from Roden M et al. CID 2005 ( Roden et al., 2005 ).

Nomenclature and frequency of Mucorales as causes of MCR and burden of the disease

MCR is caused by fungi belonging to the order Mucorales. Rhizopus , Mucor , and Lichtheimia (formerly Absidia ) species are the most common members of the order Mucorales that cause mucormycosis, accounting for >90% of all cases ( Uppuluri et al., 2021 ). Rhizopus species, are the dominant cause of MCR in the entire world responsible for >70% of all cases of MCR ( Ribes et al., 2000 ; Roden et al., 2005 ; Spellberg et al., 2005a ). In contrast, Cunninghamella , Apophysomyces , Saksenaea , Rhizomucor , Cokeromyces , Actinomucor , and Syncephalastrum species individually are responsible for fewer than 1 to 5% of reported MCR cases ( Gomes et al., 2011 ) ( Figure 1B ). Thus, Mucor species, including M. lusitanicus (formerly Mucor circinelloides f. lusitanicus ) ( Wagner et al., 2020 ), and Lichtheimia are the secondary cause of infection in the Americas and Europe, respectively. However, Apophysomyces are the secondary cause of infection in India ( Skiada et al., 2018 ; Nucci et al., 2019 ).

Although it is not possible to determine the exact burden of MCR worldwide, there has been an alarming increase in cases in the last three decades. Over the past 15 years cases have more than doubled at the MD Anderson and Fred-Hutchinson Cancer Centers ( Kontoyiannis et al., 2000 ; Marr et al., 2002 ). According to a scientific study conducted in France, there was a significant increase of 70% between 1997 and 2006. Additionally, there was a substantial rise of 175% in the prevalence of the condition between the years 1988-2006 compared to the period from 2007-2015 ( Roden et al., 2005 ). Also, a medical center in Switzerland reported >10-fold increase in MCR cases among admitted patients after 2003 ( Ambrosioni et al., 2010 ). Cases in Iran more than doubled between 2008-2014 ( Dolatabadi et al., 2018 ). In hematopoietic and allogenic stem cell transplant recipients, MCR is currently the third most prevalent invasive fungal infection, after candidiasis and aspergillosis ( Kontoyiannis et al., 2000 ; Petrikkos et al., 2012 ). That increase in MCR incidence at many transplant centers has been linked to the introduction and widespread use of voriconazole prophylaxis in these high-risk populations. However, it is not known if this association reflects a true epidemiological link or represents a marker of changing immunosuppression occurring in parallel with the evolution of transplant practices and immunosuppression strategies ( Pongas et al., 2009 ).

Before the onset of the COVID-19 era, India was recognized as hyper-endemic for MCR, a fungal infection. The estimated disease burden in India was approximately 70 times higher than the global average, with predicted cases exceeding 200,000 per year. These cases accounted for approximately 24% of all invasive mold infections ( Chakrabarti and Singh, 2014 ; Chakrabarti et al., 2019 ; Prakash and Chakrabarti, 2021 ).

Morphogenesis

Mucorales fungi can reproduce through both sexual and asexual means. Asexual reproduction involves the formation of spherical structures called sporangiospores, which are located at the tip of the sporangium (as depicted in Figure 2A ). These sporangiospores can be released and spread, eventually germinating into hyphae. On the other hand, the sexual cycle of most Mucorales fungi begins with the fusion of two opposing types, denoted as (–) and (+), which leads to the formation of zygospores ( Lee et al., 2010 ). These zygospores then germinate and develop into a sporangium at the apex, resulting in the production of sexual meiospores ( Lee et al., 2010 ). Because the sexual life cycle for sporangiospore generation is protracted, anecdotal evidence suggests that asexual sporangiospores may be the primary source of initiation, propagation and spread of infection.

www.frontiersin.org

Figure 2 (A) Morphology of Rhizopus delemar . Sporangia form at the apices of sporangiophores and contain the asexual sporangiospores. Germinated spores seen in the sporangium magnified box can be an overlay of the sporangium on released and germinated spores. (B) Under normal circumstance, alveolar macrophages (AMS) are able to phagocytize fungi and killing through LC3-associated phagocytosis (LAP+). While AMS are able to phagocytize Mucorales spores, spore melanin is able to arrest LAP to prevent phagosome maturation. However, spores are unable to grow and germinate due to iron restriction ( Frąc et al., 2018 ). In the presence of abnormal nutritional immunity ( i.e. excessive iron) spores are able to germinate and kill Ams ( Andrianaki et al., 2018 ). Courtesy of Dr. Georgios Chamilos. “Created with BioRender.com ”.

Host immune responses

Host barrier cells.

Inhalation of spores from the environment causes rhino-orbital/cerebral, sino-pulmonary MCR, the two most common disease manifestations. Cutaneous MCR is the disease’s third most prevalent presentation, and it usually the consequence of inoculation of Mucorales spores to skin/subcutaneous tissues following severe trauma or abrasions on the skin ( Sugar, 2005 ; Ibrahim et al., 2011 ) ( Figure 1C ). As MCR is characterized by extensive tissue invasion and tissue destruction and is the most angioinvasive of all fungal diseases, dissemination is very common ( Ben-Ami et al., 2009 ).

Mucorales invade nasal and alveolar epithelial cells through binding to host cell glucose-regulated protein 78 kDa (GRP78) and integrin α3β1, respectively ( Alqarihi et al., 2020 ). Watkins et al., 2018 used transcriptome sequencing (RNA-seq) to assess host transcriptional response during early stages of R. delemar infection to gain insight into governed Mucorales-host airway epithelial cell early interactions ( Watkins et al., 2018 ). The host epidermal growth factor receptor (EGFR) signaling was activated during infection, and the alveolar epithelial cell EGFR (using A549 cell line) was also phosphorylated when interacting with several Mucorales organisms. Furthermore, the EGFR co-localized with R. delemar spores during invasion of alveolar epithelial cells ( Alqarihi et al., 2020 ). EGFR inhibitors cetuximab and gefitinib protected airway epithelial cells from R. delemar invasion and injury in vitro . These findings identified EGFR activation cascade as a critical pathway in inducing invasion of alveolar epithelial cells by Mucorales and suggested that adjunctive therapy such as gefitinib can be useful in the treatment of pulmonary MCR ( Watkins et al., 2018 ). Finally, Mucorales fungi appear to hematogenously disseminate by engaging the endothelial GRP78 receptor ( Liu et al., 2010 ).

Innate immunity

The first line of effector immune cells against inhaled Mucorales spores are alveolar macrophages ( Aberdein et al., 2013 ; Ibrahim and Voelz, 2017 ). Immunocompetent mice have macrophages which are efficient in phagocytizing Mucorales spores and preventing their germination ( Diamond and Erickson, 1982 ; Waldorf et al., 1984a ; Waldorf et al., 1984b ; Waldorf, 1989 ). Contrasting to Aspergillus fumigatus conidia, macrophages from immunocompetent individuals are unable to kill phagocytized Mucorales spores, despite being able to adhere to Mucorales hyphae and damage them by oxidative and non-oxidative mechanisms ( Diamond and Clark, 1982 ; Waldorf et al., 1984b ; Waldorf, 1989 ; Lee et al., 2015 ). In contrast to macrophage from immunocompetent hosts, macrophages from diabetic mice cannot prevent spore germination, thereby resulting in established infection and lethality in infected mice ( Waldorf et al., 1984b ). Unraveling the enigma of alveolar macrophages’ enduring presence and their resilience against destruction could be essential in formulating innovative approaches for combating infections. Andrianaki et al., 2018 discovered that alveolar macrophages-phagocytized Mucorales spores retain melanin on their surface and therefore are able to halt phagosome maturation through inhibition of LC3-associated phagocytosis ( Andrianaki et al., 2018 ). Furthermore, research employing transcriptome, iron supplementation, and genetic modification of iron acquisition genes revealed that iron restriction inside macrophages modulates immunity against Rhizopus and suppresses fungus germination ( Figure 2B ) ( Andrianaki et al., 2018 ). Future studies are destined to shed more light into the important role of nutritional immunity to control Mucorales.

Mucorales are resistant to innate immune cells with hyperglycemia impairing chemotaxis and killing activities of polymorphonuclear cells, including neutrophils in the DKA settings ( Chinn and Diamond, 1982 ). As acidosis impairs transferrin’s ability to efficiently chelate iron ( Artis et al., 1982 ; Ibrahim et al., 2008b ; Kontoyiannis et al., 2012 ; Gebremariam et al., 2016 ), the released iron causes further functional impairment in phagocytes ( Cantinieaux et al., 1999 ; Guo et al., 2002 ). Immune cells from mice fed excessive amounts of iron secrete a reduced amount of interferon-gamma (IFN-γ) ( Omara and Blakley, 1994 ), a signature cytokine that orchestrates Mucorales fungal death by effector immune cells ( Gil-Lamaignere et al., 2005 ). IFN-γ and granulocyte-macrophage-colony-stimulating factor (GM-CSF) have specifically, either alone or combined, shown to improve neutrophils’ ability to damage and kill Mucorales hyphae ex vivo by increasing oxidative burst and TNF-α release ( Gil-Lamaignere et al., 2005 ).

Adaptive immunity

The role of adaptive immunity in MCR patients has not been extensively investigated. Similar to the interaction with Aspergillus , the exposure to β-glucan during the germination process of Mucorales fungi triggers dectin-1 signaling in human dendritic cells. This signaling pathway leads to the strong activation of IL-23 and Th-17 responses, similar to the immune responses observed in the presence of Aspergillus ( Chamilos et al., 2010 ). Hyphae can be destroyed in patients who elicit Mucorales-specific T-cells ( Potenza et al., 2011 ; Schmidt et al., 2013 ). Consistent with these results, T-cells that have been pulsed with Rhizopus extract and stimulated with IL-2/IL-7 produce Mucorales-specific T cells with CD4+ cells ( Castillo et al., 2018 ). Instead of non-specific signaling, these cells can produce IFN-γ, IL-5, IL-10, IL-13, and TNF-α and detect fungus antigens processed by HLA-II molecules ( Castillo et al., 2018 ). In addition, emerging evidence points out of the potential role of the benefit of adjunct immune checkpoint inhibitors (ICIs) to treat MCR. In a proof-of-concept study, Wurster et al. studied the effects of PD-1 and PD-L1 inhibitors outcomes and immunopathology of invasive pulmonary MCR in cyclophosphamide- and cortisone acetate-immunosuppressed mice. R. arrhizus -infected mice receiving either of the both PD-1 but even more so by PD-L1-inhibitor (without concomitant antifungals) had significantly improved survival, less morbidity, and lower fungal burden compared to isotype-treated infected mice ( Wurster et al., 2022 ). As inhibition of the PD-1/PD-L1 pathway is not without the potential for immune-related adverse events, future careful dose-effect studies are needed to define the “sweet spot” between ICI-induced augmentation of Mucorales immunity and potential immunotoxicities.

Pathogenicity factors

Hyphal formation.

In response to environmental stimuli, Mucorales can rapidly switch their morphogenetic programs between spores, and mycelia ( Orlowski, 1991 ). Yeast-like form development in Mucor spp. are promoted by the presence of fermentable hexose and Anaerobiosis, whereas oxygen and nutrient constraint promote hyphal growth ( Wolff et al., 2002 ). Gene targets that modulate morphogenesis were identified with gene deletion homologous recombination techniques with autotrophic markers, and this information has led to intriguing therapeutic candidates. specifically, the calcineurin pathway, for example, was found to govern yeast to mycelium transition and influenced pathogenicity in M. lusitanicus ( Lee et al., 2015 ). Specifically, the chemical inhibition of calcineurin or the disruption of the regulatory subunit gene of calcineurin (CnbR) traps the yeast-form, specifically, making it substantially less virulent in mice.

In addition to calcineurin, cyclic AMP (cAMP) and its target protein kinase A (PKA) are thought to have a role in morphogenesis control. A cross-talk between these two regulatory mechanisms is implied, calcineurin inhibits PKA ( Lee et al., 2013 ). Further gene deletions uncovered other proteins involved in the control of M. circinelloides dimorphism, such as heterotrimeric G proteins and ADP-ribosylation factors (Arfs) ( Patiño-Medina et al., 2018 ). Finally, a role in pathogenicity is thought to be played by the size of Mucor spores. Specifically, in Galleria mellonella larvae MCR model large multinucleate spores germinate faster and are more virulent than small mononucleate spores ( Li et al., 2011 ). Small spores are phagocytized more avidly, whereas larger spores can geminate inside macrophages and overpower them.

Effect of iron homeostasis

Like all pathogens, iron is crucial for Mucorales survival in the host. Mammalian cells store iron bound to iron-carrying proteins such as ferritin, lactoferrin and transferrin ( Howard, 1999 ). DKA or other types of acidosis, elevated blood glucose and low blood pH, in patients with hypoglycemia, disturb the avidity of these host proteins to bind iron, resulting in an increase in serum free iron concentration ( Artis et al., 1982 ; Ibrahim, 2014 ; Gebremariam et al., 2016 ). Increased availability of serum free iron levels in the host can enhance the ability of Mucorales to produce a rapidly invasive infection ( Boelaert, 1994 ; Boelaert et al., 1994 ; Ibrahim et al., 2007 ). A high-affinity iron absorption system and the production of siderophores are used by Mucorales to acquire exogenous iron by these two mechanisms ( Carroll et al., 2017 ; Navarro-Mendoza et al., 2018 ; Lax et al., 2020 ). A family of iron reductases (Fre), a ferroxidase (Fet3), and a high affinity iron permease (Ftr1) make up the high-affinity iron acquisition system ( Navarro-Mendoza et al., 2018 ). In R. delemar ( Fu et al., 2004 ; Ibrahim, 2010 ; Liu et al., 2015 ), M. circinelloides ( Navarro-Mendoza et al., 2018 ), and L. corymbifera , low iron availability stimulates the development of the high-affinity iron absorption system ( Schwartze et al., 2014 ). The functions of genes related with the iron uptake in Mucorales was explored using the RNAi gene silencing, particularly in R. delemar (formerly identified as R. oryzae ), which is less amenable to mutagenesis than M. circinelloides ( Ibrahim, 2010 ). In mice with DKA, reduction of the copy number of the FTR1 gene or inhibition of expression by RNAi impairs R. delemar ’s ability to accumulate iron in vitro and lower its pathogenicity ( Ibrahim, 2010 ).

Historically, patients on hemodialysis taking deferoxamine to treat iron overload toxicity had a very high risk for disseminated and frequently lethal MCR ( Boelaert et al., 1987 ; Boelaert et al., 1989 ; Boelaert et al., 1991 ; Boelaert, 1994 ). For its growth, R. delemar uses iron from ferrioxamine as a siderophore (the iron rich form of deferoxamine). According to biochemical and genetic investigations (by RNAi-mediated gene silencing) R. delemar has two surface receptors (Fob1 and Fob2) that bind ferrioxamine and enhance iron intake via a reductase/Ftr-1 mediated pathway ( Liu et al., 2015 ). In addition, three ferroxidase encoding-genes have been identified in M. circinelloides : fet3a, fet3b, and fet3c ( Navarro-Mendoza et al., 2018 ). All three M. circinelloides genes are overexpressed in the lungs of infected mice, and they are regulated by iron availability in the culture media. In addition, there is a relationship of the expression of the different ferroxidase with the fungal dimorphic state, a key virulence factor for Mucorales invasion. Specifically, during aerobic growth, fet3a is expressed specifically in yeast-like growth, while fet3b and fet3c are expressed in hyphae ( Navarro-Mendoza et al., 2018 ). As proof of concept, gene deletion studies revealed that fet3c plays a major role in M. circinelloides virulence in vivo ( Navarro-Mendoza et al., 2018 ).

In addition to using the bacterial deferoxamine as a xenosiderophore, Rhizopus species are known to synthesize and secrete their own Rhizoferrin siderophore ( Carroll et al., 2017 ). This siderophore supplies Rhizopus with iron through a receptor-mediated, and energy dependent process ( Thieken and Winkelmann, 1992 ; de Locht et al., 1994 ). A R. delemar rhizoferrin synthetase (SfnaD), a gene that has homology to the bacterial NRPS-independent siderophore (NIS) protein has been identified in a study ( Carroll et al., 2017 ). The SfnaD contains the C-terminus conserved ferric iron reductase FhuF-like transporter domain. In addition, growing the fungus in iron-rich conditions inhibited the expression of SfnaD in R. delemar , while heterologous expression of this gene allowed E. coli to synthesize the siderophore from citrate and diaminobutane, thereby confirming its identity as a NRPS-independent siderophore protein. It is important to mention that rhizoferrin was shown to be inefficient in chelating iron from serum ( Boelaert et al., 1993 ; Boelaert et al., 1994 ). Consequently, the role of rhizoferrin in Rhizopus virulence might be limited in hosts that do not have excess free-iron, while this siderophore could be operative in hosts with elevated free-iron such as DKA patients. In fact, iron regulation is quite complex in Mucorales. It depends on the specific Mucorales genus and species and the particular context (low or high iron availability).

Finally, the genome project of Mucorales fungi revealed the presence of gene orthologs to heme oxygenase which were shown to be important in acquiring iron from haemin in several fungi ( Worsham and Goldman, 1988 ; Santos et al., 2003 ). It is possible that these heme oxygenase genes are involved in acquiring iron from host hemoglobin and might explain the angioinvasive nature of the disease ( Ibrahim et al., 2008b ; García-Carnero and Mora-Montes, 2022 ).

CotH significant role in invasion

Mucorales interact actively with epithelial cells and the endothelium lining blood vessels to promote initiation of infection and angioinvasion, respectively. Among the fungal kingdom found only in Mucorales are spore coating proteins (CotH) family members, which are kinase-like proteins ( Chibucos et al., 2016 ; Gebremariam et al., 2016 ; Nguyen et al., 2016 ). CotH proteins were first described in Bacillus subtilis in which it is involved in endospore formation. The function of CotH proteins in Mucorales is to mediate invasion of host cells, including epithelial and endothelial cells ( Gebremariam et al., 2014 ; Gebremariam et al., 2016 ; Alqarihi et al., 2020 ). CotH proteins were also found to be required for normal spore formation and virulence in M. lusitanicus ( Szebenyi et al., 2023 ). The number of CotH genes is correlated to the pathogenic potential of agents of MCR, with the genera most implicated in invasive infections ( Rhizopus, Mucor , and Lichtheimia ) ( Warkentien et al., 2012 ; Warkentien et al., 2015 ; Weintrob et al., 2015 ) having multiple copies of the CotH ( Chibucos et al., 2016 ). CotH3 and CotH7 (called collectively “invasins”) promote invasion of nasal and alveolar epithelial cells through binding to host cell GRP78 and integrin α3β1, respectively ( Alqarihi et al., 2020 ). The mechanism by which CotH7 binds to integrin α3β1 on alveolar epithelial cells initiates invasion of the host is believed to be related to activation of EGFR which initiates a cascade of events involved in endocytosis ( Watkins et al., 2018 ).

In addition to epithelia, CotH3 is also involved in promotion of angioinvasion through attaching to GRP78 on endothelial cells ( Gebremariam et al., 2014 ; Gebremariam et al., 2016 ). GRP78 is a heat-shock protein belonging to the HSP70 family that is expressed on the mammalian cell membrane in response to various stressors ( Lee, 2007 ; Alqarihi et al., 2020 ). Furthermore, serum iron increases in availability, when key host conditions were encountered in DKA such as hyperglycemia and the presence of ketone bodies (e.g., β-hydroxy butyrate), have all been shown to create a “perfect storm” of increased and rapid invasion and robustly increase the expression of both GRP78 and CotH3 in the target organs of mice with DKA ( Liu et al., 2010 ; Gebremariam et al., 2016 ; Alqarihi et al., 2020 ). Furthermore, the density of GRP78 receptors is richer in sites where MCR is most common. Thus, GRP78 high expression on invaded endothelial cells and macrophages in necrotic tissues were revealed through the immunohistochemistry of the ethmoidal sinus tissue of a patient with rhino-cerebral MCR ( Shumilov et al., 2018 ). Interestingly, R. delemar ’s capacity to invade and damage endothelial cells in vitro and reduce disease severity in mice is diminished by the activity of CotH3 proteins blocking their activity, either by using anti-CotH monoclonal antibodies or by genetically attenuating CotH3 expression ( Gebremariam et al., 2016 ). As a result, the distinctive susceptibility of patients with DKA to MCR is explained by the unique and concomitant amplification of interaction between CotH3 and GRP78 under hyperglycemic/ketoacedotic settings.

Of critical importance is the ability of an anti-CotH3 monoclonal antibody to block host cell invasion and ameliorate the disease in mice when given alone after infection. Further, this antibody demonstrates synergy when given with antifungal drugs to treat severe murine MCR ( Gebremariam et al., 2019 ). A humanized version of the antibody was also shown to equally protect against the disease in mice and is currently in manufacturing ( Gu et al., 2021 ).

The first evidence of the presence of toxins in Mucorales came from the observation that even killed Mucorales spores were able to cause significant damage to host cells ( Ibrahim et al., 2005b ). This was followed by a study connecting food poisoning outbreak with Chobani yogurt to M. circinelloides ( Lee et al., 2014 ). Recently, Soliman et al. revealed that Mucorales harbor a ricin-like toxin protein of 17 kDa that is expressed during hyphal formation. The toxin has structural similarity to ricin B chain and functionally resembled ricin A chain in blocking host protein synthesis through ribosomal inactivation. Thus, this toxin was named “mucoricin”, and was shown to be critical for MCR pathogenesis in mice. In addition, mucoricin was found to be expressed in lung tissues from a patient with pulmonary MCR ( Soliman et al., 2021 ). Importantly, polyclonal antibodies targeting mucoricin were shown to protect mice from MCR ( Soliman et al., 2021 ) suggesting that further development of immunotherapies against the toxin are likely to aid in managing patients with MCR. Other unidentified toxins are likely to exist, since attenuation of mucoricin expression reduced, but not abrogated, the ability of Rhizopus to damage host cells ( Soliman et al., 2021 ). Figure 3 summarizes pathogenicity events involved in host cell invasion and tissue damage during MCR.

www.frontiersin.org

Figure 3 Postulated events that lead to adhesion and invasion and host cell death. 1) Inhaled spores bind to epithelial via CotH/integrin a3β1 followed by germination. Germlings produce mucoricin. 2) Considerable disruption of the epithelium due to invasion of the spores within hours of infection. 3) Mucoricin causes host cell death within 48 h of infection. 4) Angioinvasion of hyphae and sporulated cells occur via CotH and endothelial cell GRP78 interactions, 5) endothelial cell injury occurs after infection with R. delemar spores resulting in tissue necrosis. 6) Hematogenous dissemination results in organ seeding. Tissue edema and organ failure are the results of excessive vascular leak. “Created with BioRender.com ”.

Genomic structure and genetic manipulation to understand pathogenicity

The availability of restricted tools for genetic manipulation has been a major hindrance to the in-depth research of Mucorales fungus genes and signaling cascades. Early in the Mucormycotina lineage, a whole-genome duplication occurred, and the duplication of genes may have produced novel proteins, so expanding the sensory and signaling pathways ( Ma et al., 2009 ; Schwartze et al., 2014 ; Garcia et al., 2018 ). When sexually reproducing, Mucorales are known to be haploid and display zygotic meiosis ( Morin-Sardin et al., 2017 ). Mucor , and Rhizopus , and Lichtheimia species are the only Mucorales known so far to be amenable to genetic manipulation. Due to the paucity of dominant selection markers, genetic experimentation even with these two fungi is difficult, the limited transportation efficiency, and the rarity of chromosomal integration. RNA interference (RNAi) is the most often used approach as a result, rather than disrupting genes ( Ibrahim et al., 2010 ; Calo et al., 2014 ; Liu et al., 2015 ; Trieu et al., 2017 ). Genes involved in virulence and resistance to treatment have been identified by researchers through gene silencing. However, a major drawback of the RNAi includes the possibility of having false positive outcomes due to off-target effects ( Schmitt, 2012 ). In Mucor , gene deletion mutants are achieved by targeted integration using either dominant selection or auxotrophic markers ( Appel et al., 2004 ; Larsen et al., 2004 ; Nicolas-Molina et al., 2008 ).

Vital information on the involvement of the calcineurin pathway in MCR pathogenesis has been yielded through gene disruption by homologous recombination effectively applied in M. circineloides ( Lee et al., 2013 ; Lee et al., 2015 ). The CRISPR/Cas9 system for gene editing of genomic DNA is the most recent advancement in molecular tool development. In order to disrupt a toxin-encoding gene in R. delemar utilizing a single plasmid with pyrF as a marker and the biolistic delivery system this approach was first used in Mucorales ( Baldin et al., 2017 ). Southern blot analysis, abrogation of toxin expression, and a dramatic reduction in R. delemar’s ability to kill host cells all verified gene disruption ( Baldin et al., 2017 ). Consistent with CRISPR-Cas9- induced gene mutation by non-homologous end joining (NHEJ) the CRISPR/Cas9 has been utilized to create R. delemar pyrF mutants with a single nucleotide deletion at the fourth nucleotide before the protospacer adjacent motif (PAM) sequence ( Bruni et al., 2019 ). The CRISPR-Cas9 targeted integration method was also recently adapted to reliably and stably transform protoplasts of R. microsporus ( Lax et al., 2021 ; Lax et al., 2022 ). In M. lusitanicus , the CRISPR-Cas9 system has been successfully employed in a plasmid-free way to disrupt two genes: carB, which encodes phytoene dehydrogenase, and hmgR2, which encodes 3-hydroxy-3-methylglutaryl-CoA reductase ( Nagy et al., 2017 ). More recently, uracil auxotrohic strains of Lichtheimia corymbifera were obtained by targeted mutagenesis using CRISPR-Cas9 ( Ibragimova et al. 2020 ) These tools show promise in deciphering the role of various regulatory genes during host-pathogen interactions.

Early diagnosis of MCR remains a major unmet need as clinical & radiologic predictors lack sensitivity and specificity ( Farmakiotis and Kontoyiannis, 2016 ). Cultures are often negative in tissues (up to 55-75% ( Tarrand et al., 2005 ) as is often immunohistochemistry (IHC)/ in situ hybridization ( Lockhart et al., 2021 ). There are limited promising new antifungals with Mucorales activity ( Lamoth et al., 2022 ). Thinking of the disease and having a low threshold of initiating therapy that has activity against Mucorales is very important. In patients with hematological malignancies, delaying amphotericin B-based therapy beyond 5 days after onset of symptoms doubles 12-week mortality ( Chamilos et al., 2008 ). Early detection of MCR is critical for timely treatment implementation as a result ( Spellberg et al., 2012c ). The traditional diagnosis of MCR relies on culturing the organism from normally sterile body locations and/or tissue histology because there is currently no serology test for diagnosis of MCR ( Spellberg et al., 2005a ; Ibrahim et al., 2012 ; Cornely et al., 2019 ). Fungal elements are usually stained with Gomori methenamine-silver, hematoxylin and eosin (H&E), periodic acid-Schiff (PAS), or calcofluor white stain and Mucorales can be isolated on Sabouraud-dextrose agar incubated at 25–37 °C ( Lass-Flörl, 2009 ). However, these procedures are insufficiently sensitive and frequently result in a misdiagnosis due to: 1) possible contamination of the plates, given the widespread nature of Mucorales fungi, cultures can result in false positives; and 2) lack of growth as a result of laboratory mishandling of specimens ( e.g. homogenization can damage hyphal components and kill the fungus) resulting in false negatives ( Spellberg and Ibrahim, 2015 ). The paradox of the poor recovery of Zygomycetes hyphae from tissue specimens remains unclear, and it may result from failure of current culture methods to mimic physiologic conditions found in hyphae-laden infected. Experimental evidence suggests that simulating Mucorales growth under necrotic or semi-anaerobic tissue conditions enhances culture yield ( Kontoyiannis et al., 2007 ). Even the “gold standard” that of histopathology detection of the characteristic nature of the board ribbon-like aseptate Mucorales hyphae is far from perfect. Minimal influence on the outcome of therapy can be achieved with definitive histologic identification based on morphology which can lead to error and often occur at a late stage of the infection ( Dadwal and Kontoyiannis, 2018 ). Radiological clues, in the appropriate clinical context, make the early suspicion of MCR possible in patient with hematologic cancer. The illness may be distinguished from invasive pulmonary aspergillosis through the radiological clues in chest CT along with the presence of a reversed halo sign in patients with a hematologic malignancy or neutropenia that have a strong predictive value for MCR ( Chamilos et al., 2005 ; Spellberg et al., 2005a ; Georgiadou et al., 2011 ; Legouge et al., 2014 ; Jung et al., 2015 ). Thus, the development of polymerase chain reaction (PCR)-based technologies and Matrix Assisted Laser Desorption/Ionization-Time of Flight Mass Spectrometry (MALDI-TOF MS) which are recent advancements in molecular diagnostics, have generated significant excitement the potential to speed up both the diagnosis and treatment of MCR (see below).

Molecular diagnostics

A potential useful tool for diagnosing MCR are real-time PCR-based approaches, particularly in lung infections caused by Lichtheimia, Mucor, Rhizopus , and Rhizomucor spp ( McCarthy and Walsh, 2016 ; McCarthy et al., 2017 ). PCR based diagnosis is particularly promising, in view of the early and rapid dissemination of MCR (in contrast to Aspergillus) The PCR amplification of CotH3, a Mucorales-specific protein, has demonstrated to be specific and sensitive for MCR diagnosis ( Baldin et al., 2018 ). CotH3 was not effectively amplified from urine, plasma, and bronchoalveolar lavage collected from mice infected with Aspergillus fumigatus but was effectively amplified in Mucorales fungi-infected mice ( Baldin et al., 2018 ). To identify DNA from the most common MCR agents a multiplex real-time PCR (MRT-PCR) technology has also been created ( Nagao et al., 2005 ; Walsh, 2012 ). Several studies showed significant inter-laboratory standardization and several supportive retrospective studies and preclinical data correlating MCR burden with qPCR kinetics ( Millon et al., 2013 ; Caillot et al., 2016 ; Millon et al., 2016 ; Springer et al., 2016 ). The nucleotide sequence of the ITS1 ribosomal DNA region from strains belonging to R. oryzae and R. microsporus , as well as the sequence of the ITS2 region for Mucor spp. belonging to M. circinelloides, M. racemosus, M. plumbeus, and M. velutinosus ( Bernal-Martinez et al., 2013 ), were used to design primers and molecular probes ( Springer et al., 2016 ). In tissue and serum samples from patients with rhinoorbital/cerebral MCR, a semi-nested PCR-based technique amplifying the 18S region of rDNA unique to Mucorales was shown to be more reliable than ITS2 PCR in identifying infection ( Zaman et al., 2017 ). For circulating Mucorales identification in patients with proven MCR, other quantitative PCR approaches have investigated a mix of hydrolysis probes targeting Mucor, Rhizopus , Lichtheimia , and Rhizomucor ( Millon et al., 2013 ). In 9 out of 10 blood samples from individuals diagnosed with the condition Mucorales DNA was found, which indicates that quantitative PCR might be a valuable screening technique in high-risk patients ( Caillot et al., 2016 ). These data have been further validated in a recent multicenter Study (MODIMUCOR study) ( Millon et al., 2022 ). Mucorales have been identified using other promising technologies such as MALDI-TOF MS ( Yaman et al., 2012 ). However, MALDI-TOF MS has yet to be proven as a reliable method for detecting MCR in clinical samples. As a diagnostic tool the requirement for fungus culture prior to identification, as well as the lack of substantial libraries for uncommon Mucorales has been restricted ( Cornely et al., 2019 ). Other emerging technologies are multiplex pan mold PCR ( Alanio and Bretagne, 2017 ), and a sequence-based identification/shot gun metagenomics ( Hoang et al., 2022 ).

Treatment of MCR

Reversal of underlying poor prognostic factors such as neutropenia, hyperglycemia, low threshold of suspicion and early initiation of effective antifungal therapy and in selected cases, surgical debridement of affected tissues, and antifungal therapy, are the cornerstones in the management of MCR ( Kontoyiannis and Lewis, 2011 ; Chitasombat and Kontoyiannis, 2016 ).

Surgical intervention

To resect all necrotic regions, surgical debridement should be carried out as soon as feasible and should be thorough. In cases of rhino-orbital/cerebral infection surgeries sometimes deformity can result ( Vironneau et al., 2014 ). Better outcomes have been associated with extensive and repeat surgical debridement of the rhino-orbital/cerebral MCR which have been shown by many uncontrolled investigations ( Gil-Lamaignere et al., 2005 ; Chamilos et al., 2010 ; Potenza et al., 2011 ). After radical surgery, 90% of patients achieved local infection control, compared to 41.6% in patients who underwent less extensive surgery ( Vironneau et al., 2014 ). However, in modern times, such disfiguring surgeries are much less common in the era of early rhinoscopic evaluation of the sinuses ( Davoudi et al., 2015 ). In pulmonary MCR, although patient selection also plays a role (patients with terminal underlying disease are typically excluded from surgery), studies have shown that patients treated with a combined medical-surgical approach had a better outcome than patients who did not undergo surgery ( Lee et al., 1999 ).

Treatment with antifungal drugs

In vitro data indicate that a limited number of FDA-approved antifungals (amphotericin B-based formulations, and the triazoles posaconazole. isavuconazole and possibly itraconazole) have activity against Mucorales and that there are species-specific differences in susceptibility to azoles (e.g., high posaconazole minimum inhibitory concentrations [MICs] in some Mucor species, high MIC to isavuconazole in Rhizomucor , multidrug resistance [MDR] in Cunninghamella and in some Rhizopus spp.) ( Almyroudis et al., 2007 ; Lamoth and Kontoyiannis, 2019 ; Borman et al., 2021 ). However, despite the useful information of in vitro studies the in vitro/in vivo correlation in the management of complicated opportunistic mold infections in humans such is MCR remains problematic ( Lamoth et al., 2020 ).

Most of the treatment experience is derived from amphotericin B-based therapy. The therapy of choice has become Lipid amphotericin B formulations ( e.g. liposomal amphotericin B [L-AMB], and amphotericin B lipid complex [ABLC]) since they can be given in higher doses than amphotericin B-deoxycholate and have improved nephrotoxicity index ( Fera et al., 2009 ; Vehreschild et al., 2013 ; Cornely et al., 2019 ). As a “step-down” treatment following the primary therapy with L-AMB, the broad-spectrum oral triazoles of posaconazole and isavuconazole can be used ( Kontoyiannis and Lewis, 2011 ; Allen et al., 2015 ; Marty et al., 2016 ). There are no randomized clinical trials evaluating the effectiveness of antifungal medications since MCR is an uncommon condition, affecting many host groups, presenting with different clinical syndromes, and caused by a variety of Mucorales ( Figure 1 ). However, a multicenter open-label single-arm research (VITAL study) including 37 patients with MCR found that isavuconazole monotherapy is as effective as L-AMB or L-AMB plus posaconazole ( Marty et al., 2016 ). In fact, clinically relevant dosages of isavuconazole, which is licensed by the FDA and the European Medicines Agency for the treatment of MCR patients were equated to tissue clearance and survival in mice ( Gebremariam et al., 2020 ). Figure 4 introduces an algorithm for MCR treatment.

www.frontiersin.org

Figure 4 An Algorithm for Mucormycosis Treatment.

The poor outcomes of MCR with currently available monotherapy, particularly in patients with hematologic malignancies, has stimulated interest in studying various combinations of antifungal agents ( Spellberg et al., 2012b ). In contrast to L-AMB + posaconazole combination ( Ibrahim et al., 2009 ), comparative effectiveness of isavuconazole + L-AMB in treating diabetic mice, showed synergy in treating mice infected with either R. delemar or M. circinelloides ( Gebremariam et al., 2021 ). Given the lack of easy extrapolation from experimental models to the complexity of human MCR, the synergy between L-AMB and isavuconazole therapy is yet to be determined in clinical trials.

Despite harboring the echinocandin-target enzyme glucan synthase (FKS) needed for 1,3-β-glucan production ( Ma et al. 2009 ), Mucorales species show innate in vitro resistance to echinocandins because glucans are not a major component of the Mucorales cell wall ( Ibrahim et al., 2005a ). However, a synergistic relationship between echinocandins and lipid formulation amphotericin B was observed in DKA mice infected with Rhizopus showing enhanced survival when compared to monotherapy ( Spellberg et al., 2005b ; Ibrahim et al., 2008a ). These experimental studies are consistent with data obtained from a retrospective study employing limited 41 diabetic patients with rhino-orbital MCR. Specifically, patients treated with a combination of caspofungin and amphotericin B-based drugs had better survival than those treated with amphotericin B-based drugs alone ( Reed et al., 2008 ). It is thought that immune detention and phagocytosis of invading hyphae is allowed when immunological epitopes on Mucorales cell wall, which are unmasked by echinocandins inhibition of β- glucan synthesis ( Reed et al., 2008 ). However, in MCR patients with hematologic malignancies and hematopoietic cell transplant recipients, a retrospective cohort study using propensity score analysis found that the combination L-AMB with posaconazole (suspension) or with echinocandins, or posaconazole with echinocandins resulted in no differences in 6-week mortality between monotherapy and combination treatment ( Kyvernitakis et al., 2016 ). It was revealed that L-AMB + micafungin treatment provided little enhancement in survival of neutropenic mice versus L-AMB monotherapy which are in line with data obtained in neutropenic patients ( Ibrahim et al., 2008a ). Overall, a combination of lipid formulation amphotericin B and echinocandins have been shown by the human retrospective and murine experimental results to help patients with DKA than those with hematologic malignancies or hematopoietic cell transplant recipients. It would be of interest to revisit the merits of combination therapy, with other conventional or investigational antifungals.

Antifungal drugs in development

Some investigational drugs are currently in development with demonstrated in vitro activity against Mucorales and in experimental models of MCR. In a delayed treatment model of immunosuppressed mice infected with R. arrhizus var. arrhizus , the 1-tetrazole fungal-specific 14 a-lanosterol demethylase (CYP51) inhibitor VT-1161 demonstrated equivalent effectiveness to high dosage L-AMB ( Gebremariam et al., 2015 ). When administered as prophylaxis, both VT-1161 and posaconazole enhanced lifespan and reduced tissue fungal load in immunosuppressed mice infected with R. arrhizus var. arrhizus . Furthermore, VT-1161 was superior to posaconazole in terms of extending mice survival time when used as a continuous treatment ( Gebremariam et al., 2015 ). Future clinical studies are needed to evaluate the therapeutic impact of tetrazoles in human MCR.

Manogepix (formerly APX001A and E1210) is a first-in-class antifungal drug with good activity against several fungal pathogens including Mucorales fungi ( Shaw and Ibrahim, 2020 ; Lamoth et al., 2022 ). The fungal Gwt1 enzyme which catalyzed inositol acylation is inhibited by manogepix, an early step in the glycosylphosphatidylinositol (GPI)-anchor biosynthesis pathway ( Umemura et al., 2003 ). Treatment with fosmanogepix (the prodrug of manogepix) substantially enhanced and prolonged median survival time of mice infected with either R. arrihzus var. arrhizus or R. arrihzus var. R. delemar , when compared to placebo. Furthermore, a 1-2 log decrease in both lung and kidney fungal loads resulted from fosmanogepix treatment ( Gebremariam et al., 2020 ). Further, a combination of fosmanogepix and L-AMB was found to be superior to monotherapy in treating immunosuppressed mice infected with R. arrihzus var. R. delemar with enhanced survival, tissue fungal clearance and histology improvement of infected lungs ( Gebremariam et al., 2022 ). Beyond mice studies, no clinical data exist regarding the efficacy of manogepix, given alone or in combination for primary or salvage therapy of MCR. Recently, it was shown that Rhizopus hyphae killing can be enhanced by delivering sub-micromolar concentrations of amphotericin B through liposomes targeted to the fungal hyphae by the inclusion of dectin-1 receptor which binds to fungal β-glucans ( Choudhury et al., 2022 ). These exciting results of enhancing the amphotericin B therapeutic index with lower and less toxic concentration are yet to be verified in animal models or clinical trials.

Adjunctive therapies

Adjunctive therapies are crucial in managing mucormycosis. A promising adjunctive therapy is iron chelation, specifically with deferasirox, which has demonstrated potential in inhibiting the growth of Mucorales fungi by reducing the availability of iron, an essential nutrient for their proliferation ( Ibrahim, 2006 ). Additionally, immune modulation therapies such as granulocyte transfusions and cytokine therapies are being studied to enhance the host’s immune response against Mucorales infections ( Lanternier et al., 2015 ). While these adjunctive therapies show promise, further research and clinical trials are necessary to determine their optimal use and effectiveness. In individuals with DKA suspected of having MCR, the restoration of the host’s ability to chelate iron resulted in enhanced activity of neutrophils in killing Mucorales ex vivo ( Gebremariam et al., 2016 ). This was achieved by reversing acidemia (acidosis) with sodium bicarbonate. Further, the administration of sodium bicarbonate partially prevented the capacity of R. delemar to invade endothelial cells ( Gebremariam et al., 2016 ). Moreover, in a mouse model of ketoacidosis, treatment with sodium bicarbonate protected against invasive lung infection ( Gebremariam et al., 2016 ). These findings suggest that individuals with DKA suspected of having Mucorales infection may benefit from the rapid correction of hyperglycemia and acidemia using insulin and sodium bicarbonate, respectively, to improve the host’s defense mechanisms ( Gebremariam et al., 2016 ).

Restriction of available serum iron with new generation of xenosiderophores, inhibits fungal growth and protects DKA mice against MCR ( Ibrahim, 2006 ; Ibrahim et al., 2007 ). In patients with DM it is suggested in case reports that it is beneficial to use iron chelation therapy as an adjunctive treatment ( Spellberg, 2009 ). Adding deferasirox to L-AMB treatment was found to be harmful primarily in patients with hematologic malignancies in a small (20 patient) multi-center, placebo controlled, double-blind study (DEFEAT Mucor) ( Spellberg et al., 2012a ). The data do not support the use of deferasirox as an initial supplementary therapy for MCR in hematologic malignancies patients although the population imbalances in this small phase II study make generalizable inferences problematic. These findings are not surprising since hematologic malignancies patients generally do not suffer from iron overload due to acidosis or hyperglycemia.

The use of hyperbaric oxygen (HBO) is another therapy that is likely to be useful in conjunction with surgery and antifungal therapy. HBO therapy raises blood oxygen levels and boosts neutrophil activity ( Lerche et al., 2022 ). Adjunctive HBO proved promising in diabetic patients (94% survival), but not in those with hematologic malignancies or bone marrow transplants (33% survival; p 0.02) ( Price and Stevens, 1980 ; Ferguson et al., 1988 ; Kajs-Wyllie, 1995 ; Barratt et al., 2001 ; John et al., 2005 ). A better rate of survival was linked to prolonged courses of HBO ( John et al., 2005 ).

Based on limited in vitro data and anecdotal case reports, strategies that boost the immune system, such as the administration of granulocyte (macrophage) colony stimulating factor or interferon-γ, or possibly check point inhibitors or their combination have been advocated as adjuvant therapy ( Abzug and Walsh, 2004 ; Gil-Lamaignere et al., 2005 ). A combination of Interferon-γ with nivolumab (a monoclonal antibody that reduces programmed death-1 [PD-1] expression on T-cells) was found to be effective in an immunocompromised patient with intractable MCR in a recent case report ( Grimaldi et al., 2017 ).

MCR in the era of COVID-19

COVID-19-associated (CAM) MCR has recently emerged as an important superinfection among COVID-19 patients with documented cases from various regions of the world, and most notably in India ( Raut and Huy, 2021 ; Ravani et al., 2021 ). Between May and August of 2021, more than 47,000 cases were reported, among mainly diabetic patients suffering from COVID-19 infection in India alone forcing the government to declare MCR as an epidemic. Whether COVID-19 infection by itself predisposes patients to MCR is not clear. Both in India and the rest of the world, the vast majority of excess cases of MCR during the COVID-19 pandemic have likely been attributable to a combination of DM and corticosteroid ( John et al., 2021 ; Raut and Huy, 2021 ; Ravani et al., 2021 ; Singh et al., 2021 ). Almost 1/3 of the recent reported Indian MCR cases were among non-COVID-19 infected patients ( Patel et al., 2021 ; Ravani et al., 2021 ), thereby underscoring the high baseline rate of infection in this country which was previously estimated to be 70-fold higher than any other part of the world ( Patel et al., 2021 ). Furthermore, the large majority of MCR cases in COVID-19 patients in India have been of the rhino-orbital-cerebral (ROC) type ( Ravani et al., 2021 ; Singh et al., 2021 ), and pulmonary infection has been rare. The pathogenesis of CAM remains an enigma. It is possible that COVID-19 predispose patients to newly onset DM or the ones with preexisting DM to experience worsening of glycemic control or full blown DKA, as SARS CoV-2 infection is associated with high expression of angiotensin-converting enzyme 2 (ACE2) receptor in pancreatic islets (potentially destroying these cells), along with increased insulin resistance due to cytokine storm ( Kothandaraman et al., 2021 ). Interestingly, high expression of GRP78 in COVID-19 patients has been reported, possibly as a result of the viral-induced endoplasmic reticulum stress cascade ( Sabirli et al., 2021 ). It was also recently shown that GRP78 forms a complex with ACE2 to act as an auxiliary receptor to the SARS COV-2 ( Carlos et al., 2021 ). Thus, with GRP78 being a receptor to Mucorales fungi ( Gebremariam et al., 2014 ; Gebremariam et al., 2016 ; Alqarihi et al., 2020 ), there is an increased probability that the presence of elevated GRP78 levels in COVID-19 patients specifically predispose to MCR.

Despite advances in risk stratification, dissection of pathogenesis of the disease, imaging and increasingly the introduction of non-culture-based diagnostics, MCR continues to be associated with high rates of death and disability. Further improvements in molecular diagnostics and the establishment of large patient registries are key components of ongoing efforts. We believe that disease outcomes will further improve by the combination of much earlier diagnostics (surveillance vs. adjunct diagnostics), immuno-restoration/immunotherapeutic strategies, and the introduction of potent new antifungals. Further investments on developing pathophysiologically appropriate and phylogenetically disparate model systems of MCR ( e.g. flies ( Shirazi et al., 2014 ), Galleria mellonella ( Maurer et al., 2019 ), zebra fish ( Wurster et al., 2021 ) and mice ( Jacobsen, 2019 ; Stevens et al., 2020 )), along with advances in the molecular toolbox systems would further shed lights on the complex pathophysiology of this important disease.

Author contributions

AA: Conceptualization, Writing – original draft, Writing – review & editing. DK: Writing – original draft, Writing – review & editing, Conceptualization, Funding acquisition, Supervision. AI: Conceptualization, Funding acquisition, Investigation, Supervision, Writing – original draft, Writing – review & editing

This study was supported by Public Health Service grant R01 AI063503 to A.S.I and Robert C Hickey endowment to DPK.

Conflict of interest

AI has received research support from and served on advisory boards for Amplyx, Astellas, Cidara and Navigen. AI owns shares in Vitalex Biosciences, a start-up company that is developing immunotherapies and diagnostics for MCR. DK reports honoraria and research support from Gilead Sciences and Astellas, Inc, received consultant fees from Astellas Pharma, Merck, and Gilead Sciences, and is a member of the Data Review Committee of Cidara Therapeutics, AbbVie, and the Mycoses Study Group.

The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Aberdein, J. D., Cole, J., Bewley, M. A., Marriott, H. M., Dockrell, D. H. (2013). Alveolar macrophages in pulmonary host defence the unrecognized role of apoptosis as a mechanism of intracellular bacterial killing. Clin. Exp. Immunol. 174 (2), 193–202. doi: 10.1111/cei.12170

PubMed Abstract | CrossRef Full Text | Google Scholar

Abzug, M. J., Walsh, T. J. (2004). Interferon-gamma and colony-stimulating factors as adjuvant therapy for refractory fungal infections in children. Pediatr. Infect. Dis. J. 23 (8), 769–773. doi: 10.1097/01.inf.0000134314.65398.bf

Alanio, A., Bretagne, S. (2017). Performance evaluation of multiplex PCR including Aspergillus -not so simple! Med. Mycol. 55 (1), 56–62. doi: 10.1093/mmy/myw080

Allen, D., Wilson, D., Drew, R., Perfect, J. (2015). Azole antifungals: 35 years of invasive fungal infection management. Expert Rev. Anti Infect. Ther. 13 (6), 787–798. doi: 10.1586/14787210.2015.1032939

Almyroudis, N. G., Sutton, D. A., Fothergill, A. W., Rinaldi, M. G., Kusne, S. (2007). In vitro susceptibilities of 217 clinical isolates of zygomycetes to conventional and new antifungal agents. Antimicrob. Agents Chemother. 51 (7), 2587–2590. doi: 10.1128/AAC.00452-07

Alqarihi, A., Gebremariam, T., Gu, Y., Swidergall, M., Alkhazraji, S., Soliman, S. S. M., et al. (2020). GRP78 and integrins play different roles in host cell invasion during mucormycosis. mBio 11 (3), e01087–e01020. doi: 10.1128/mBio.01087-20

Ambrosioni, J., Bouchuiguir-Wafa, K., Garbino, J. (2010). Emerging invasive zygomycosis in a tertiary care center: epidemiology and associated risk factors. Int. J. Infect. Dis. 14, e100–e1e3. doi: 10.1016/j.ijid.2009.11.024

Andrianaki, A. M., Kyrmizi, I., Thanopoulou, K., Baldin, C., Drakos, E., Soliman, S. S. M., et al. (2018). Iron restriction inside macrophages regulates pulmonary host defense against Rhizopus species. Nat. Commun. 9 (1), 3333. doi: 10.1038/s41467-018-05820-2

Appel, K. F., Wolff, A. M., Arnau, J. (2004). A multicopy vector system for genetic studies in Mucor circinelloides and other zygomycetes. Mol. Genet. Genomics 271 (5), 595–602. doi: 10.1007/s00438-004-1008-6

Artis, W. M., Fountain, J. A., Delcher, H. K., Jones, H. E. (1982). A mechanism of susceptibility to mucormycosis in diabetic ketoacidosis: transferrin and iron availability. Diabetes 31 (12), 1109–1114. doi: 10.2337/diacare.31.12.1109

Available at: https://www.cdc.gov/fungal/infections/hospitalized.html .

Google Scholar

Baldin, C., Soliman, S. S. M., Jeon, H. H., Alkhazraji, S., Gebremariam, T., Gu, Y., et al. (2018). PCR-based approach targeting Mucorales specific gene family for the diagnosis of mucormycosis. J. Clin. Microbiol . 56, e00746–18. doi: 10.1128/JCM.00746-18

Baldin, C., Soliman, S., Jeon, H. H., Gebremariam, T., Skory, C. D., Edwards, J. E. J., et al. (2017). editors. Optimization of the CRISPR/Cas9 system to manipulate gene function in Rhizopus delemar . Open Forum Infect. Dis. 4 (Suppl 1), S116.

Barratt, D. M., Van Meter, K., Asmar, P., Nolan, T., Trahan, C., Garcia-Covarrubias, L., et al. (2001). Hyperbaric oxygen as an adjunct in zygomycosis: randomized controlled trial in a murine model. Antimicrob. Agents Chemother. 45 (12), 3601–3602. doi: 10.1128/AAC.45.12.3601-3602.2001

Ben-Ami, R., Luna, M., Lewis, R. E., Walsh, T. J., Kontoyiannis, D. P. (2009). A clinicopathological study of pulmonary mucormycosis in cancer patients: extensive angioinvasion but limited inflammatory response. J. Infect. 59 (2), 134–138. doi: 10.1016/j.jinf.2009.06.002

Bernal-Martinez, L., Buitrago, M. J., Castelli, M. V., Rodriguez-Tudela, J. L., Cuenca-Estrella, M. (2013). Development of a single tube multiplex real-time PCR to detect the most clinically relevant Mucormycetes species. Clin. Microbiol. Infect. 19 (1), E1–E7. doi: 10.1111/j.1469-0691.2012.03976.x

Boelaert, J. R. (1994). Mucormycosis (zygomycosis): is there news for the clinician? J. Infect. 28 Suppl 1 (5 Pt 2), 1–6. doi: 10.1016/S0163-4453(94)95896-3

Boelaert, J. R., de Locht, M., Van Cutsem, J., Kerrels, V., Cantinieaux, B., Verdonck, A., et al. (1993). Mucormycosis during deferoxamine therapy is a siderophore-mediated infection. In vitro and in vivo animal studies. J. Clin. Invest. 91 (5), 1979–1986. doi: 10.1172/JCI116419

Boelaert, J. R., Fenves, A. Z., Coburn, J. W. (1989). Registry on mucormycosis in dialysis patients [letter]. J. Infect. Dis. 160 (5), 914. doi: 10.1093/infdis/160.5.914

Boelaert, J. R., Fenves, A. Z., Coburn, J. W. (1991). Deferoxamine therapy and mucormycosis in dialysis patients: report of an international registry. Am. J. Kidney Dis. 18 (6), 660–667. doi: 10.1016/S0272-6386(12)80606-8

Boelaert, J. R., Van Cutsem, J., de Locht, M., Schneider, Y. J., Crichton, R. R. (1994). Deferoxamine augments growth and pathogenicity of Rhizopus , while hydroxypyridinone chelators have no effect. Kidney Int. 45 (3), 667–671. doi: 10.1038/ki.1994.89

Boelaert, J. R., Vergauwe, P. L., Vandepitte, J. M. (1987). Mucormycosis infection in dialysis patients [letter]. Ann. Internal Med. 107 (5), 782–783. doi: 10.7326/0003-4819-107-5-782_2

CrossRef Full Text | Google Scholar

Borman, A. M., Fraser, M., Patterson, Z., Palmer, M. D., Johnson, E. M. (2021). In vitro antifungal drug resistance profiles of clinically relevant members of the mucorales (Mucoromycota) especially with the newer triazoles. J. Fungi (Basel). 7 (4), 271. doi: 10.3390/jof7040271

Bruni, G. O., Zhong, K., Lee, S. C., Wang, P. (2019). CRISPR-Cas9 induces point mutation in the mucormycosis fungus Rhizopus delemar. Fungal Genet. Biol. 124, 1–7. doi: 10.1016/j.fgb.2018.12.002

Caillot, D., Valot, S., Lafon, I., Basmaciyan, L., Chretien, M. L., Sautour, M., et al. (2016). Is it time to include CT “Reverse halo sign” and qPCR targeting mucorales in serum to EORTC-MSG criteria for the diagnosis of pulmonary mucormycosis in leukemia patients? Open Forum Infect. Dis. 3 (4), ofw190.

PubMed Abstract | Google Scholar

Calo, S., Shertz-Wall, C., Lee, S. C., Bastidas, R. J., Nicolás, F. E., Granek, J. A., et al. (2014). Antifungal drug resistance evoked via RNAi-dependent epimutations. Nature 513 (7519), 555–558. doi: 10.1038/nature13575

Cantinieaux, B., Janssens, A., Boelaert, J. R., Lejeune, M., Vermylen, C., Kerrels, V., et al. (1999). Ferritin-associated iron induces neutrophil dysfunction in hemosiderosis. J. Lab. Clin. Med. 133 (4), 353–361. doi: 10.1016/S0022-2143(99)90066-5

Carlos, A. J., Ha, D. P., Yeh, D.-W., Van Krieken, R., Tseng, C.-C., Zhang, P., et al. (2021). The chaperone GRP78 is a host auxiliary factor for SARS-CoV-2 and GRP78 depleting antibody blocks viral entry and infection. J. Biol. Chem. 296. doi: 10.1016/j.jbc.2021.100759

Carroll, C. S., Grieve, C. L., Murugathasan, I., Bennet, A. J., Czekster, C. M., Liu, H., et al. (2017). The rhizoferrin biosynthetic gene in the fungal pathogen Rhizopus delemar is a novel member of the NIS gene family. Int. J. Biochem. Cell Biol. 89, 136–146. doi: 10.1016/j.biocel.2017.06.005

Castillo, P., Wright, K. E., Kontoyiannis, D. P., Walsh, T., Patel, S., Chorvinsky, E., et al. (2018). A new method for reactivating and expanding T cells specific for Rhizopus oryzae. Mol. Ther. Methods Clin. Dev. 9, 305–312. doi: 10.1016/j.omtm.2018.03.003

Chakrabarti, A., Kaur, H., Savio, J., Rudramurthy, S. M., Patel, A., Shastri, P., et al. (2019). Epidemiology and clinical outcomes of invasive mould infections in Indian intensive care units (FISF study). J. Crit. Care 51, 64–70. doi: 10.1016/j.jcrc.2019.02.005

Chakrabarti, A., Singh, R. (2014). Mucormycosis in India: unique features. Mycoses 57 Suppl 3, 85–90. doi: 10.1111/myc.12243

Chamilos, G., Ganguly, D., Lande, R., Gregorio, J., Meller, S., Goldman, W. E., et al. (2010). Generation of IL-23 producing dendritic cells (DCs) by airborne fungi regulates fungal pathogenicity via the induction of T(H)-17 responses. PloS One 5 (9), e12955. doi: 10.1371/journal.pone.0012955

Chamilos, G., Lewis, R. E., Kontoyiannis, D. P. (2008). Delaying amphotericin B-based frontline therapy significantly increases mortality among patients with hematologic Malignancy who have zygomycosis. Clin. Infect. Dis. 47 (4), 503–509. doi: 10.1086/590004

Chamilos, G., Marom, E. M., Lewis, R. E., Lionakis, M. S., Kontoyiannis, D. P. (2005). Predictors of pulmonary zygomycosis versus invasive pulmonary aspergillosis in patients with cancer. Clin. Infect. Dis. 41 (1), 60–66. doi: 10.1086/430710

Chibucos, M. C., Soliman, S., Gebremariam, T., Lee, H., Daugherty, S., Orvis, J., et al. (2016). An integrated genomic and transcriptomic survey of mucormycosis-causing fungi. Nat. Commun. 7, 12218. doi: 10.1038/ncomms12218

Chinn, R. Y., Diamond, R. D. (1982). Generation of chemotactic factors by Rhizopus oryzae in the presence and absence of serum: relationship to hyphal damage mediated by human neutrophils and effects of hyperglycemia and ketoacidosis. Infect. Immunity 38 (3), 1123–1129. doi: 10.1128/iai.38.3.1123-1129.1982

Chitasombat, M. N., Kontoyiannis, D. P. (2016). Treatment of mucormycosis in transplant patients: role of surgery and of old and new antifungal agents. Curr. Opin. Infect. Dis. 29 (4), 340–345. doi: 10.1097/QCO.0000000000000277

Choudhury, Q. J., Ambati, S., Lewis, Z. A., Meagher, R. B. (2022). Targeted delivery of antifungal liposomes to Rhizopus delemar . J. Fungi (Basel) 8 (4), 352. doi: 3390/jof8040352

Cornely, O. A., Alastruey-Izquierdo, A., Arenz, D., Chen, S. C. A., Dannaoui, E., Hochhegger, B., et al. (2019). 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. 19 (12), e405–ee21. doi: 10.1016/S1473-3099(19)30312-3

Dadwal, S. S., Kontoyiannis, D. P. (2018). Recent advances in the molecular diagnosis of mucormycosis. Expert Rev. Mol. Diagnost. 18 (10), 845–854. doi: 10.1080/14737159.2018.1522250

Davoudi, S., Kumar, V. A., Jiang, Y., Kupferman, M., Kontoyiannis, D. P. (2015). Invasive mould sinusitis in patients with haematological Malignancies: a 10 year single-centre study. J. Antimicrob. Chemother. 70 (10), 2899–2905. doi: 10.1093/jac/dkv198

de Locht, M., Boelaert, J. R., Schneider, Y. J. (1994). Iron uptake from ferrioxamine and from ferrirhizoferrin by germinating spores of Rhizopus microsporus. Biochem. Pharmacol. 47 (10), 1843–1850. doi: 10.1016/0006-2952(94)90314-X

Diamond, R. D., Clark, R. A. (1982). Damage to Aspergillus fumigatus and Rhizopus oryzae hyphae by oxidative and nonoxidative microbicidal products of human neutrophils in vitro. Infect. Immun. 38 (2), 487–495. doi: 10.1128/iai.38.2.487-495.1982

Diamond, R. D., Erickson, N. F., 3rd. (1982). Chemotaxis of human neutrophils and monocytes induced by Cryptococcus neoformans. Infect. Immun. 38 (1), 380–382. doi: 10.1128/iai.38.1.380-382.1982

Dolatabadi, S., Ahmadi, B., Rezaei-Matehkolaei, A., Zarrinfar, H., Skiada, A., Mirhendi, H., et al. (2018). Mucormycosis in Iran: A six-year retrospective experience. J. Mycol. Médicale 28 (2), 269–273. doi: 10.1016/j.mycmed.2018.02.014

Farmakiotis, D., Kontoyiannis, D. P. (2016). Mucormycoses. Infect. Dis. Clin. North Am. 30 (1), 143–163. doi: 10.1016/j.idc.2015.10.011

Fera, M. T., La Camera, E., De Sarro, A. (2009). New triazoles and echinocandins: mode of action, in vitro activity and mechanisms of resistance. Expert Rev. Anti Infect. Ther. 7 (8), 981–998. doi: 10.1586/eri.09.67

Ferguson, B. J., Mitchell, T. G., Moon, R., Camporesi, E. M., Farmer, J. (1988). Adjunctive hyperbaric oxygen for treatment of rhinocerebral mucormycosis. Rev. Infect. Dis. 10 (3), 551–559. doi: 10.1093/clinids/10.3.551

Frąc, M., Hannula, S. E., Bełka, M., Jędryczka, M. (2018). Fungal biodiversity and their role in soil health. Front. Microbiol. 9. doi: 10.3389/fmicb.2018.00707

Fu, Y., Lee, H., Collins, M., Tsai, H. F., Spellberg, B., Edwards, J. E., Jr., et al. (2004). Cloning and functional characterization of the Rhizopus oryzae high affinity iron permease (rFTR1) gene. FEMS Microbiol. Lett. 235 (1), 169–176. doi: 10.1016/j.femsle.2004.04.031

Garcia, A., Vellanki, S., Lee, S. C. (2018). Genetic tools for investigating Mucorales fungal pathogenesis. Curr. Clin. Microbiol. Rep. 5 (3), 173–180. doi: 10.1007/s40588-018-0097-7

García-Carnero, L. C., Mora-Montes, H. M. (2022). Mucormycosis and COVID-19-associated mucormycosis: insights of a deadly but neglected mycosis. J. Fungi 8 (5), 445. doi: 10.3390/jof8050445

Gebremariam, T., Alkhazraji, S., Alqarihi, A., Wiederhold, N. P., Shaw, K. J., Patterson, T. F., et al (2020). Fosmanogepix (APX001) is effective in the treatment of pulmonary murine mucormycosis due to Rhizopus arrhizus . Antimicrob. Agents Chemother. 64, e00178–20. doi: 10.1128/AAC.00178-20

Gebremariam, T., Alkhazraji, S., Soliman, S. S. M., Gu, Y., Jeon, H. H., Zhang, L., et al. (2019). Anti-CotH3 antibodies protect mice from mucormycosis by prevention of invasion and augmenting opsonophagocytosis. Sci. Adv. 5 (6), eaaw1327. doi: 10.1126/sciadv.aaw1327

Gebremariam, T., Gu, Y., Alkhazraji, S., Youssef, E., Shaw, K. J., Ibrahim, A. S. (2022). The combination treatment of fosmanogepix and liposomal amphotericin B is superior to monotherapy in treating experimental invasive mold infections. Antimicrob. Agents Chemother. 66 (7), e0038022. doi: 10.1128/aac.00380-22

Gebremariam, T., Gu, Y., Singh, S., Kitt, T. M., Ibrahim, A. S. (2021). Combination treatment of liposomal amphotericin B and isavuconazole is synergistic in treating experimental mucormycosis. J. Antimicrob. Chemother. 76 (10), 2636–2639. doi: 10.1093/jac/dkab233

Gebremariam, T., Lin, L., Liu, M., Kontoyiannis, D. P., French, S., Edwards, J. E., Jr., et al. (2016). Bicarbonate correction of ketoacidosis alters host-pathogen interactions and alleviates mucormycosis. J. Clin. Invest . 126 (6), 2280–2294. doi: 10.1172/JCI82744

Gebremariam, T., Liu, M., Luo, G., Bruno, V., Phan, Q. T., Waring, A. J., et al. (2014). CotH3 mediates fungal invasion of host cells during mucormycosis. J. Clin. Invest. 124 (1), 237–250. doi: 10.1172/JCI71349

Gebremariam, T., Wiederhold, N. P., Fothergill, A. W., Garvey, E. P., Hoekstra, W. J., Schotzinger, R. J., et al. (2015). VT-1161 Protects Immunosuppressed Mice from Rhizopus arrhizus var. arrhizus Infection. Antimicrob. Agents Chemother. 59 (12), 7815–7817. doi: 10.1128/AAC.01437-15

Georgiadou, S. P., Sipsas, N. V., Marom, E. M., Kontoyiannis, D. P. (2011). The diagnostic value of halo and reversed halo signs for invasive mold infections in compromised hosts. Clin. Infect. Dis. 52 (9), 1144–1155. doi: 10.1093/cid/cir122

Gil-Lamaignere, C., Simitsopoulou, M., Roilides, E., Maloukou, A., Winn, R. M., Walsh, T. J. (2005). Interferon- gamma and Granulocyte-Macrophage Colony-Stimulating Factor Augment the Activity of Polymorphonuclear Leukocytes against Medically Important Zygomycetes. J. Infect. Dis. 191 (7), 1180–1187. doi: 10.1086/428503

Gomes, M. Z., Lewis, R. E., Kontoyiannis, D. P. (2011). Mucormycosis caused by unusual mucormycetes, non- Rhizopus, -Mucor, and -Lichtheimia species. Clin. Microbiol. Rev. 24 (2), 411–445. doi: 10.1128/CMR.00056-10

Grimaldi, D., Pradier, O., Hotchkiss, R. S., Vincent, J.-L. (2017). Nivolumab plus interferon-γ; in the treatment of intractable mucormycosis. Lancet Infect. Dis. 17 (1), 18. doi: 10.1016/S1473-3099(16)30541-2

Gu, Y., Alqarihi, A., Singh, S., Gebremariam, T., Alkhazraji, S., Youssef, E. G., et al. (2021). A humanized antibody targeting the CotH invasins is protective against murine mucormycosis. Open Forum Infect Dis . 8(Suppl 1):S71–2. doi: 10.1093/ofid/ofab466.119

Guo, D., Jaber, B. L., Lee, S., Perianayagam, M. C., King, A. J., Pereira, B. J., et al. (2002). Impact of iron dextran on polymorphonuclear cell function among hemodialysis patients. Clin. Nephrol. 58 (2), 134–142. doi: 10.5414/CNP58134

Hoang, M. T. V., Irinyi, L., Hu, Y., Schwessinger, B., Meyer, W. (2022). Long-reads-based metagenomics in clinical diagnosis with a special focus on fungal infections. Front. Microbiol. 12. doi: 10.3389/fmicb.2021.708550

Howard, D. H. (1999). Acquisition, transport, and storage of iron by pathogenic fungi. Clin. Microbiol. Rev. 12 (3), 394–404. doi: 10.1128/CMR.12.3.394

Ibrahim, A. S. (2006). Deferiprone iron chelation as a novel therapy for experimental mucormycosis. J. Antimicrob. Chemother. 58 (5), 1070–1073. doi: 10.1093/jac/dkl350

Ibrahim, A. S. (2010). The high affinity iron permease is a key virulence factor required for Rhizopus oryzae pathogenesis. Curr. Infect. Dis. Rep. 77 (3), 587–604. doi: 10.1111/j.1365-2958.2010.07234.x

Ibrahim, A. S. (2014). Host-iron assimilation: pathogenesis and novel therapies of mucormycosis. Mycoses 57 Suppl 3, 13–17. doi: 10.1111/myc.12232

Ibrahim, A. S., Bowman, J. C., Avanessian, V., Brown, K., Spellberg, B., Edwards, J. E., Jr., et al. (2005a). Caspofungin inhibits Rhizopus oryzae 1,3-beta-D-glucan synthase, lowers burden in brain measured by quantitative PCR, and improves survival at a low but not a high dose during murine disseminated zygomycosis. Antimicrob. Agents Chemother. 49 (2), 721–727. doi: 10.1128/AAC.49.2.721-727.2005

Ibrahim, A. S., Edwards, J. E., Filler, S. G., Spellberg, B. (2011). “Mucormycosis and entomophtoramycosis (Zygomycosis),” in Essentials of Clinical Mycology , 2nd Ed. Eds. Kauffman PP, C. A., Sobel, J. D., Dismukes, W. E. (New York: Springer), 265–280.

Ibragimova, A., Szebenyi, C., Sinka, R., Alzyoud, E. I., Homa, M., Vagvolgyi, C., Nagy, G., Papp, T.. (2020). CRISPR-Cas9-base mutagenesis of the mucormycosis-causing fungus Lichtheimia corymbifera. Int. J. Mol. Sci. 22, 3727. doi: 10.3390/ijms21103727

Ibrahim, A. S., Gebermariam, T., Fu, Y., Lin, L., Husseiny, M. I., French, S. W., et al. (2007). The iron chelator deferasirox protects mice from mucormycosis through iron starvation. J. Clin. Invest. 117 (9), 2649–2657. doi: 10.1172/JCI32338

Ibrahim, A. S., Gebremariam, T., Fu, Y., Edwards, J. E., Jr., Spellberg, B. (2008a). Combination echinocandin-polyene treatment of murine mucormycosis. Antimicrob. Agents Chemother. 52 (4), 1556–1558. doi: 10.1128/AAC.01458-07

Ibrahim, A. S., Gebremariam, T., Lin, L., Luo, G., Husseiny, M. I., Skory, C. D., et al. (2010). The high affinity iron permease is a key virulence factor required for Rhizopus oryzae pathogenesis. Mol. Microbiol. 77 (3), 587–604. doi: 10.1111/j.1365-2958.2010.07234.x

Ibrahim, A. S., Gebremariam, T., Schwartz, J. A., Edwards, J. E., Jr., Spellberg, B. (2009). Posaconazole mono- or combination therapy for treatment of murine zygomycosis. Antimicrob. Agents Chemother. 53 (2), 772–775. doi: 10.1128/AAC.01124-08

Ibrahim, A. S., Spellberg, B., Avanessian, V., Fu, Y., Edwards, J. E., Jr. (2005b). Rhizopus oryzae adheres to, is phagocytosed by, and damages endothelial cells in vitro. Infect. Immun. 73 (2), 778–783. doi: 10.1128/IAI.73.2.778-783.2005

Ibrahim, A. S., Spellberg, B., Edwards, J., Jr. (2008b). Iron acquisition: a novel perspective on mucormycosis pathogenesis and treatment. Curr. Opin. Infect. Dis. 21 (6), 620–625. doi: 10.1097/QCO.0b013e3283165fd1

Ibrahim, A. S., Spellberg, B., Walsh, T. J., Kontoyiannis, D. P. (2012). Pathogenesis of mucormycosis. Clin. Infect. Dis. 54 Suppl 1, S16–S22. doi: 10.1093/cid/cir865

Ibrahim, A. S., Voelz, K. (2017). The mucormycete–host interface. Curr. Opin. Microbiol. 40 (Supplement C), 40–45. doi: 10.1016/j.mib.2017.10.010

Jacobsen, I. D. (2019). Animal models to study mucormycosis. J. Fungi (Basel Switzerland) 5 (2), 27. doi: 10.3390/jof5020027

John, B. V., Chamilos, G., Kontoyiannis, D. P. (2005). Hyperbaric oxygen as an adjunctive treatment for zygomycosis. Clin. Microbiol. Infect. 11 (7), 515–517. doi: 10.1111/j.1469-0691.2005.01170.x

John, T. M., Jacob, C. N., Kontoyiannis, D. P. (2021). When uncontrolled diabetes mellitus and severe COVID-19 converge: the perfect storm for mucormycosis. J. Fungi (Basel). 7 (4), 298. doi: 10.3390/jof7040298

Jung, J., Kim, M. Y., Lee, H. J., Park, Y. S., Lee, S. O., Choi, S. H., et al. (2015). Comparison of computed tomographic findings in pulmonary mucormycosis and invasive pulmonary aspergillosis. Clin. Microbiol. Infect. 21 (7), 684 e11–8. doi: 10.1016/j.cmi.2015.03.019

Kajs-Wyllie, M. (1995). Hyperbaric oxygen therapy for rhinocerebral fungal infection. J. Neurosci. Nurs. 27 (3), 174–181. doi: 10.1097/01376517-199506000-00006

Kontoyiannis, D. P., Chamilos, G., Hassan, S. A., Lewis, R. E., Albert, N. D., Tarrand, J. J. (2007). Increased culture recovery of Zygomycetes under physiologic temperature conditions. Am. J. Clin. Pathol. 127 (2), 208–212. doi: 10.1309/7KU5XWURYM0151YN

Kontoyiannis, D. P., Lewis, R. E. (2011). How I treat mucormycosis. Blood 118 (5), 1216–1224. doi: 10.1182/blood-2011-03-316430

Kontoyiannis, D. P., Wessel, V. C., Bodey, G. P., Rolston, K. V. (2000). Zygomycosis in the 1990s in a tertiary-care cancer center. Clin. Infect. Dis. 30 (6), 851–856. doi: 10.1086/313803

Konotoyiannis, D. P., Lewis, R. E., Lortholary, O., Spellber, B., Petrikkos, G., Roilides, E., Ibrahim, A., Walsh, T. J. (2012). Future directions in mucormycosis research. Clin. Infect. Dis. 54 Suppl 1, S79–S85. doi: 10.1093/cid/cir886

Kothandaraman, N., Rengaraj, A., Xue, B., Yew, W. S., Velan, S. S., Karnani, N., et al. (2021). COVID-19 endocrinopathy with hindsight from SARS. Am. J. Physiol. Endocrinol. Metab. 320 (1), E139–Ee50. doi: 10.1152/ajpendo.00480.2020

Kyvernitakis, A., Torres, H. A., Jiang, Y., Chamilos, G., Lewis, R. E., Kontoyiannis, D. P. (2016). Initial use of combination treatment does not impact survival of 106 patients with haematologic Malignancies and mucormycosis: a propensity score analysis. Clin. Microbiol. Infect 22(9):811.e1-811.e8. doi: 10.1016/j.cmi.2016.03.029

Lamoth, F., Kontoyiannis, D. P. (2019). Therapeutic challenges of non- aspergillus invasive mold infections in immunosuppressed patients. Antimicrob. Agents Chemother. 63 (11), e01244–e01219. doi: 10.1128/AAC.01244-19

Lamoth, F., Lewis, R. E., Kontoyiannis, D. P. (2020). Role and interpretation of antifungal susceptibility testing for the management of invasive fungal infections. J. Fungi (Basel). 7 (1):17. doi: 10.3390/jof7010017

Lamoth, F., Lewis, R. E., Kontoyiannis, D. P. (2022). Investigational antifungal agents for invasive mycoses: A clinical perspective. Clin. Infect. Dis 75 (3), 534–544. doi: 10.1093/cid/ciab1070

Lanternier, F., Poiree, S., Elie, C., Garcia-Hermoso, D., Bakouboula, P., Sitbon, K., et al. (2015). Prospective pilot study of high-dose (10 mg/kg/day) liposomal amphotericin B (L-AMB) for the initial treatment of mucormycosis. J. Antimicrob. Chemother. 70 (11), 3116–3123. doi: 10.1093/jac/dkv236

Larsen, G. G., Appel, K. F., Wolff, A. M., Nielsen, J., Arnau, J. (2004). Characterisation of the Mucor circinelloides regulated promoter gpd1P. Curr. Genet. 45 (4), 225–234. doi: 10.1007/s00294-003-0484-2

Lass-Flörl, C. (2009). Zygomycosis: conventional laboratory diagnosis. Clin. Microbiol. Infect. 15 Suppl 5, 60–65. doi: 10.1111/j.1469-0691.2009.02999.x

Lax, C., Navarro-Mendoza, M. I., Pérez-Arques, C., Navarro, E., Nicolás, F. E., Garre, V. (2021). Stable and reproducible homologous recombination enables CRISPR-based engineering in the fungus Rhizopus microsporus. Cell Rep. Methods 1 (8), 100124. doi: 10.1016/j.crmeth.2021.100124

Lax, C., Navarro-Mendoza, M. I., Pérez-Arques, C., Navarro, E., Nicolás, F. E., Garre, V. (2022). Transformation and CRISPR-Cas9-mediated homologous recombination in the fungus Rhizopus microsporus. STAR Protoc. 3 (1), 101237. doi: 10.1016/j.xpro.2022.101237

Lax, C., Pérez-Arques, C., Navarro-Mendoza, M. I., Cánovas-Márquez, J. T., Tahiri, G., Pérez-Ruiz, J. A., et al. (2020). Genes, pathways, and mechanisms involved in the virulence of mucorales. Genes (Basel) 11 (3):317. doi: 10.3390/genes11030317

Lee, A. S. (2007). GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 67 (8), 3496–3499. doi: 10.1158/0008-5472.CAN-07-0325

Lee, S. C., Billmyre, R. B., Li, A., Carson, S., Sykes, S. M., Huh, E. Y., et al. (2014). Analysis of a food-borne fungal pathogen outbreak: virulence and genome of a Mucor circinelloides isolate from yogurt. mBio 5 (4), e01390–e01314. doi: 10.1128/mBio.01390-14

Lee, S. C., Li, A., Calo, S., Heitman, J. (2013). Calcineurin plays key roles in the dimorphic transition and virulence of the human pathogenic zygomycete Mucor circinelloides. PloS Pathogens 9 (9), e1003625. doi: 10.1371/journal.ppat.1003625

Lee, S. C., Li, A., Calo, S., Inoue, M., Tonthat, N. K., Bain, J. M., et al. (2015). Calcineurin orchestrates dimorphic transitions, antifungal drug responses and host-pathogen interactions of the pathogenic mucoralean fungus Mucor circinelloides. Mol. Microbiol. 97 (5), 844–865. doi: 10.1111/mmi.13071

Lee, F. Y. W., Mossad, S. B., Adal, K. A. (1999). Pulmonary mucormycosis: the last 30 years. Arch. Internal Med. 159 (12), 1301–1309. doi: 10.1001/archinte.159.12.1301

Lee, S. C., Ni, M., Li, W., Shertz, C., Heitman, J. (2010). The evolution of sex: a perspective from the fungal kingdom. Microbiol. Mol. Biol. Rev. 74 (2), 298–340. doi: 10.1128/MMBR.00005-10

Legouge, C., Caillot, D., Chrétien, M. L., Lafon, I., Ferrant, E., Audia, S., et al. (2014). The reversed halo sign: pathognomonic pattern of pulmonary mucormycosis in leukemic patients with neutropenia? Clin. Infect. Dis. 58 (5), 672–678. doi: 10.1093/cid/cit929

Lerche, C. J., Schwartz, F., Pries-Heje, M. M., Fosbøl, E. L., Iversen, K., Jensen, P. Ø., et al. (2022). Potential advances of adjunctive hyperbaric oxygen therapy in infective endocarditis. Front. Cell Infect. Microbiol. 12. doi: 10.3389/fcimb.2022.805964

Li, C. H., Cervantes, M., Springer, D. J., Boekhout, T., Ruiz-Vazquez, R. M., Torres-Martinez, S. R., et al. (2011). Sporangiospore size dimorphism is linked to virulence of Mucor circinelloides . PloS Pathog. 7 (6), e1002086. doi: 10.1371/journal.ppat.1002086

Liu, M., Lin, L., Gebremariam, T., Luo, G., Skory, C. D., French, S. W., et al. (2015). Fob1 and Fob2 Proteins Are Virulence Determinants of Rhizopus oryzae via Facilitating Iron Uptake from Ferrioxamine. PloS Pathog. 11 (5), e1004842. doi: 10.1145/2818302

Liu, M., Spellberg, B., Phan, Q. T., Fu, Y., Lee, A. S., Edwards, J. E., Jr., et al. (2010). The endothelial cell receptor GRP78 is required for mucormycosis pathogenesis in diabetic mice. J. Clin. Invest. 120 (6), 1914–1924. doi: 10.1172/JCI42164

Lockhart, S. R., Bialek, R., Kibbler, C. C., Cuenca-Estrella, M., Jensen, H. E., Kontoyiannis, D. P. (2021). Molecular techniques for genus and species determination of fungi from fresh and paraffin-embedded formalin-fixed tissue in the revised EORTC/MSGERC definitions of invasive fungal infection. Clin. Infect. Dis. 72 (Suppl 2), S109–Ss13. doi: 10.1093/cid/ciaa1836

Ma, L. J., Ibrahim, A. S., Skory, C., Grabherr, M. G., Burger, G., Butler, M., et al. (2009). Genomic analysis of the basal lineage fungus Rhizopus oryzae reveals a whole-genome duplication. PloS Genet. 5 (7), e1000549. doi: 10.1371/journal.pgen.1000549

Marr, K. A., Carter, R. A., Crippa, F., Wald, A., Corey, L. (2002). Epidemiology and outcome of mould infections in hematopoietic stem cell transplant recipients. Clin. Infect. Dis. 34 (7), 909–917. doi: 10.1086/339202

Marty, F. M., Ostrosky-Zeichner, L., Cornely, O. A., Mullane, K. M., Perfect, J. R., Thompson, G. R., 3rd, et al. (2016). Isavuconazole treatment for mucormycosis: a single-arm open-label trial and case-control analysis. Lancet Infect. Dis. 16 (7), 828–837. doi: 10.1016/S1473-3099(16)00071-2

Maurer, E., Hörtnagl, C., Lackner, M., Grässle, D., Naschberger, V., Moser, P., et al. (2019). Galleria mellonella as a model system to study virulence potential of mucormycetes and evaluation of antifungal treatment. Med. Mycol. 57 (3), 351–362. doi: 10.1093/mmy/myy042

McCarthy, M. W., Kontoyiannis, D. P., Cornely, O. A., Perfect, J. R., Walsh, T. J. (2017). Novel agents and drug targets to meet the challenges of resistant fungi. J. Infect. Dis. 216 (suppl_3), S474–SS83. doi: 10.1093/infdis/jix130

McCarthy, M. W., Walsh, T. J. (2016). PCR methodology and applications for the detection of human fungal pathogens. Expert Rev. Mol. Diagn. 16 (9), 1025–1036. doi: 10.1080/14737159.2016.1219253

Millon, L., Caillot, D., Berceanu, A., Bretagne, S., Lanternier, F., Morio, F., et al. (2022). Evaluation of serum Mucorales PCR for the diagnosis of Mucormycoses: The MODIMUCOR prospective trial. Clin. Infect. Dis 75 (5), 777–785. doi: 10.1093/cid/ciab1066

Millon, L., Herbrecht, R., Grenouillet, F., Morio, F., Alanio, A., Letscher-Bru, V., et al. (2016). 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. 22 (9), 810.e1–.e8. doi: 10.1016/j.cmi.2015.12.006

Millon, L., Larosa, F., Lepiller, Q., Legrand, F., Rocchi, S., Daguindau, E., et al. (2013). Quantitative polymerase chain reaction detection of circulating DNA in serum for early diagnosis of mucormycosis in immunocompromised patients. Clin. Infect. Dis. 56 (10), e95–e101. doi: 10.1093/cid/cit094

Morin-Sardin, S., Nodet, P., Coton, E., Jany, J. L. (2017). Mucor: a janus-faced fungal genus with human health impact and industrial applications. Fungal Biol. Rev. 31 (1), 12–32. doi: 10.1016/j.fbr.2016.11.002

Nagao, K., Ota, T., Tanikawa, A., Takae, Y., Mori, T., Udagawa, S., et al. (2005). Genetic identification and detection of human pathogenic Rhizopus species, a major mucormycosis agent, by multiplex PCR based on internal transcribed spacer region of rRNA gene. J. Dermatol. Sci. 39 (1), 23–31. doi: 10.1016/j.jdermsci.2005.01.010

Nagy, G., Szebenyi, C., Csernetics, Á, Vaz, A. G., Tóth, E. J., Vágvölgyi, C., et al. (2017). Development of a plasmid free CRISPR-Cas9 system for the genetic modification of Mucor circinelloides. Sci. Rep. 7 (1), 16800. doi: 10.1038/s41598-017-17118-2

Navarro-Mendoza, M. I., Pérez-Arques, C., Murcia, L., Martínez-García, P., Lax, C., Sanchis, M., et al. (2018). Components of a new gene family of ferroxidases involved in virulence are functionally specialized in fungal dimorphism. Sci. Rep. 8 (1), 7660. doi: 10.1038/s41598-018-26051-x

Nguyen, K. B., Sreelatha, A., Durrant, E. S., Lopez-Garrido, J., Muszewska, A., Dudkiewicz, M., et al. (2016). Phosphorylation of spore coat proteins by a family of atypical protein kinases. Proc. Natl. Acad. Sci. 113 (25), E3482–E3E91. doi: 10.1073/pnas.1605917113

Nicolas-Molina, F. E., Navarro, E., Ruiz-Vazquez, R. M. (2008). Lycopene over-accumulation by disruption of the negative regulator gene crgA in Mucor circinelloides. Appl. Microbiol. Biotechnol. 78 (1), 131–137. doi: 10.1007/s00253-007-1281-5

Nucci, M., Engelhardt, M., Hamed, K. (2019). Mucormycosis in South America: A review of 143 reported cases. Mycoses 62 (9), 730–738. doi: 10.1111/myc.12958

Omara, F. O., Blakley, B. R. (1994). The effects of iron deficiency and iron overload on cell-mediated immunity in the mouse. Br. J. Nutr. 72 (6), 899–909. doi: 10.1079/BJN19940094

Orlowski, M. (1991). Mucor dimorphism. Microbiol. Rev. 55 (2), 234–258. doi: 10.1128/mr.55.2.234-258.1991

Patel, A., Agarwal, R., Rudramurthy, S., Shevkani, M., Xess, I., Sharma, R., et al. (2021). Multicenter epidemiologic study of coronavirus disease–associated mucormycosis, India. Emerg. Infect. Dis. J. 27 (9), 2349–2359. doi: 10.3201/eid2709.210934

Patiño-Medina, J. A., Maldonado-Herrera, G., Pérez-Arques, C., Alejandre-Castañeda, V., Reyes-Mares, N. Y., Valle-Maldonado, M. I., et al. (2018). Control of morphology and virulence by ADP-ribosylation factors (Arf) in Mucor circinelloides. Curr. Genet. 64 (4), 853–869. doi: 10.1007/s00294-017-0798-0

Petrikkos, G., Skiada, A., Lortholary, O., Roilides, E., Walsh, T. J., Kontoyiannis, D. P. (2012). Epidemiology and clinical manifestations of mucormycosis. Clin. Infect. Dis. 54 Suppl 1, S23–S34. doi: 10.1093/cid/cir866

Pongas, G. N., Lewis, R. E., Samonis, G., Kontoyiannis, D. P. (2009). Voriconazole-associated zygomycosis: a significant consequence of evolving antifungal prophylaxis and immunosuppression practices? Clin. Microbiol. Infect. 15 Suppl 5, 93–97. doi: 10.1111/j.1469-0691.2009.02988.x

Potenza, L., Vallerini, D., Barozzi, P., Riva, G., Forghieri, F., Zanetti, E., et al. (2011). 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 118 (20), 5416–5419. doi: 10.1182/blood-2011-07-366526

Prakash, H., Chakrabarti, A. (2021). Epidemiology of mucormycosis in India. Microorganisms 9 (3):523. doi: 10.3390/microorganisms9030523

Price, J. C., Stevens, D. L. (1980). Hyperbaric oxygen in the treatment of rhinocerebral mucormycosis. Laryngoscope 90 (5 Pt 1), 737–747. doi: 10.1288/00005537-198005000-00002

Raut, A., Huy, N. T. (2021). Rising incidence of mucormycosis in patients with COVID-19: another challenge for India amidst the second wave? Lancet Respir. Med 9 (8), e77. doi: 10.1016/S2213-2600(21)00265-4

Ravani, S. A., Agrawal, G. A., Leuva, P. A., Modi, P. H., Amin, K. D. (2021). Rise of the phoenix: Mucormycosis in COVID-19 times. Indian J. Ophthalmol. 69 (6), 1563–1568. doi: 10.4103/ijo.IJO_310_21

Reed, C., Bryant, R., Ibrahim, A. S., Edwards, J., Jr., Filler, S. G., Goldberg, R., et al. (2008). Combination polyene-caspofungin treatment of rhino-orbital-cerebral mucormycosis. Clin. Infect. Dis. 47 (3), 364–371. doi: 10.1086/589857

Ribes, J. A., Vanover-Sams, C. L., Baker, D. J. (2000). Zygomycetes in human disease. Clin. Microbiol. Rev. 13 (2), 236–301. doi: 10.1128/CMR.13.2.236

Roden, M. M., Zaoutis, T. E., Buchanan, W. L., Knudsen, T. A., Sarkisova, T. A., Schaufele, R. L., et al. (2005). Epidemiology and outcome of zygomycosis: a review of 929 reported cases. Clin. Infect. Dis. 41 (5), 634–653. doi: 10.1086/432579

Sabirli, R., Koseler, A., Goren, T., Turkcuer, I., Kurt, O. (2021). High GRP78 levels in Covid-19 infection: A case-control study. Life Sci. 265, 118781 doi: 10.1016/j.lfs.2020.118781

Santos, R., Buisson, N., Knight, S., Dancis, A., Camadro, J. M., Lesuisse, E. (2003). Haemin uptake and use as an iron source by Candida albicans : role of CaHMX1-encoded haem oxygenase. Microbiology 149 (Pt 3), 579–588. doi: 10.1099/mic.0.26108-0

Schmidt, S., Tramsen, L., Perkhofer, S., Lass-Florl, C., Hanisch, M., Roger, F., et al. (2013). Rhizopus oryzae hyphae are damaged by human natural killer (NK) cells, but suppress NK cell mediated immunity. Immunobiology 218 (7), 939–944. doi: 10.1016/j.imbio.2012.10.013

Schmitt, K. (2012). Overcoming drawbacks of gene silencing with RNAi. Genet. Eng. Biotechnol. News. 32 (21), 40–41. doi: 10.1089/gen.32.21.19

Schwartze, V. U., Winter, S., Shelest, E., Marcet-Houben, M., Horn, F., Wehner, S., et al. (2014). Gene expansion shapes genome architecture in the human pathogen Lichtheimia corymbifera : an evolutionary genomics analysis in the ancient terrestrial mucorales (Mucoromycotina). PloS Genet. 10 (8), e1004496. doi: 10.1371/journal.pgen.1004496

Shaw, K. J., Ibrahim, A. S. (2020). Fosmanogepix: A review of the first-in-class broad spectrum agent for the treatment of invasive fungal infections. J. Fungi (Basel). 6 (4):239. doi: 10.3390/jof6040239

Shirazi, F., Farmakiotis, D., Yan, Y., Albert, N., Do, K.-A., Kontoyiannis, D. P. (2014). Diet modification and metformin have a beneficial effect in a fly model of obesity and mucormycosis. PloS One 9 (9), e108635–e. doi: 10.1371/journal.pone.0108635

Shumilov, E., Bacher, U., Perske, C., Mohr, A., Eiffert, H., Hasenkamp, J., et al. (2018). In situ validation of the endothelial cell receptor GRP78 in a case of Rhinocerebral mucormycosis. Antimicrob. Agents Chemother. 62 (5):e00172-18. doi: 10.1128/AAC.00172-18

Singh, A. K., Singh, R., Joshi, S. R., Misra, A. (2021). Mucormycosis in COVID-19: A systematic review of cases reported worldwide and in India. Diabetes Metab. Syndr. 15 (4), 102146. doi: 10.1016/j.dsx.2021.05.019

Skiada, A., Lass-Floerl, C., Klimko, N., Ibrahim, A., Roilides, E., Petrikkos, G. (2018). Challenges in the diagnosis and treatment of mucormycosis. Med. Mycol. 56 (suppl_1), 93–101. doi: 10.1093/mmy/myx101

Soliman, S. S. M., Baldin, C., Gu, Y., Singh, S., Gebremariam, T., Swidergall, M., et al. (2021). Mucoricin is a ricin-like toxin that is critical for the pathogenesis of mucormycosis. Nat. Microbiol. 6 (3), 313–326. doi: 10.1038/s41564-020-00837-0

Spellberg, B. (2009). Safety and outcomes of open-label deferasirox iron chelation therapy for mucormycosis. PloS Pathog. 53 (7), 3122–3125. doi: 10.1128/AAC.00361-09

Spellberg, B., Edwards, J., Ibrahim, A. (2005a). Novel perspectives on mucormycosis: pathophysiology, presentation, and management. Clin. Microbiol. Rev. 18, 556–569. doi: 10.1128/CMR.18.3.556-569.2005

Spellberg, B., Fu, Y., Edwards, J. E., Jr., Ibrahim, A. S. (2005b). Combination therapy with amphotericin B lipid complex and caspofungin acetate of disseminated zygomycosis in diabetic ketoacidotic mice. Antimicrob. Agents Chemother. 49 (2), 830–832. doi: 10.1128/AAC.49.2.830-832.2005

Spellberg, B., Ibrahim, A. S. (2015). “Mucormycosis,” in Harrison’s Principles of Internal Medicine, 19e . Eds. Kasper, D., Fauci, A., Hauser, S., Longo, D., Jameson, J. L., Loscalzo, J. (New York, NY: McGraw-Hill Education).

Spellberg, B., Ibrahim, A. S. (2018). “Mucormycosis,” in Harrison’s Principles of Internal Medicine, 20e . Eds. Jameson, J. L., Fauci, A. S., Kasper, D. L., Hauser, S. L., Longo, D. L., Loscalzo, J. (New York, NY: McGraw-Hill Education).

Spellberg, B., Ibrahim, A. S., Chin-Hong, P. V., Kontoyiannis, D. P., Morris, M. I., Perfect, J. R., et al. (2012a). The Deferasirox-AmBisome Therapy for Mucormycosis (DEFEAT Mucor) study: a randomized, double-blinded, placebo-controlled trial. J. Antimicrob. Chemother. 67 (3), 715–722. doi: 10.1093/jac/dkr375

Spellberg, B., Ibrahim, A., Roilides, E., Lewis, R. E., Lortholary, O., Petrikkos, G., et al. (2012b). Combination therapy for mucormycosis: why, what, and how? Clin. Infect. Dis. 54 Suppl 1, S73–S78. doi: 10.1093/cid/cir885

Spellberg, B., Kontoyiannis, D. P., Fredricks, D., Morris, M. I., Perfect, J. R., Chin-Hong, P. V., et al. (2012c). Risk factors for mortality in patients with mucormycosis. Med. Mycol. 50 (6), 611–618. doi: 10.3109/13693786.2012.669502

Springer, J., Lackner, M., Ensinger, C., Risslegger, B., Morton, C. O., Nachbaur, D., et al. (2016). Clinical evaluation of a Mucorales-specific real-time PCR assay in tissue and serum samples. J. Med. Microbiol. 65 (12), 1414–1421. doi: 10.1099/jmm.0.000375

Stevens, V. M., Mueller, S. W., Reynolds, P. M., MacLaren, R., Kiser, T. H. (2020). Extrapolating antifungal animal data to humans - Is it reliable? Curr. Fungal Infect. Rep. 14 (1), 50–62. doi: 10.1007/s12281-020-00370-x

Sugar, A. M. (2005). “Agents of mucormycosis and related species,” in Principles and Practice of Infectious Diseases. 2 , 6th ed. Eds. Mandell, G. L., Bennett, J. E., Dolin, R. (Philadelphia, PA: Elsevier Churchill Livingstone), 2973–2984.

Szebenyi, C., Gu, Y., Gebremariam, T., Kocsubé, S., Kiss-Vetráb, S., Jáger, O., et al. (2023). cotH genes are necessary for normal spore formation and virulence in Mucor lusitanicus. mBio 14 (1), e0338622. doi: 10.1128/mbio.03386-22

Tarrand, J. J., Han, X. Y., Kontoyiannis, D. P., May, G. S. (2005). Aspergillus hyphae in infected tissue: evidence of physiologic adaptation and effect on culture recovery. J. Clin. Microbiol. 43 (1), 382–386. doi: 10.1128/JCM.43.1.382-386.2005

Thieken, A., Winkelmann, G. (1992). Rhizoferrin: a complexone type siderophore of the Mucorales and entomophthorales (Zygomycetes). FEMS Microbiol. Lett. 73 (1-2), 37–41. doi: 10.1111/j.1574-6968.1992.tb05285.x

Tribble, D. R., Warkentien, T., Rodriguez, C. (2013). Trauma Infectious Diseases Outcomes Study Group of the Infectious Disease Clinical Research P. Mucormycosis after a tornado in Joplin, Missouri. N Engl. J. Med. 368 (11), 1067. doi: 10.1056/NEJMc1300111

Trieu, T. A., Navarro-Mendoza, M. I., Pérez-Arques, C., Sanchis, M., Capilla, J., Navarro-Rodriguez, P., et al. (2017). RNAi-based functional genomics identifies new virulence determinants in mucormycosis. PloS Pathogens 13 (1), e1006150. doi: 10.1371/journal.ppat.1006150

Umemura, M., Okamoto, M., Nakayama, K.-I., Sagane, K., Tsukahara, K., Hata, K., et al. (2003). GWT1 gene is required for inositol acylation of glycosylphosphatidylinositol anchors in yeast. J. Biol. Chem. 278 (26), 23639–23647. doi: 10.1074/jbc.M301044200

Uppuluri, P., Alqarihi, A., Ibrahim, A. S. (2021). Encyclopedia of Mycology 1:600–612. doi: 10.1016/B978-0-12-809633-8.21013-3

Vehreschild, J. J., Birtel, A., Vehreschild, M. J., Liss, B., Farowski, F., Kochanek, M., et al. (2013). Mucormycosis treated with posaconazole: review of 96 case reports. Crit. Rev. Microbiol. 39 (3), 310–324. doi: 10.3109/1040841X.2012.711741

Vironneau, P., Kania, R., Morizot, G., Elie, C., Garcia-Hermoso, D., Herman, P., et al. (2014). Local control of rhino-orbito-cerebral mucormycosis dramatically impacts survival. Clin. Microbiol. Infect. 20 (5), O336–O339. doi: 10.1111/1469-0691.12408

Wagner, L., Stielow, J. B., de Hoog, G. S., Bensch, K., Schwartze, V. U., Voigt, K., et al. (2020). A new species concept for the clinically relevant Mucor circinelloides complex. Persoonia 44, 67–97. doi: 10.3767/persoonia.2020.44.03

Waldorf, A. R. (1989). Pulmonary defense mechanisms against opportunistic fungal pathogens. Immunol. Series 47 (4), 243–271.

Waldorf, A. R., Levitz, S. M., Diamond, R. D. (1984a). In vivo bronchoalveolar macrophage defense against Rhizopus oryzae and Aspergillus fumigatus. J. Infect. Dis. 150 (5), 752–760. doi: 10.1093/infdis/150.5.752

Waldorf, A. R., Ruderman, N., Diamond, R. D. (1984b). Specific susceptibility to mucormycosis in murine diabetes and bronchoalveolar macrophage defense against Rhizopus. J. Clin. Invest. 74 (1), 150–160. doi: 10.1172/JCI111395

Walsh, T. J., Gamaletsou, M. N., McGinnis, M. R., Hayden, R. T., Kontoyiannis, D. P. (2012). Early clinical and laboratory diagnosis of invasive pulmonary, extrapulmonary, and disseminated mucormycosis (zygomycosis). Clin. Infect. Dis. 54 Suppl 1, S55–S60. doi: 10.1093/cid/cir868

Warkentien, T., Rodriguez, C., Lloyd, B., Wells, J., Weintrob, A., Dunne, J. R., et al. (2012). Invasive mold infections following combat-related injuries. Clin. Infect. Dis. 55 (11), 1441–1449. doi: 10.1093/cid/cis749

Warkentien, T. E., Shaikh, F., Weintrob, A. C., Rodriguez, C. J., Murray, C. K., Lloyd, B. A., et al. (2015). Impact of mucorales and other invasive molds on clinical outcomes of polymicrobial traumatic wound infections. J. Clin. Microbiol. 53 (7), 2262–2270. doi: 10.1128/JCM.00835-15

Watkins, T. N., Gebremariam, T., Swidergall, M., Shetty, A. C., Graf, K. T., Alqarihi, A., et al. (2018). Inhibition of EGFR signaling protects from mucormycosis. mBio 9 (4), e01384–e01318. doi: 10.1128/mBio.01384-18

Weintrob, A. C., Weisbrod, A. B., Dunne, J. R., Rodriguez, C. J., Malone, D., Lloyd, B. A., et al. (2015). Combat trauma-associated invasive fungal wound infections: epidemiology and clinical classification. Epidemiol. Infect. 143 (1), 214–224. doi: 10.1017/S095026881400051X

Wolff, A. M., Appel, K. F., Petersen, J. B., Poulsen, U., Arnau, J. (2002). Identification and analysis of genes involved in the control of dimorphism in Mucor circinelloides (syn. racemosus). FEMS Yeast Res. 2 (2), 203–213. doi: 10.1111/j.1567-1364.2002.tb00085.x

Worsham, P. L., Goldman, W. E. (1988). Quantitative plating of Histoplasma capsulatum without addition of conditioned medium or siderophores. J. Med. Vet. Mycol. 26 (3), 137–143. doi: 10.1080/02681218880000211

Wurster, S., Albert, N. D., Bharadwaj, U., Kasembeli, M. M., Tarrand, J. J., Daver, N., et al. (2022). Blockade of the PD-1/PD-L1 immune checkpoint pathway improves infection outcomes and enhances fungicidal host defense in a murine model of invasive pulmonary mucormycosis. Front. Immunol. 13. doi: 10.3389/fimmu.2022.838344

Wurster, S., Ruiz, O. E., Samms, K. M., Tatara, A. M., Albert, N. D., Kahan, P. H., et al. (2021). EGF-mediated suppression of cell extrusion during mucosal damage attenuates opportunistic fungal invasion. Cell Rep. 34 (12), 108896. doi: 10.1016/j.celrep.2021.108896

Yaman, G., Akyar, I., Can, S. (2012). Evaluation of the MALDI TOF-MS method for identification of Candida strains isolated from blood cultures. Diagn. Microbiol. Infect. Dis. 73 (1), 65–67. doi: 10.1016/j.diagmicrobio.2012.01.013

Zaman, K., Rudramurthy, S. M., Das, A., Panda, N., Honnavar, P., Kaur, H., et al. (2017). Molecular diagnosis of rhino-orbito-cerebral mucormycosis from fresh tissue samples. J. Med. Microbiol. 66 (8), 1124–1129. doi: 10.1099/jmm.0.000560

Keywords: mucormycosis, invasive fungal infections, immunosuppression, pathogenicity, DKA, rhizopus, Mucorales, COVID-19-associated mucormycosis

Citation: Alqarihi A, Kontoyiannis DP and Ibrahim AS (2023) Mucormycosis in 2023: an update on pathogenesis and management. Front. Cell. Infect. Microbiol. 13:1254919. doi: 10.3389/fcimb.2023.1254919

Received: 07 July 2023; Accepted: 05 September 2023; Published: 21 September 2023.

Reviewed by:

Copyright © 2023 Alqarihi, Kontoyiannis and Ibrahim. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) . The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Dimitrios P. Kontoyiannis, [email protected] ; Ashraf S. Ibrahim, [email protected]

† These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

  • DOI: 10.3389/fcimb.2023.1254919
  • Corpus ID: 262205350

Mucormycosis in 2023: an update on pathogenesis and management

  • Abdullah Alqarihi , D. Kontoyiannis , Ashraf S. Ibrahim
  • Published in Frontiers in Cellular and… 21 September 2023
  • Medicine, Environmental Science

Figures from this paper

figure 1

11 Citations

Mucormycosis: update on clinical presentation, diagnosis, and treatment, genetic basis for mucormycosis progression in covid-19 patients: from susceptibility to severity, comprehensive review on the virulence factors and therapeutic strategies with the aid of artificial intelligence against mucormycosis., corona-associated mucormycosis: case series reports and review of the literature, co-occurrence of gram-negative rods in patients with hematologic malignancy and sinopulmonary mucormycosis, experimental models to study the pathogenesis and treatment of mucormycosis, potential fungal zoonotic pathogens in cetaceans: an emerging concern, molecular architecture of chitin and chitosan-dominated cell walls in zygomycetous fungal pathogens by solid-state nmr, epithelial responses to fungal pathogens., editorial: advancements in the understanding of mucorales biology and the management of mucormycosis, 189 references, pathogenesis of mucormycosis., epidemiology and clinical manifestations of mucormycosis., mucormycosis and covid-19-associated mucormycosis: insights of a deadly but neglected mycosis, novel perspectives on mucormycosis: pathophysiology, presentation, and management, host‐iron assimilation: pathogenesis and novel therapies of mucormycosis, mucormycosis in south america: a review of 143 reported cases.

  • Highly Influential

The mucormycete-host interface.

Mucormycosis treated with posaconazole: review of 96 case reports, when uncontrolled diabetes mellitus and severe covid-19 converge: the perfect storm for mucormycosis, mucormycosis in iran: a six-year retrospective experience., related papers.

Showing 1 through 3 of 0 Related Papers

Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

jof-logo

Article Menu

  • Subscribe SciFeed
  • Recommended Articles
  • PubMed/Medline
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

Epidemiology and diagnosis of mucormycosis: an update.

mucormycosis research paper pdf

1. Introduction

2. epidemiology, 2.1. incidence of mucormycosis, 2.2. causative agents, 2.3. predisposing factors/underlying conditions, 2.3.1. diabetes mellitus and ketoacidosis, 2.3.2. hematological malignancy and hematopoietic stem cell transplantation, 2.3.3. solid organ malignancies and solid organ transplantation, 2.3.4. corticosteroids and other immunosuppressive agents, 2.3.5. iron overload, 2.3.6. breakthrough mucormycosis, 2.3.7. other, 2.3.8. no underlying disease, 2.3.9. healthcare associated, 3. diagnosis, 3.1. clinical diagnosis, 3.2. routine laboratory diagnosis, 3.2.1. histopathology, 3.2.2. direct microscopy, 3.2.3. culture, 3.3. applied and emerging molecular methods, 3.4. non-invasive diagnostic methods: an eye to the future, 3.4.1. molecular, 3.4.2. serology, 3.4.3. metabolomics-breath test, 4. conclusions, author contributions, conflicts of interest.

  • Fürbringer, P. Beobachtungen über Lungenmycose beim Menschen. Virchows Arch. 1876 , 66 , 330–365. [ Google Scholar ] [ CrossRef ]
  • Paltauf, A. Mycosis mucorina: Ein Beitrag zur Kenntnis der menschilchen Fadenpiltzer-krankungen. Virchows Arch. Pathol. Anat. 1885 , 102 , 543–564. [ Google Scholar ] [ CrossRef ]
  • 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 ] [ PubMed ]
  • Slavin, M.; Van Hal, S.; Sorrell, T.; Lee, A.; Marriott, D.; Daveson, K.; Kennedy, K.; Hajkowicz, K.; Halliday, C.; Athan, E.; et al. Invasive infections due to filamentous fungi other than Aspergillus: Epidemiology and determinants of mortality. Clin. Microbiol. Infect. 2015 , 21 , 490.e1–490.e10. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • 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 ] [ PubMed ] [ Green Version ]
  • Prakash, H.; Ghosh, A.; Rudramurthy, S.; Paul, R.; 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 ]
  • Chander, J.; Singla, N.; Kaur, M.; Punia, R.S.; Attri, A.; Alastruey-Izquierdo, A.; Stchigel, A.M.; Cano-Lira, J.F.; 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 ]
  • 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 ]
  • Jeong, W.; Keighley, C.; Wolfe, R.; Lee, W.L.; Slavin, M.A.; Kong, D.C.; 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 ]
  • Lanternier, F.; Dannaoui, E.; Morizot, G.; Elie, C.; Garcia-Hermoso, D.; Huerre, M.; Bitar, D.; Dromer, F.; Lortholary, O.; The French Mycosis Study Group. A Global Analysis of Mucormycosis in France: The RetroZygo Study (2005–2007). Clin. Infect. Dis. 2012 , 54 , S35–S43. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Patel, A.; Kaur, H.; Xess, I.; Michael, J.S.; Savio, J.; Rudramurthy, S.; Singh, R.; Shastri, P.; Umabala, P.; Sardana, R. A multicenter 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 ] [ PubMed ]
  • 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 ]
  • 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 ]
  • 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 ] [ PubMed ] [ Green Version ]
  • Rees, J.R.; Pinner, R.W.; Hajjeh, R.A.; Brandt, M.E.; Reingold, A.L. The Epidemiological Features of Invasive Mycotic Infections in the San Francisco Bay Area, 1992–1993: Results of Population-Based Laboratory Active Surveillance. Clin. Infect. Dis. 1998 , 27 , 1138–1147. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Saegeman, V.; Maertens, J.; Meersseman, W.; Spriet, I.; Verbeken, E.; Lagrou, K. Increasing Incidence of Mucormycosis in University Hospital, Belgium. Emerg. Infect. Dis. 2010 , 16 , 1456–1458. [ Google Scholar ] [ CrossRef ]
  • Guinea, J.; Escribano, P.; Vena, A.; Muñoz, P.; Martínez-Jiménez, M.D.C.; Padilla, B.; Bouza, E. Increasing incidence of mucormycosis in a large Spanish hospital from 2007 to 2015: Epidemiology and microbiological characterization of the isolates. PLoS ONE 2017 , 12 , e0179136. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • 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 , 1–6. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chakrabarti, A.; Das, A.; Sharma, A.; Panda, N.; Das, S.; Gupta, K.; Sakhuja, V. Ten Years’ Experience in Zygomycosis at a Tertiary Care Centre in India. J. Infect. 2001 , 42 , 261–266. [ Google Scholar ] [ CrossRef ]
  • 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 ]
  • 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 ]
  • Chakrabarti, A.; Singh, R. Mucormycosis in India: Unique features. Mycoses 2014 , 57 (Suppl. 3), 85–90. [ Google Scholar ] [ CrossRef ]
  • 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 ] [ Green Version ]
  • 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 ] [ Green Version ]
  • Lu, X.-L.; Najafzadeh, M.; Dolatabadi, S.; Ran, Y.-P.; Ende, A.G.V.D.; Shen, Y.-N.; Li, C.-Y.; Xi, L.-Y.; Hao, F.; Zhang, Q.-Q.; et al. Taxonomy and epidemiology of Mucor irregularis, agent of chronic cutaneous mucormycosis. Pers. Mol. Phylogeny Evol. Fungi 2013 , 30 , 48–56. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • 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 ]
  • Pagano, L.; Valentini, C.G.; Posteraro, B.; Girmenia, C.; Ossi, C.; Pan, A.; Candoni, A.; Nosari, A.; Riva, M.; Cattaneo, C.; et al. Zygomycosis in Italy: A Survey of FIMUA-ECMM (Federazione Italiana Di Micopatologia Umana ed Animale and European Confederation of Medical Mycology). J. Chemother. 2009 , 21 , 322–329. [ Google Scholar ] [ CrossRef ]
  • 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 ]
  • 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 ]
  • 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. Médicale 2018 , 28 , 269–273. [ Google Scholar ] [ CrossRef ]
  • Vaezi, A.; Moazeni, M.; Rahimi, M.T.; De Hoog, S.; Badali, H. Mucormycosis in Iran: A systematic review. Mycoses 2016 , 59 , 402–415. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • El Zein, S.; El-Sheikh, J.; El Zakhem, A.; Ibrahim, D.; Bazarbachi, A.; Kanj, S.S. Mucormycosis in hospitalized patients at a tertiary care center in Lebanon: A case series. Infection 2018 , 46 , 811–821. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kennedy, K.; Daveson, K.; Slavin, M.; Van Hal, S.; Sorrell, T.; Lee, A.; Marriott, D.; Chapman, B.; Halliday, C.; Hajkowicz, K.; et al. Mucormycosis in Australia: Contemporary epidemiology and outcomes. Clin. Microbiol. Infect. 2016 , 22 , 775–781. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Stemler, J.; Hamed, K.; Salmanton-García, J.; Rezaei-Matehkolaei, A.; Gräfe, S.K.; Sal, E.; Zarrouk, M.; Seidel, D.; Khedr, R.A.; 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 . [ Google Scholar ] [ CrossRef ]
  • WHO. Global Report on Diabetes. 2016. Available online: https://www.who.int/publications/i/item/global-report-on-diabetes (accessed on 10 September 2020).
  • Whiting, D.R.; Guariguata, L.; Weil, C.; Shaw, J. IDF Diabetes Atlas: Global estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res. Clin. Pr. 2011 , 94 , 311–321. [ Google Scholar ] [ CrossRef ]
  • Prakash, H.; Chakrabarti, A. Global Epidemiology of Mucormycosis. J. Fungi 2019 , 5 , 26. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Manesh, A.; Rupali, P.; Sullivan, M.O.; Mohanraj, P.; Rupa, V.; George, B.; Michael, J.S. Mucormycosis—A clinicoepidemiological review of cases over 10 years. Mycoses 2019 , 62 , 391–398. [ Google Scholar ] [ CrossRef ]
  • Pagano, L.; Offidani, M.; Fianchi, L.; Nosari, A.; Candoni, A.; Piccardi, M.; Corvatta, L.; D’Antonio, D.; Girmenia, C.; Martino, P.; et al. Mucormycosis in hematologic patients. Haematologica 2004 , 89 , 207–214. [ Google Scholar ]
  • Xhaard, A.; Lanternier, F.; Porcher, R.; Dannaoui, E.; Bergeron, A.; Clement, L.; Lacroix, C.; Herbrecht, R.; Legrand, F.; Mohty, M.; et al. Mucormycosis after allogeneic haematopoietic stem cell transplantation: A French Multicentre Cohort Study (2003–2008). Clin. Microbiol. Infect. 2012 , 18 , E396–E400. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Park, B.J.; Pappas, P.G.; Wannemuehler, K.A.; Alexander, B.D.; Anaissie, E.J.; Andes, D.R.; Baddley, J.W.; Brown, J.M.; Brumble, L.M.; Freifeld, A.G.; et al. Invasive Non-AspergillusMold Infections in Transplant Recipients, United States, 2001–2006. Emerg. Infect. Dis. 2011 , 17 , 1855–1864. [ Google Scholar ] [ CrossRef ]
  • Pappas, P.G.; Alexander, B.D.; Andes, D.R.; Hadley, S.; Kauffman, C.A.; Freifeld, A.; Anaissie, E.J.; Brumble, L.M.; Herwaldt, L.; Ito, J.; et al. Invasive Fungal Infections among Organ Transplant Recipients: Results of the Transplant-Associated Infection Surveillance Network (TRANSNET). Clin. Infect. Dis. 2010 , 50 , 1101–1111. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Azie, N.; Neofytos, D.; Pfaller, M.; Meier-Kriesche, H.-U.; Quan, S.-P.; Horn, D. The PATH (Prospective Antifungal Therapy) Alliance ® registry and invasive fungal infections: Update 2012. Diagn. Microbiol. Infect. Dis. 2012 , 73 , 293–300. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Almyroudis, N.G.; A Sutton, D.; Linden, P.K.; Rinaldi, M.G.; Fung, J.; Kusne, S. Zygomycosis in Solid Organ Transplant Recipients in a Tertiary Transplant Center and Review of the Literature. Arab. Archaeol. Epigr. 2006 , 6 , 2365–2374. [ Google Scholar ] [ CrossRef ]
  • 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 ] [ PubMed ]
  • Steinbach, W.J.; Cramer, R.A.; Perfect, B.Z.; Asfaw, Y.G.; Sauer, T.C.; Najvar, L.K.; Kirkpatrick, W.R.; Patterson, T.F.; Benjamin, D.K.; Heitman, J.; et al. Calcineurin Controls Growth, Morphology, and Pathogenicity in Aspergillus fumigatus. Eukaryot. Cell 2006 , 5 , 1091–1103. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lax, C.; Pérez-Arques, C.; Navarro-Mendoza, M.I.; Cánovas-Márquez, J.T.; Tahiri, G.; Pérez-Ruiz, J.A.; Osorio-Concepción, M.; Navarro, E.; Navarro, E.; Garre, V.; et al. Genes, Pathways, and Mechanisms Involved in the Virulence of Mucorales. Genes 2020 , 11 , 317. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kontoyiannis, D.P.; Lewis, R.E. How I treat mucormycosis. Blood 2011 , 118 , 1216–1224. [ Google Scholar ] [ CrossRef ]
  • Hoang, K.; Abdo, T.; Reinersman, J.M.; Lu, R.; Higuita, N.I.A. A case of invasive pulmonary mucormycosis resulting from short courses of corticosteroids in a well-controlled diabetic patient. Med. Mycol. Case Rep. 2020 , 29 , 22–24. [ Google Scholar ] [ CrossRef ]
  • Arce-Salinas, C.A.; Silva-Perez, E. Mucormycosis complications in systemic lupus erythematosus. Lupus 2010 , 19 , 985–988. [ Google Scholar ] [ CrossRef ]
  • Nogueira, E.L.; Ind, P.W.; Friedland, J.S.; Salama, A.D. Mucormycosis May Mimic Disease Relapse in Wegener’s Granulomatosis. J. Rheumatol. 2010 , 37 , 1364–1365. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bhatt, H.; Zilani, G.; Hayhurst, C. Orbitocerebral mucormycosis and intracranial haemorrhage: A role for caution with steroids in suspected giant cell arteritis. BMJ Case Rep. 2018 , 2018 , 2017224086. [ Google Scholar ] [ CrossRef ]
  • Ibrahim, A.; Spellberg, B.; Edwards, J., Jr. Iron acquisition: A novel prospective on mucormycosis pathogenesis and treatment. Curr. Opin. Infect. Dis. 2008 , 21 , 620–625. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Artis, W.M.; Fountain, J.A.; Delcher, H.K.; Jones, H.E. A mechanism of susceptibility to mucormycosis in diabetic ketoacidosis: Transferrin and iron availability. Diabetes 1982 , 31 , 1109–1114. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Boelaert, J.R.; Van Roost, G.F.; Vergauwe, P.L.; Verbanck, J.J.; De Vroey, C.; Segaert, M.F. The role of desferrioxamine in dialysis-associated mucormycosis: Report of three cases and review of the literature. Clin. Nephrol. 1988 , 29 , 261–266. [ Google Scholar ] [ PubMed ]
  • Singh, N.; Sun, H.-Y. Iron overload and unique susceptibility of liver transplant recipients to disseminated disease due to opportunistic pathogens. Liver Transplant. 2008 , 14 , 1249–1255. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kontoyiannis, D.P.; Lionakis, M.S.; Lewis, R.E.; Chamilos, G.; Healy, M.; Perego, C.; Safdar, A.; Kantarjian, H.; Champlin, R.; Walsh, T.J.; et al. Zygomycosis in a Tertiary-Care Cancer Center in the Era ofAspergillus-Active Antifungal Therapy: A Case-Control Observational Study of 27 Recent Cases. J. Infect. Dis. 2005 , 191 , 1350–1360. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Marty, F.M.; Cosimi, L.A.; Baden, L.R. Breakthrough Zygomycosis after Voriconazole Treatment in Recipients of Hematopoietic Stem-Cell Transplants. N. Engl. J. Med. 2004 , 350 , 950–952. [ Google Scholar ] [ CrossRef ]
  • Imhof, A.; Balajee, S.A.; Fredricks, D.N.; Englund, J.A.; Marr, K.A. Breakthrough Fungal Infections in Stem Cell Transplant Recipients Receiving Voriconazole. Clin. Infect. Dis. 2004 , 39 , 743–746. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Siwek, G.T.; Dodgson, K.J.; De Margarida, M.-S.; Bartelt, L.A.; Kilborn, S.B.; Hoth, P.; Diekema, D.J.; Pfaller, M.A. Invasive Zygomycosis in Hematopoietic Stem Cell Transplant Recipients Receiving Voriconazole Prophylaxis. Clin. Infect. Dis. 2004 , 39 , 584–587. [ Google Scholar ] [ CrossRef ]
  • Lamaris, G.A.; Ben-Ami, R.; Lewis, R.E.; Chamilos, G.; Samonis, G.; Kontoyiannis, D.P. Increased Virulence of Zygomycetes Organisms Following Exposure to Voriconazole: A Study Involving Fly and Murine Models of Zygomycosis. J. Infect. Dis. 2009 , 199 , 1399–1406. [ Google Scholar ] [ CrossRef ]
  • Auberger, J.; Lass-Flörl, C.; Aigner, M.; Clausen, J.; Gastl, G.; Nachbaur, D. Invasive fungal breakthrough infections, fungal colonization and emergence of resistant strains in high-risk patients receiving antifungal prophylaxis with posaconazole: Real-life data from a single-centre institutional retrospective observational study. J. Antimicrob. Chemother. 2012 , 67 , 2268–2273. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Rausch, C.R.; DiPippo, A.J.; Bose, P.; Kontoyiannis, D.P. Breakthrough Fungal Infections in Patients with Leukemia Receiving Isavuconazole. Clin. Infect. Dis. 2018 , 67 , 1610–1613. [ Google Scholar ] [ CrossRef ]
  • Lamoth, F.; Chung, S.J.; Damonti, L.; Alexander, B.D. Changing Epidemiology of Invasive Mold Infections in Patients Receiving Azole Prophylaxis. Clin. Infect. Dis. 2017 , 64 , 1619–1621. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lionakis, M.S.; Lewis, R.E.; Kontoyiannis, D.P. Breakthrough Invasive Mold Infections in the Hematology Patient: Current Concepts and Future Directions. Clin. Infect. Dis. 2018 , 67 , 1621–1630. [ Google Scholar ] [ CrossRef ]
  • Antinori, S.; Nebuloni, M.; Magni, C.; Fasan, M.; Adorni, F.; Viola, A.; Corbellino, M.; Galli, M.; Vago, G.; Parravicini, C.; et al. Trends in the postmortem diagnosis of opportunistic invasive fungal infections in patients with AIDS: A retrospective study of 1630 autopsies performed between 1984 and 2002. Am. J. Clin. Pathol. 2009 , 132 , 221–227. [ Google Scholar ] [ CrossRef ]
  • Moreira, J.; Varon, A.; Galhardo, M.C.; Santos, F.; Lyra, M.; Castro, R.; Oliveira, R.; Lamas, C. The burden of mucormycosis in HIV-infected patients: A systematic review. J. Infect. 2016 , 73 , 181–188. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kerezoudis, P.; Watts, C.R.; Bydon, M.; Dababneh, A.S.; Deyo, C.N.; Frye, J.M.; Kelley, P.C.; Kemp, A.M.; Palraj, B.V.; Pupillo, G.T. Diagnosis and Treatment of Isolated Cerebral Mucormycosis: Patient-Level Data Meta-Analysis and Mayo Clinic Experience. World Neurosurg. 2019 , 123 , 425–434. [ Google Scholar ] [ CrossRef ]
  • Skiada, A.; Petrikkos, G. Cutaneous mucormycosis. Skinmed 2013 , 11 , 155–159. [ Google Scholar ]
  • 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 ]
  • Rao, C.Y.; Kurukularatne, C.; Garcia-Diaz, J.B.; Kemmerly, S.A.; Reed, D.; Fridkin, S.K.; Morgan, J. Implications of Detecting the Mold Syncephalastrum in Clinical Specimens of New Orleans Residents After Hurricanes Katrina and Rita. J. Occup. Environ. Med. 2007 , 49 , 411–416. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Walsh, T.J.; Hospenthal, D.R.; Petraitis, V.; Kontoyiannis, D.P. Necrotizing Mucormycosis of Wounds Following Combat Injuries, Natural Disasters, Burns, and Other Trauma. J. Fungi 2019 , 5 , 57. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Bhadauria, D.; Etta, P.; Chelappan, A.; Gurjar, M.; Kaul, A.; Sharma, R.K.; Gupta, A.; Prasad, N. Isolated bilateral renal mucormycosis in apparently immunocompetent patients—A case series from India and review of the literature. Clin. Kidney J. 2018 , 11 , 769–776. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Liang, G.-Z.; Xu, W.-Q.; Zheng, X.-L.; Mei, H.; Lv, G.-X.; Shen, Y.-N.; Li, D.-M.; Liu, W. Successful Treatment by Surgery of a Primary Cutaneous Mucormycosis Caused by Mucor irregularis . Mycopathologia 2018 , 183 , 445–449. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Rammaert, B.; Lanternier, F.; Zahar, J.-R.; Dannaoui, E.; Bougnoux, M.-E.; Lecuit, M.; Lortholary, O. Healthcare-associated mucormycosis. Clin. Infect. Dis. 2012 , 54 (Suppl. 1), S44–S54. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • 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 ] [ PubMed ] [ Green Version ]
  • Petrikkos, G.; Skiada, A.; Sambatakou, H.; Toskas, A.; Vaiopoulos, G.; Giannopoulou, M.; Katsilambros, N. Mucormycosis: Ten-Year Experience at a Tertiary-Care Center in Greece. Eur. J. Clin. Microbiol. Infect. Dis. 2003 , 22 , 753–756. [ Google Scholar ] [ CrossRef ]
  • 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 ]
  • LeMaile-Williams, M.; Burwell, L.A.; Salisbury, D.; Noble-Wang, J.; Arduino, M.J.; 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 ]
  • Cheng, V.C.C.; Chan, J.F.-W.; Ngan, A.H.Y.; To, K.K.; 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 ]
  • Hampson, F.G.; Ridgway, E.; Feeley, K.; Reilly, J. A fatal case of disseminated zygomycosis associated with the use of blood glucose self-monitoring equipment. J. Infect. 2005 , 51 , e269–e272. [ Google Scholar ] [ CrossRef ]
  • Prabhu, S.; Alqahtani, M.; Al Shehabi, M. A fatal case of rhinocerebral mucormycosis of the jaw after dental extractions and review of literature. J. Infect. Public Health 2018 , 11 , 301–303. [ Google Scholar ] [ CrossRef ]
  • 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 ]
  • 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 ]
  • Chamilos, G.; Marom, E.M.; Lewis, R.E.; Lionakis, M.S.; Kontoyiannis, D.P. Predictors of Pulmonary Zygomycosis versus Invasive Pulmonary Aspergillosis in Patients with Cancer. Clin. Infect. Dis. 2005 , 41 , 60–66. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Jiménez-Zarazúa, O.; Vélez-Ramírez, L.N.; Alcocer-León, M.; Utrilla-Álvarez, J.D.; Martínez-Rivera, M.A.; Flores-Saldaña, G.A.; Mondragón, J.D. A case of concomitant pulmonary tuberculosis and mucormycosis in an insulin-dependent diabetic patient. J. Clin. Tuberc. Other Mycobact. Dis. 2019 , 16 , 100105. [ Google Scholar ]
  • Legouge, C.; Caillot, D.; Chretien, M.L.; Lafon, I.; Ferrant, E.; Audia, S.; Pages, P.-B.; Roques, M.; Estivalet, L.; Martin, L.; et al. The reversed halo sign: Pathognomonic pattern of pulmonary mucormycosis in leukemic patients with neutropenia? Clin. Infect. Dis. 2014 , 58 , 672–678. [ Google Scholar ] [ CrossRef ]
  • Jung, J.; Kim, M.Y.; Lee, H.; Park, Y.; Lee, S.-O.; Choi, S.-H.; Kim, Y.; Woo, J.; 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 ] [ Green Version ]
  • Nam, B.D.; Kim, T.J.; Lee, K.S.; Kim, T.S.; Han, J.; Chung, M.J. Pulmonary Mucormycosis: Risk factors, Radiologic findings, and Pathologic Correlation. Eur. Radiol. 2018 , 28 , 788–795. [ Google Scholar ] [ CrossRef ]
  • Liu, Y.; Wu, H.; Huang, F.; Fan, Z.; Xu, B. Utility of 18F-FDG PET/CT in Diagnosis and Management of Mucormycosis. Clin. Nucl. Med. 2013 , 38 , e370–e371. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Guarner, J.; Brandt, M.E. Histopathologic Diagnosis of Fungal Infections in the 21st Century. Clin. Microbiol. Rev. 2011 , 24 , 247–280. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Ribes, J.A.; Vanover-Sams, C.L.; Baker, D.J. Zygomycetes in human disease. Clin. Microbiol. Rev. 2000 , 13 , 236–301. [ Google Scholar ] [ CrossRef ]
  • McDermott, N.E.; Barrett, J.; Hipp, J.; Merino, M.J.; Lee, C.-C.R.; Waterman, P.; Domingo, D.L.; Walsh, T.J. Successful treatment of periodontal mucormycosis: Report of a case and literature review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endodontol. 2010 , 109 , e64–e69. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Cornely, O.A.; Alastruey-Izquierdo, A.; Arenz, D.; Chen, S.C.A.; Dannaoui, E.; Hochhegger, B.; Hoenigl, M.; Jensen, H.E.; Lagrou, K.; Mucormycosis ECMM MSG Global Guideline Writing Group; 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 ]
  • 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 ]
  • 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. Using immunohistochemistry to assess the accuracy of histomorphologic diagnosis of aspergillosis and mucormycosis. Clin. Infect. Dis. 2015 , 61 , 1664–1670. [ Google Scholar ]
  • Son, H.-J.; Song, J.S.; Choi, S.; Jung, J.; Kim, M.J.; Chong, Y.P.; Lee, S.-O.; Choi, S.-H.; Kim, Y.S.; Woo, J.H.; et al. A comparison of histomorphologic diagnosis with culture- and immunohistochemistry-based diagnosis of invasive aspergillosis and mucormycosis. Infect. Dis. 2020 , 52 , 279–283. [ Google Scholar ] [ CrossRef ]
  • Cassagne, C.; Ranque, S.; Normand, A.-C.; Fourquet, P.; Thiebault, S.; Planard, C.; Hendrickx, M.; Piarroux, R. Mould Routine Identification in the Clinical Laboratory by Matrix-Assisted Laser Desorption Ionization Time-Of-Flight Mass Spectrometry. PLoS ONE 2011 , 6 , e28425. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • 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 ] [ PubMed ]
  • Zaman, K.; Rudramurthy, S.M.; Das, A.; Panda, N.; Honnavar, P.; Kaur, H.; Chakrabarti, A. Molecular diagnosis of rhino-orbito-cerebral mucormycosis from fresh tissue samples. J. Med. Microbiol. 2017 , 66 , 1124–1129. [ Google Scholar ] [ CrossRef ]
  • Alanio, A.; Garcia-Hermoso, D.; Mercier-Delarue, S.; Lanternier, F.; Gits-Muselli, M.; Menotti, J.; Denis, B.; Bergeron, A.; Legrand, M.; Lortholary, O.; et al. Molecular identification of Mucor in human tissues: Contribution of PCR electrospray-ionization mass spectrometry. Clin. Microbiol. Infect. 2015 , 21 , 594.e1–594.e5. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lengerova, M.; Racil, Z.; Hrncirova, K.; Kocmanova, I.; Volfova, P.; Ricna, D.; Bejdak, P.; Moulis, M.; Pavlovsky, Z.; Winbergerova, B.; et al. Rapid detection and identification of Mucormycetesand 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 ] [ PubMed ] [ Green Version ]
  • Dadwal, S.S.; Kontoyiannis, D.P. Recent advances in the molecular diagnosis of mucormycosis. Expert Rev. Mol. Diagn. 2018 , 18 , 845–854. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Springer, J.; Goldenberger, D.; Schmidt, F.; Weisser, M.; Wehrle-Wieland, E.; Einsele, H.; Frei, R.; Loeffler, J. Development and application of two independent real-time PCR assays to detect clinically relevant Mucorales species. J. Med. Microbiol. 2016 , 65 , 227–234. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Caramalho, R.; Madl, L.; Rosam, K.; Rambach, G.; Speth, C.; Pallua, J.; Larentis, T.; Araujo, R.; Alastruey-Izquierdo, A.; Lass-Flörl, C.; et al. Evaluation of a Novel Mitochondrial Pan-Mucorales Marker for the Detection, Identification, Quantification, and Growth Stage Determination of Mucormycetes. J. Fungi 2019 , 5 , 98. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Hata, D.J.; Buckwalter, S.P.; Pritt, B.S.; Roberts, G.D.; Wengenack, N.L. Real-time PCR method for detection of zygomycetes. J. Clin. Microbiol. 2008 , 46 , 2353–2358. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • 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 , 00746-18. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kasai, M.; Harrington, S.M.; Francesconi, A.; Petraitis, V.; Petraitiene, R.; Beveridge, M.G.; Knudsen, T.; Milanovich, J.; Cotton, M.P.; Hughes, J.; et al. Detection of a Molecular Biomarker for Zygomycetes by Quantitative PCR Assays of Plasma, Bronchoalveolar Lavage, and Lung Tissue in a Rabbit Model of Experimental Pulmonary Zygomycosis. J. Clin. Microbiol. 2008 , 46 , 3690–3702. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bernal-Martínez, L.; Buitrago, M.J.; Castelli, M.V.; Rodriguez-Tudela, J.L.; Cuenca-Estrella, M. Development of a single tube multiplex real-time PCR to detect the most clinically relevant Mucormycetes species. Clin. Microbiol. Infect. 2013 , 19 , E1–E7. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Mercier, T.; Reynders, M.B.; 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 ]
  • 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 ]
  • Springer, J.; White, P.L.; Kessel, J.; Wieters, I.; Teschner, D.; Korczynski, D.; Liebregts, T.; Cornely, O.A.; Schwartz, S.; Elgeti, T.; et al. A comparison of Aspergillus and Mucorales PCR testing of different bronchoscopic alveolar lavage fluid fractions from patients with suspected invasive pulmonary fungal disease. J. Clin. Microbiol. 2018 , 56 , e01655-17. [ Google Scholar ] [ PubMed ] [ Green Version ]
  • Scherer, E.; Iriart, X.; Bellanger, A.P.; Dupont, D.; Guitard, J.; Gabriel, F.; Cassaing, S.; Charpentier, E.; Guenounou, S.; Cornet, M.; et al. Quantitative PCR (qPCR) Detection of Mucorales DNA in Bronchoalveolar Lavage Fluid to Diagnose Pulmonary Mucormycosis. J. Clin. Microbiol. 2018 , 56 , JCM.00289-18. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chan-Tack, K.M.; Nemoy, L.L.; Perencevich, E.N. Central venous catheter-associated fungemia secondary to mucormycosis. Scand. J. Infect. Dis. 2005 , 37 , 925–927. [ Google Scholar ] [ CrossRef ]
  • Dizbay, M.; Adisen, E.; Kustimur, S.; Sari, N.; Cengiz, B.; Yalcin, B.; Kalkanci, A.; Gonul, I.I.; Sugita, T. Fungemia and cutaneous zygomycosis due to Mucor circinelloides in an intensive care unit patient: Case report and review of literature. Jpn. J. Infect. Dis. 2009 , 62 , 146–148. [ Google Scholar ]
  • Bard, J.D.; Mangahis, A.; Hofstra, T.C.; Bender, J.M. First case report of bloodstream infection by Rhizomucor pusillus in a child with hemophagocytic lymphohistiocytosis. Med. Mycol. Case Rep. 2014 , 5 , 20–23. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Arroyo, M.A.; Schmitt, B.H.; Davis, T.E.; Relich, R.F. Detection of the Dimorphic Phases of Mucor circinelloides in Blood Cultures from an Immunosuppressed Female. Case Rep. Infect. Dis. 2016 , 2016 . [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Millon, L.; Scherer, E.; Rocchi, S.; Bellanger, A.-P. Molecular Strategies to Diagnose Mucormycosis. J. Fungi 2019 , 5 , 24. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Millon, L.; LaRosa, F.; Lepiller, Q.; Legrand, F.; Rocchi, S.; Daguindau, E.; Scherer, E.; Bellanger, A.-P.; Leroy, J.; Grenouillet, F. Quantitative Polymerase Chain Reaction Detection of Circulating DNA in Serum for Early Diagnosis of Mucormycosis in Immunocompromised Patients. Clin. Infect. Dis. 2013 , 56 , e95–e101. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • 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 ] [ Green Version ]
  • Legrand, M.; Gits-Muselli, M.; Boutin, L.; Garcia-Hermoso, D.; Maurel, V.; Soussi, S.; Benyamina, M.; Ferry, A.; Chaussard, M.; Hamane, S.; et al. Detection of Circulating Mucorales DNA in Critically Ill Burn Patients: Preliminary Report of a Screening Strategy for Early Diagnosis and Treatment. Clin. Infect. Dis. 2016 , 63 , 1312–1317. [ Google Scholar ] [ CrossRef ]
  • Springer, J.; Lackner, M.; Ensinger, C.; Risslegger, B.; Morton, C.O.; Nachbaur, D.; Lass-Flörl, C.; Einsele, H.; Heinz, W.J.; Loeffler, J. Clinical evaluation of a Mucorales-specific real-time PCR assay in tissue and serum samples. J. Med. Microbiol. 2016 , 65 , 1414–1421. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Ino, K.; Nakase, K.; Nakamura, A.; Nakamori, Y.; Sugawara, Y.; Miyazaki, K.; Monma, F.; Fujieda, A.; Sugimoto, Y.; Ohishi, K.; et al. Management of Pulmonary Mucormycosis Based on a Polymerase Chain Reaction (PCR) Diagnosis in Patients with Hematologic Malignancies: A Report of Four Cases. Intern. Med. 2017 , 56 , 707–711. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • 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 , e00094-18. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • 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. [ Google Scholar ]
  • 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 ]
Current Species NamesPrevious Names/Synonyms
Lichtheimia corymbiferaAbsidia corymbifera, Mycocladus corymbifer
Lichtheimia ornataAbsidia ornata
Lichtheimia ramosaAbsidia ramosa, Mycocladus ramosus
Mucor ardhlaengiktusMucor ellipsoideus, Mucor circinelloides f. circinelloides
Mucor circinelloidesRhizomucor regularior, Rhizomucor variabilis var. regularior
Mucor griseocyanusMucor circinelloides f. griseocyanus
Mucor irregularisRhizomucor variabilis
Mucor jansseniiMucor circinelloides f. janssenii
Mucor lusitanicusMucor circinelloides f. lusitanicus
Rhizopus arrhizus (incl. var. delemar)Rhizopus oryzae
Rhizopus microsporusRhizopus microsporus var. azygosporus, var. chinensis, var. oligosporus, var. rhizopodiformis, var. tuberosus
ReferenceCharacteristics of StudiesRisk Factors/Underlying Diseases (%)
Countries of Origin of CasesProspective StudyMulticenter StudyTime PeriodTotal no. of ptsDMHMHSCTSOM/ SOTAI/COTrauma HIVNone
Roden et al. 2005 [ ]GlobalNoYes1940–20039293615.851/718 219
Jeong et al. 2019 [ ]GlobalNoYes2000–20178514032 1/143/3320 18
Skiada et al. 2011 [ ]EuropeYesYes2005–20072301744 5/4441728
Lanternier et al. 2012 [ ]FranceNoYes2005–20071012350122/3131811
Pagano et al. 2009 [ ]ItalyYesYes2004–200760186238/3/502173
Kontoyiannis et al. 2016 [ ]USANo Yes2005–20145555240116/15NA42NA
Nucci et al. 2019 [ ]South AmericaNoYes1960–201814342112/13NA2027.7
Corzo-Leon et al. 2017 [ ]MexicoNoYes1982–20164187217 1/12.30.74
Chakrabarti et al. 2006 [ ]IndiaNoNo2000–200417873.61.1 /0.61.77.30.611.8
Chakrabarti et al. 2009 [ ]IndiaYesNo2006–200775449 /5291113
Prakash et al. 2019 [ ]IndiaYesYes2013–201530356.86 /69.910-10.5
Patel et al. 2020 [ ]IndiaYesYes2016–201746574811.5/6.5/3.76.9-11.8
Dolatabadi et al. 2018 [ ]IranNoYes2008–201420875323/3NA4-2
Vaezi et al. 2016 [ ]IranNoYes1990–2015984861/23NA1-10
El Zein et al. 2018 [ ]LebanonNoNo2008–2018203565 /570---
Kennedy et al. 2016 [ ]AustraliaNoYes2004–2012742748.6183/1112/ 5323 11
Stemler et al. 2020 [ ]Middle East and North AfricaNoYes1968–201931049.716.5 2/1721.6120.35.8
MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

Skiada, A.; Pavleas, I.; Drogari-Apiranthitou, M. Epidemiology and Diagnosis of Mucormycosis: An Update. J. Fungi 2020 , 6 , 265. https://doi.org/10.3390/jof6040265

Skiada A, Pavleas I, Drogari-Apiranthitou M. Epidemiology and Diagnosis of Mucormycosis: An Update. Journal of Fungi . 2020; 6(4):265. https://doi.org/10.3390/jof6040265

Skiada, Anna, Ioannis Pavleas, and Maria Drogari-Apiranthitou. 2020. "Epidemiology and Diagnosis of Mucormycosis: An Update" Journal of Fungi 6, no. 4: 265. https://doi.org/10.3390/jof6040265

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

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

Affiliations.

  • 1 Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF) partner site Bonn-Cologne, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; Clinical Trials Center Cologne, University Hospital of Cologne, Cologne, Germany. Electronic address: [email protected].
  • 2 Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Madrid, Spain.
  • 3 Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany.
  • 4 Centre for Infectious Diseases and Microbiology Laboratory Services, New South Wales Health Pathology, and the Department of Infectious Diseases, Westmead Hospital, School of Medicine, University of Sydney, Sydney, NSW, Australia.
  • 5 Université Paris-Descartes, Faculté de Médecine, APHP, Hôpital Européen Georges Pompidou, Unité de Parasitologie-Mycologie, Service de Microbiologie, Paris, France.
  • 6 Radiology, Hospital São Lucas da Pontificia Universidade Catolica do Rio Grande do Sul (PUCRS), Escola de Medicina, Porto Alegre, Brazil; Radiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil.
  • 7 Section of Infectious Diseases and Tropical Medicine and Division of Pulmonology, Medical University of Graz, Graz, Austria; Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, USA.
  • 8 Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
  • 9 Department of Microbiology, Immunology and Transplantation, KU Leuven and Clinical Department of Laboratory Medicine and National Reference Center for Mycosis, University Hospitals Leuven, Leuven, Belgium.
  • 10 Infectious Diseases Clinic, Sant'Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
  • 11 Divisions of Critical Care and Pulmonology, Department of Medicine, Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences University of the Witwatersrand, Johannesburg, South Africa.
  • 12 Infectious Diseases Unit, 3rd Department of Paediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Thessaloniki, Greece; Hippokration General Hospital, Thessaloniki, Greece.
  • 13 Division of Infectious Diseases, Department of Medicine, Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
  • 14 Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany.
  • 15 Department of Infectious Diseases, Hacettepe University School of Medicine, Ankara, Turkey.
  • 16 Institut Pasteur, National Reference Center for Invasive Mycoses and Antifungals, Department of Mycology, CNRS UMR2000, Parasitology-Mycology Laboratory, Lariboisière, Saint-Louis, Fernand Widal Hospitals, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France.
  • 17 Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands; Centre of Expertise in Mycology RadboudUMC/Canisius Wilhelmina Hospital, Nijmegen, The Netherlands; Ministry of Health, Directorate General of Health Services, Ibri, Oman.
  • 18 Department of Medical Microbiology, Hacettepe University School of Medicine, Sıhhiye Ankara, Turkey.
  • 19 Department of Medical Mycology/Invasive Fungi Research Center (IFRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
  • 20 Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Infectious Diseases Unit, Tel Aviv Medical Center, Tel- Aviv, Israel.
  • 21 Dermatology Service & Mycology Department, Hospital General de México "Dr. Eduardo Liceaga", Mexico City, Mexico.
  • 22 Infectious Diseases Unit, Istituto Giannina Gaslini Children's Hospital, Genoa, Italy.
  • 23 Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
  • 24 Special Mycology Laboratory, Division of Infectious Diseases, Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
  • 25 Department of Epidemiology and Infectious Diseases, Hospital General Dr Manuel Gea González, Mexico City, Mexico; Medical Mycology and Fungal Immunology/Wellcome Trust Strategic Award Program, Aberdeen Fungal Group, University of Aberdeen, King's College, Aberdeen, UK.
  • 26 Oncohematology Clinic, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia.
  • 27 InfectiousDisease Research Program, Department of Paediatric Hematology/Oncology and Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.
  • 28 Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación v Sanitaria Gregorio Marañón, Madrid, Spain; Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
  • 29 Diagnostic and Interventional Radiology, Thoracic Clinic, University Hospital Heidelberg, Heidelberg, Germany.
  • 30 Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, CA, USA.
  • 31 Department of Internal Medicine, Division of Infectious Diseases, American University of Beirut Medical Center, Beirut, Lebanon.
  • 32 Department of Clinical Mycology, Allergology and Immunology, North Western State Medical University, St Petersburg, Russia.
  • 33 Division of Hygiene and Medical Microbiology, Department of Hygiene, Microbiology and Public Health, Medical University Innsbruck, Innsbruck, Austria.
  • 34 Infectious Diseases Service, Department of Medicine and Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland; Institute of Microbiology, Department of Laboratories, Lausanne University Hospital, Lausanne, Switzerland.
  • 35 Institut Pasteur, National Reference Center for Invasive Mycoses and Antifungals, Department of Mycology, Paris Descartes University, Necker-Enfants Malades University Hospital, Department of Infectious Diseases and Tropical Medicine, Centre d'Infectiologie Necker-Pasteur, Institut Imagine, AP-HP, Paris, France.
  • 36 Division of Infectious Diseases, Department of Internal Medicine, Catholic Hematology Hospital, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul, Korea.
  • 37 Division of Paediatric Haematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe-University, Frankfurt, Germany.
  • 38 School of Medicine and Pharmacy, University Mohammed the fifth, Hay Riad, Rabat, Morocco.
  • 39 Laboratory of Antimicrobial Chemotherapy, Ion Ionescu de la Brad University, Iaşi, Romania.
  • 40 Department of Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Potsdam, Germany.
  • 41 Department of Medical Microbiology and Infectious Diseases, Centre of Expertise in Mycology Radboudumc/Canisius Wilhelmina Hospital, Nijmegen, Netherlands.
  • 42 Clinical Microbiology Laboratory, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands.
  • 43 Department of Infectious Diseases, Alfred Health & Monash University, Melbourne, Australia.
  • 44 Department of Internal Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
  • 45 Department of Medical Microbiology & Parasitology, College of Medicine, University of Lagos, Yaba, Lagos, Nigeria; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
  • 46 Department of Hematology, Fondazione Policlinico Universitario A. Gemelli -IRCCS- Universita Cattolica del Sacro Cuore, Roma, Italy.
  • 47 Federal University of Health Sciences of Porto Alegre, Hospital Dom Vicente Scherer, Porto Alegre, Brazil.
  • 48 Infectious Diseases Clinic, Vedanta Institute of Medical Sciences, Navarangpura, Ahmeddabad, India.
  • 49 Institute of Hematology and Blood Transfusion, Prague, Czech Republic.
  • 50 UK NHS Mycology Reference Centre, Manchester University NHS Foundation Trust, Manchester, UK.
  • 51 Hämatologie & Internistische Onkologie, Lukas-Krankenhaus Bünde, Onkologische Ambulanz, Bünde, Germany.
  • 52 Department of Medical Mycology/Invasive Fungi Research Center (IFRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Center of Expertise in Microbiology, Infection Biology and Antimicrobial Pharmacology, Tehran, Iran; Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
  • 53 Department of Hemato Oncology, Amrita Institute of Medical Sciences, Amrita Viswa Vidyapeetham University, Kochi, India.
  • 54 Division of Infectious Diseases, University of Pittsburgh Medical Center and VA Pittsburgh Healthcare System, Infectious Diseases Section, University of Pittsburgh, Pittsburgh, PA, USA.
  • 55 Infectious Diseases Unit, Szent Istvan and Szent Laszlo Hospital, Budapest, Hungary.
  • 56 Department of Infectious Diseases, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
  • 57 University of Melbourne, Melbourne, VIC, Australia; The National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Parkville, Melbourne, VIC, Australia.
  • 58 P D Hinduja Hospital & Medical Research Centre, Department of Medicine, Veer Sarvarkar Marg, Mumbai, India.
  • 59 Los Angeles County and University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA.
  • 60 Division of Pediatric Infectious Diseases, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
  • 61 Department of Infectious Diseases, Singapore General Hospital, Singapur, Singapore.
  • 62 Department for Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
  • 63 Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF) partner site Bonn-Cologne, Cologne, Germany; Department of Internal Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt, Germany.
  • 64 Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF) partner site Bonn-Cologne, Cologne, Germany; Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt, Germany.
  • 65 Departments of Medicine, Pediatrics, Microbiology & Immunology, Weill Cornell Medicine, and New York Presbyterian Hospital, New York City, NY, USA.
  • 66 Public Health Wales Microbiology Cardiff, UHW, Heath Park, Cardiff, UK.
  • 67 Fungus Testing Laboratory, University of Texas Health Science Center, San Antonio, TX, USA.
  • 68 Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • 69 Department of Medical Microbiology, Postgraduate Institute of Medical Education & Research, Chandigarh, India.
  • PMID: 31699664
  • PMCID: PMC8559573
  • DOI: 10.1016/S1473-3099(19)30312-3

Mucormycosis is a difficult to diagnose rare disease with high morbidity and mortality. Diagnosis is often delayed, and disease tends to progress rapidly. Urgent surgical and medical intervention is lifesaving. Guidance on the complex multidisciplinary management has potential to improve prognosis, but approaches differ between health-care settings. From January, 2018, authors from 33 countries in all United Nations regions analysed the published evidence on mucormycosis management and provided consensus recommendations addressing differences between the regions of the world as part of the "One World One Guideline" initiative of the European Confederation of Medical Mycology (ECMM). Diagnostic management does not differ greatly between world regions. Upon suspicion of mucormycosis appropriate imaging is strongly recommended to document extent of disease and is followed by strongly recommended surgical intervention. First-line treatment with high-dose liposomal amphotericin B is strongly recommended, while intravenous isavuconazole and intravenous or delayed release tablet posaconazole are recommended with moderate strength. Both triazoles are strongly recommended salvage treatments. Amphotericin B deoxycholate is recommended against, because of substantial toxicity, but may be the only option in resource limited settings. Management of mucormycosis depends on recognising disease patterns and on early diagnosis. Limited availability of contemporary treatments burdens patients in low and middle income settings. Areas of uncertainty were identified and future research directions specified.

Copyright © 2019 Elsevier Ltd. All rights reserved.

PubMed Disclaimer

Figure 1:. Cutaneous and rhino-orbito-cerebral mucormycosis

(A) Extensive primary cutaneous mucormycosis of the left leg…

Figure 2:. Diagnostic pathway for mucormycosis

Depending on the geographical location not all recommended tests…

Figure 3:. Radiographic signs of mucormycosis

Four imaging signs can suggest pulmonary mucormycosis in an…

Figure 4:. Hyphal morphology in mucormycosis and…

Figure 4:. Hyphal morphology in mucormycosis and aspergillosis

(A) Typical hyphal morphology in mucormycosis lesions…

Figure 5:. Optimal treatment pathways for mucormycosis…

Figure 5:. Optimal treatment pathways for mucormycosis in adults

Depending on the geographical location not…

  • Pharmacoeconomic evaluation of isavuconazole, posaconazole, and voriconazole for the treatment of invasive mold diseases in hematological patients: initial therapy prior to pathogen differential diagnosis in China. Han G, Xu Q, Lv Q, Li X, Shi X. Han G, et al. Front Public Health. 2023 Dec 19;11:1292162. doi: 10.3389/fpubh.2023.1292162. eCollection 2023. Front Public Health. 2023. PMID: 38179563 Free PMC article.

Similar articles

  • ESCMID and ECMM joint clinical guidelines for the diagnosis and management of mucormycosis 2013. Cornely OA, Arikan-Akdagli S, Dannaoui E, Groll AH, Lagrou K, Chakrabarti A, Lanternier F, Pagano L, Skiada A, Akova M, Arendrup MC, Boekhout T, Chowdhary A, Cuenca-Estrella M, Freiberger T, Guinea J, Guarro J, de Hoog S, Hope W, Johnson E, Kathuria S, Lackner M, Lass-Flörl C, Lortholary O, Meis JF, Meletiadis J, Muñoz P, Richardson M, Roilides E, Tortorano AM, Ullmann AJ, van Diepeningen A, Verweij P, Petrikkos G; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group; European Confederation of Medical Mycology. Cornely OA, et al. Clin Microbiol Infect. 2014 Apr;20 Suppl 3:5-26. doi: 10.1111/1469-0691.12371. Clin Microbiol Infect. 2014. PMID: 24479848
  • [Expert consensus on diagnosis and treatment of severe COVID-19 associated pulmonary aspergillosis and mucormycosis]. Critical Care Medicine Group of Chinese Association of Chest Physicians; Chinese Thoracic Society. Critical Care Medicine Group of Chinese Association of Chest Physicians, et al. Zhonghua Jie He He Hu Xi Za Zhi. 2024 Jan 12;47(1):10-23. doi: 10.3760/cma.j.cn112147-20230823-00098. Zhonghua Jie He He Hu Xi Za Zhi. 2024. PMID: 38062689 Chinese.
  • Challenges in the diagnosis and treatment of mucormycosis. Skiada A, Lass-Floerl C, Klimko N, Ibrahim A, Roilides E, Petrikkos G. Skiada A, et al. Med Mycol. 2018 Apr 1;56(suppl_1):93-101. doi: 10.1093/mmy/myx101. Med Mycol. 2018. PMID: 29538730 Free PMC article. Review.
  • ECMM/ISHAM recommendations for clinical management of COVID-19 associated mucormycosis in low- and middle-income countries. Rudramurthy SM, Hoenigl M, Meis JF, Cornely OA, Muthu V, Gangneux JP, Perfect J, Chakrabarti A; ECMM and ISHAM. Rudramurthy SM, et al. Mycoses. 2021 Sep;64(9):1028-1037. doi: 10.1111/myc.13335. Epub 2021 Jul 26. Mycoses. 2021. PMID: 34133816 Free PMC article.
  • Mucormycosis: New Developments into a Persistently Devastating Infection. Danion F, Aguilar C, Catherinot E, Alanio A, DeWolf S, Lortholary O, Lanternier F. Danion F, et al. Semin Respir Crit Care Med. 2015 Oct;36(5):692-705. doi: 10.1055/s-0035-1562896. Epub 2015 Sep 23. Semin Respir Crit Care Med. 2015. PMID: 26398536 Review.
  • Risk factors and clinical presentation of rhino-orbital mucormycosis: Lesson learnt during Covid pandemic. Kusumesh R, Singh V, Sinha S, Ali MS, Mishra SK, Ambasta A, Prakash V, Sinha P, Sinha BP, Priyadarshi RN. Kusumesh R, et al. J Family Med Prim Care. 2024 Apr;13(4):1354-1361. doi: 10.4103/jfmpc.jfmpc_1259_23. Epub 2024 Apr 22. J Family Med Prim Care. 2024. PMID: 38827672 Free PMC article.
  • Insights into invasive fungal infection diagnostic and treatment capacities in tertiary care centres of Germany. Salmanton-García J, Simon M, Groll AH, Kurzai O, Lahmer T, Lehrnbecher T, Schroeder M, Cornely OA, Stemler J. Salmanton-García J, et al. JAC Antimicrob Resist. 2024 May 29;6(3):dlae083. doi: 10.1093/jacamr/dlae083. eCollection 2024 Jun. JAC Antimicrob Resist. 2024. PMID: 38812581 Free PMC article.
  • In vitro antifungal activity of MMV Pathogen Box® compounds alone or in combination with antifungal drugs against mucormycosis agents. Almeida-Silva F, Tenório-Alvarenga PH, da Costa RV, Coelho RA, de Sousa Araújo GR, Zancopé-Oliveira RM, Frases S, Almeida-Paes R. Almeida-Silva F, et al. Curr Res Microb Sci. 2024 May 15;6:100242. doi: 10.1016/j.crmicr.2024.100242. eCollection 2024. Curr Res Microb Sci. 2024. PMID: 38799088 Free PMC article.
  • Humans vs. Fungi: An Overview of Fungal Pathogens against Humans. Thambugala KM, Daranagama DA, Tennakoon DS, Jayatunga DPW, Hongsanan S, Xie N. Thambugala KM, et al. Pathogens. 2024 May 17;13(5):426. doi: 10.3390/pathogens13050426. Pathogens. 2024. PMID: 38787278 Free PMC article. Review.
  • Corona-Associated Mucormycosis: Case Series Reports and Review of the Literature. Fitero A, Negrut N, John HT, Ferician A, Marian P. Fitero A, et al. J Fungi (Basel). 2024 Apr 24;10(5):305. doi: 10.3390/jof10050305. J Fungi (Basel). 2024. PMID: 38786660 Free PMC article.
  • Chamilos G, Lewis RE, Kontoyiannis DP. Delaying amphotericin B-based frontline therapy significantly increases mortality among patients with hematologic malignancy who have zygomycosis. Clin Infect Dis 2008; 47: 503–09. - PubMed
  • Vaughan C, Bartolo A, Vallabh N, Leong SC. A meta-analysis of survival factors in rhino-orbital-cerebral mucormycosis—has anything changed in the past 20 years? Clin Otolaryngol 2018; 43: 1454–64. - PubMed
  • Sun HY, Singh N. Mucormycosis: its contemporary face and management strategies. Lancet Infect Dis 2011; 11: 301–11. - PubMed
  • Tissot F, Agrawal S, Pagano L, et al. ECIL-6 guidelines for the treatment of invasive candidiasis, aspergillosis and mucormycosis in leukemia and hematopoietic stem cell transplant patients. Haematologica 2017; 102: 433–44. - PMC - PubMed
  • Kung HC, Huang PY, Chen WT, et al. 2016 guidelines for the use of antifungal agents in patients with invasive fungal diseases in Taiwan. J Microbiol Immunol Infect 2018; 51: 1–17. - PubMed

Publication types

  • Search in MeSH

Related information

  • Cited in Books

Grants and funding

  • R01 AI063503/AI/NIAID NIH HHS/United States
  • R01 AI117211/AI/NIAID NIH HHS/United States
  • R21 AI132923/AI/NIAID NIH HHS/United States

LinkOut - more resources

Full text sources.

  • Elsevier Science
  • Europe PubMed Central
  • PubMed Central
  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Generative AI Can Harm Learning

59 Pages Posted: 18 Jul 2024

Hamsa Bastani

University of Pennsylvania - The Wharton School

Osbert Bastani

University of Pennsylvania - Department of Computer and Information Science

Özge Kabakcı

International Business School - Budapest (IBS)

Rei Mariman

Independent; Independent

Date Written: July 15, 2024

Generative artificial intelligence (AI) is poised to revolutionize how humans work, and has already demonstrated promise in significantly improving human productivity. However, a key remaining question is how generative AI affects learning , namely, how humans acquire new skills as they perform tasks. This kind of skill learning is critical to long-term productivity gains, especially in domains where generative AI is fallible and human experts must check its outputs. We study the impact of generative AI, specifically OpenAI's GPT-4, on human learning in the context of math classes at a high school. In a field experiment involving nearly a thousand students, we have deployed and evaluated two GPT based tutors, one that mimics a standard ChatGPT interface (called GPT Base) and one with prompts designed to safeguard learning (called GPT Tutor). These tutors comprise about 15% of the curriculum in each of three grades. Consistent with prior work, our results show that access to GPT-4 significantly improves performance (48% improvement for GPT Base and 127% for GPT Tutor). However, we additionally find that when access is subsequently taken away, students actually perform worse than those who never had access (17% reduction for GPT Base). That is, access to GPT-4 can harm educational outcomes. These negative learning effects are largely mitigated by the safeguards included in GPT Tutor. Our results suggest that students attempt to use GPT-4 as a "crutch" during practice problem sessions, and when successful, perform worse on their own. Thus, to maintain long-term productivity, we must be cautious when deploying generative AI to ensure humans continue to learn critical skills. * HB, OB, and AS contributed equally

Keywords: Generative AI, Human Capital Development, Education, Human-AI Collaboration, Large Language Models

Suggested Citation: Suggested Citation

University of Pennsylvania - The Wharton School ( email )

3641 Locust Walk Philadelphia, PA 19104-6365 United States

University of Pennsylvania - Department of Computer and Information Science ( email )

3330 Walnut Street Philadelphia, PA 19104 United States

Alp Sungu (Contact Author)

International business school - budapest (ibs) ( email ), independent ( email ), do you have a job opening that you would like to promote on ssrn, paper statistics, related ejournals, econometrics: computer programs & software ejournal.

Subscribe to this fee journal for more curated articles on this topic

Randomized Social Experiments eJournal

Environment for innovation ejournal, generative ai ejournal.

Subscribe to this free journal for more curated articles on this topic

Mucormycosis Research: A global outlook through bibliometric approaches

  • Iberoamerican Journal of Science Measurement and Communication 3(1)
  • CC BY-NC 4.0

Shri Ram at Thapar Institute of Engineering & Technology

  • Thapar Institute of Engineering & Technology

Hemant Sharma at Sikkim University

  • Sikkim University

Arun Kumar Rai at Sikkim University

Discover the world's research

  • 25+ million members
  • 160+ million publication pages
  • 2.3+ billion citations

Devi Dayal

  • Carlos Alberto Gómez Cano

Javier Gonzalez-Argote

  • Gargee Dubey

Payam Tabarsi

  • Elham Askari

Francisco López-Muñoz

  • Robert N. Weinreb

Sasan Moghimi

  • J MYCOL MED

Tarun Kumar Suvvari

  • MYCOPATHOLOGIA
  • Deepak Garg

Valliappan Muthu

  • Tanmaya Kataria
  • OPHTHAL PLAST RECONS

Zesemayat K. Mekonnen

  • Davin C. Ashraf
  • Tyler Jankowski

Bryan Winn

  • Recruit researchers
  • Join for free
  • Login Email Tip: Most researchers use their institutional email address as their ResearchGate login Password Forgot password? Keep me logged in Log in or Continue with Google Welcome back! Please log in. Email · Hint Tip: Most researchers use their institutional email address as their ResearchGate login Password Forgot password? Keep me logged in Log in or Continue with Google No account? Sign up

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 08 July 2024

Pathogenicity and transmissibility of bovine H5N1 influenza virus

  • Amie J. Eisfeld 1   na1 ,
  • Asim Biswas 1   na1 ,
  • Lizheng Guan 1   na1 ,
  • Chunyang Gu 1   na1 ,
  • Tadashi Maemura 1   na1 ,
  • Sanja Trifkovic 1 ,
  • Tong Wang 1 ,
  • Lavanya Babujee 1 ,
  • Randall Dahn 1 ,
  • Peter J. Halfmann   ORCID: orcid.org/0000-0002-1648-1625 1 ,
  • Tera Barnhardt 2 ,
  • Gabriele Neumann 1 ,
  • Yasuo Suzuki 3 ,
  • Alexis Thompson   ORCID: orcid.org/0000-0003-4405-5313 4 ,
  • Amy K. Swinford 5 ,
  • Kiril M. Dimitrov   ORCID: orcid.org/0000-0002-5525-4492 5 ,
  • Keith Poulsen 6 &
  • Yoshihiro Kawaoka   ORCID: orcid.org/0000-0001-5061-8296 1 , 7 , 8 , 9  

Nature ( 2024 ) Cite this article

13k Accesses

1696 Altmetric

Metrics details

We are providing an unedited version of this manuscript to give early access to its findings. Before final publication, the manuscript will undergo further editing. Please note there may be errors present which affect the content, and all legal disclaimers apply.

  • Influenza virus
  • Viral pathogenesis
  • Viral transmission

Highly pathogenic H5N1 avian influenza (HPAI H5N1) viruses occasionally infect, but typically do not transmit, in mammals. In the Spring of 2024, an unprecedented outbreak of HPAI H5N1 in bovine herds occurred in the US, with virus spread within and between herds, infections in poultry and cats, and spillover into humans, collectively indicating an increased public health risk 1-4 . Here, we characterized an HPAI H5N1 virus isolated from infected cow milk in mice and ferrets. Like other HPAI H5N1 viruses, the bovine H5N1 virus spread systemically, including to the mammary glands of both species; however, this tropism was also observed for an older HPAI H5N1 virus isolate. Importantly, bovine HPAI H5N1 virus bound to sialic acids expressed in human upper airways and inefficiently transmitted to exposed ferrets (one of four exposed ferrets seroconverted without virus detection). Bovine HPAI H5N1 virus thus possesses features that may facilitate infection and transmission in mammals.

You have full access to this article via your institution.

Similar content being viewed by others

mucormycosis research paper pdf

Characterization of highly pathogenic avian influenza H5Nx viruses in the ferret model

mucormycosis research paper pdf

Potential pandemic risk of circulating swine H1N2 influenza viruses

mucormycosis research paper pdf

Risk assessment of a highly pathogenic H5N1 influenza virus from mink

Author information.

These authors contributed equally: Amie J. Eisfeld, Asim Biswas, Lizheng Guan, Chunyang Gu, Tadashi Maemura

Authors and Affiliations

Influenza Research Institute, Dept. of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA

Amie J. Eisfeld, Asim Biswas, Lizheng Guan, Chunyang Gu, Tadashi Maemura, Sanja Trifkovic, Tong Wang, Lavanya Babujee, Randall Dahn, Peter J. Halfmann, Gabriele Neumann & Yoshihiro Kawaoka

Heritage Vet Partners, Johnson, KS, USA

Tera Barnhardt

Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan

Yasuo Suzuki

Texas A&M Veterinary Medical Diagnostic Laboratory, Canyon, TX, USA

Alexis Thompson

Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, USA

Amy K. Swinford & Kiril M. Dimitrov

Wisconsin Veterinary Diagnostic Laboratory, University of Wisconsin-Madison, Madison, WI, USA

Keith Poulsen

Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan

Yoshihiro Kawaoka

The University of Tokyo Pandemic Preparedness, Infection and Advanced research center (UTOPIA), University of Tokyo, Tokyo, Japan

The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Yoshihiro Kawaoka .

Supplementary information

Reporting summary, rights and permissions.

Reprints and permissions

About this article

Cite this article.

Eisfeld, A.J., Biswas, A., Guan, L. et al. Pathogenicity and transmissibility of bovine H5N1 influenza virus. Nature (2024). https://doi.org/10.1038/s41586-024-07766-6

Download citation

Received : 01 June 2024

Accepted : 28 June 2024

Published : 08 July 2024

DOI : https://doi.org/10.1038/s41586-024-07766-6

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

By submitting a comment you agree to abide by our Terms and Community Guidelines . If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

mucormycosis research paper pdf

Cookies on GOV.UK

We use some essential cookies to make this website work.

We’d like to set additional cookies to understand how you use GOV.UK, remember your settings and improve government services.

We also use cookies set by other sites to help us deliver content from their services.

You have accepted additional cookies. You can change your cookie settings at any time.

You have rejected additional cookies. You can change your cookie settings at any time.

King's Speech 2024: background briefing notes

Read the briefing notes on the announcements made in the 2024 King’s Speech.

The King's Speech 2024: background briefing notes

PDF , 556 KB , 104 pages

This file may not be suitable for users of assistive technology.

Updates to this page

The attachment on this page has been updated.

First published.

Sign up for emails or print this page

Is this page useful.

  • Yes this page is useful
  • No this page is not useful

Help us improve GOV.UK

Don’t include personal or financial information like your National Insurance number or credit card details.

To help us improve GOV.UK, we’d like to know more about your visit today. Please fill in this survey (opens in a new tab) .

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Elsevier - PMC COVID-19 Collection

Logo of pheelsevier

Mucormycosis in COVID-19: A systematic review of cases reported worldwide and in India

Awadhesh kumar singh.

a Department of Diabetes & Endocrinology, G. D Hospital & Diabetes Institute, Kolkata, West Bengal, India

Shashank R. Joshi

b Department of Diabetes & Endocrinology, Lilavati Hospital & Joshi Clinic, Mumbai, Maharashtra, India

Anoop Misra

c Fortis C-DOC Hospital for Diabetes & Allied Sciences, New Delhi, India

d National Diabetes, Obesity and Cholesterol Foundation, New Delhi, India

e Diabetes Foundation (India), New Delhi, India

Background and aims

There are increasing case reports of rhino-orbital mucormycosis in people with coronavirus disease 2019 (COVID-19), especially from India. Diabetes mellitus (DM) is an independent risk factor for both severe COVID-19 and mucormycosis. We aim to conduct a systematic review of literature to find out the patient's characteristics having mucormycosis and COVID-19.

We searched the electronic database of PubMed and Google Scholar from inception until May 13, 2021 using keywords. We retrieved all the granular details of case reports/series of patients with mucormycosis, and COVID-19 reported world-wide. Subsequently we analyzed the patient characteristics, associated comorbidities, location of mucormycosis, use of steroids and its outcome in people with COVID-19.

Overall, 101 cases of mucormycosis in people with COVID-19 have been reported, of which 82 cases were from India and 19 from the rest of the world. Mucormycosis was predominantly seen in males (78.9%), both in people who were active (59.4%) or recovered (40.6%) from COVID-19. Pre-existing diabetes mellitus (DM) was present in 80% of cases, while concomitant diabetic ketoacidosis (DKA) was present in 14.9%. Corticosteroid intake for the treatment of COVID-19 was recorded in 76.3% of cases. Mucormycosis involving nose and sinuses (88.9%) was most common followed by rhino-orbital (56.7%). Mortality was noted in 30.7% of the cases.

An unholy trinity of diabetes, rampant use of corticosteroid in a background of COVID-19 appears to increase mucormycosis. All efforts should be made to maintain optimal glucose and only judicious use of corticosteroids in patients with COVID-19.

1. Introduction

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been associated with a wide range of opportunistic bacterial and fungal infections [ 1 ]. Both Aspergill us and Candida have been reported as the main fungal pathogens for co-infection in people with COVID-19 [ 2 ]. Recently, several cases of mucormycosis in people with COVID-19 have been increasingly reported world-wide, in particular from India. The primary reason that appears to be facilitating Mucorales spores to germinate in people with COVID-19 is an ideal environment of low oxygen (hypoxia), high glucose (diabetes, new-onset hyperglycemia, steroid-induced hyperglycemia), acidic medium (metabolic acidosis, diabetic ketoacidosis [DKA]), high iron levels (increased ferritins) and decreased phagocytic activity of white blood cells (WBC) due to immunosuppression (SARS-CoV-2 mediated, steroid-mediated or background comorbidities) coupled with several other shared risk factors including prolonged hospitalization with or without mechanical ventilators.

Phycomycosis or zygomycosis was first described in 1885 by Paltauf [ 3 ] and later coined as Mucormycosis in 1957 by Baker [ 4 ] an American pathologist for an aggressive infection caused by Rhizopus . Mucormycosis is an uncommon but a fatal fungal infection that usually affects patients with altered immunity. Mucormycosis is an angioinvasive disease caused by mold fungi of the genus Rhizopus , Mucor , Rhizomucor, Cunninghamella and Absidia of Order- Mucorales, Class- Zygomycetes [ 5 ]. The Rhizopus Oryzae is most common type and responsible for nearly 60% of mucormycosis cases in humans and also accounts for 90% of the Rhino-orbital-cerebral (ROCM) form [ 6 ]. Mode of contamination occurs through the inhalation of fungal spores.

Globally, the prevalence of mucormycosis varied from 0.005 to 1.7 per million population, while its prevalence is nearly 80 times higher (0.14 per 1000) in India compared to developed countries, in a recent estimate of year 2019–2020 [ [7] , [8] , [9] ]. In other words, India has highest cases of the mucormycosis in the world. Notwithstanding, India is already having second largest population with diabetes mellitus (DM) and was the diabetes capital of the world, until recently [ 10 ]. Importantly, DM has been the most common risk factor linked with mucormycosis in India, although hematological malignancies and organ transplant takes the lead in Europe and the USA [ 9 ]. Nevertheless, DM remains the leading risk factor associated with mucormycosis globally, with an overall mortality of 46% [ 11 ]. Indeed, presence of DM was an independent risk factor (Odds ratio [OR] 2.69; 95% Confidence Interval 1.77–3.54; P < 0.001) in a large 2018 meta-analysis of 851 cases of rarely occurring mucormycosis, and the most common species isolated was Rhizopus (48%) [ 11 ]. While long term use of corticosteroids have often been associated with several opportunistic fungal infection including aspergillosis and mucormycosis, even a short course of corticosteroids has recently been reported to link with mucormycosis especially in people with DM. A cumulative prednisone dose of greater than 600 mg or a total methyl prednisone dose of 2–7 g given during the month before, predisposes immunocompromised people to mucormycosis [ 12 ]. There are few case reports of mucormycosis resulting from even a short course (5–14 days) of steroid therapy, especially in people with DM [ 13 ]. Surprisingly, 46% of the patients had received corticosteroids within the month before the diagnosis of mucormycosis in the European Confederation of Medical Mycology study [ 14 ].

These findings need a relook in the context of COVID-19 pandemic where corticosteroids are often being used. There has been a steep rise in case reports/series of mucormycosis in people with COVID-19 especially from India. Similarly, many cases are being reported from other parts of globe. Several anecdotal cases are also being reported in grey literature such as the print and electronic media. These finding are unprecedented and carry an immense public health importance, primarily because fatality rate with mucormycosis is pretty high. Especially the intracranial involvement of mucormycosis increases the fatality rate to as high as 90% [ 15 ]. Moreover, rapidity of dissemination of mucormycosis is an extraordinary phenomenon and even a delay of 12 h in the diagnosis could be fatal, the reason 50% of cases of mucormycosis have been historically diagnosed only in the post-mortem autopsy series [ 16 ]. This prompted us to conduct a systematic review of published case reports/series of mucormycosis in people with COVID-19, to know its temporal associations in relation to comorbidities, association with drugs being used in COVID-19 and overall characteristics of patients with its outcome. We additionally postulated a mechanistic explanation as to why mucormycosis could be increasingly linked to COVID-19 and is being reported increasingly from India.

A systematic literature search was conducted in the electronic database of PubMed and Google Scholar from inception until May 13, 2021 using keyword “COVID-19”, “SARS CoV-2”, AND “Mucormycosis”, “Zygomycosis”, “Phycomycosis, “Mucorales”, “Mucor”, “Rhizopus”, “Rhizomucor”, “Cunninghamella”, and “Absidia”. Details of all the cases that reported mucormycosis (both confirmed and suspected) in people with COVID-19 so far, were retrieved. Characteristics of each patient was collected on excel sheet and analyzed on various endpoints and outcomes. Two authors independently checked the veracity of data.

Overall, 28 articles were found to report the original case(s) from the database of PubMed (24/28) and Google Scholar (4/28) [ [17] , [18] , [19] , [20] , [21] , [22] , [23] , [24] , [25] , [26] , [27] , [28] , [29] , [30] , [31] , [32] , [33] , [34] , [35] , [36] , [37] , [38] , [39] , [40] , [41] , [42] , [43] , [44] ]. A total of 101 cases of mucormycosis (including confirmed [95/101] and suspected [6/101]) in people with confirmed (RT-PCR diagnosis) COVID-19 were retrieved ( Table 1 ). Largely, 82 cases (81.2%) of mucormycosis in patients with COVID-19 were reported from India, followed by 9 cases (8.9%) from USA and 3 cases (3.1%) from Iran. Only 19 (18.8%) cases as of now were reported from other parts of the world. One study by Satish et al. [ 25 ] that reported 11 case-series of mucormycosis in people with COVID-19 from India lacked granular detail of every patient and therefore excluded from some of the analysis. Pooled data from this study showed mucormycosis was predominantly seen in males (78.9%), both in people who were active (59.4%) or recovered (40.6%) from COVID-19. Recovered COVID-19 was defined as those who were either discharged from hospital or in-hospital but 2-weeks had passed post-detection, although there was evident overlap across the cases. Hyperglycemia at presentation (due to pre-existing DM or new-onset hyperglycemia or new-onset diabetes or diabetic ketoacidosis [DKA]) was the single most important risk factor observed in majority of cases (83.3%) of mucormycosis in people with COVID-19, followed by cancer (3.0%). Pre-existing DM accounted for 80% of cases, while concomitant DKA was present in nearly 15% of people with mucormycosis and COVID-19. History of corticosteroid intake for the treatment of COVID-19 was present in 76.3% of cases, followed by remdesivir (20.6%) and tocilizumab (4.1%). Commonest organ involved with mucormycosis was nose and sinus (88.9%), followed by rhino-orbital (56.7%) and ROCM type (22.2%). Overall mortality was noted in 30.7% of the cases. Table 2 summarizes the findings from 101 cases of mucormycosis in people with COVID-19.

Table 1

Mucormycosis in COVID-19 – Summary of 101 cases reported world-wide till May’ 2021.

First author Place (of report) N Age, range, M/F Comorbidities Confirmed/Suspected COVID-19 (Active/Recovered)Treatment received for COVID-19 Confirmed/Suspected MucorLocation of mucormycosis Outcome
DMCancerSteroidTocilizumabRemdesivirNasal/SinusOrbitCNSBoneLungGIT
Mehta et al. Mumbai160, MYNConfirm, AYYNConfirmYYNNNNDeath
Garg et al. Chandigarh155, MYNConfirm, AYNYConfirmNNNNYNImproving
Maini et al. Mumbai138, MNNConfirm, RYNYConfirmYYNNNNImproved
Saldanha et al. Mangalore132, FYNConfirm, ANRNRNRConfirmYYNNNNImproved
Revannavar et al. Mangalore1Middle age, FY, NDDNConfirm, ANNNConfirmYYYNNNImproving
Sen et al. Mumbai646.2–73.9, M: 6Y: AllNConfirm,
A: 1
R: 5
Y: 5
N: 1
NNConfirm: 5,
Suspect: 1
Y: AllY: AllY: 5
N: 1
NNNImproving
Sarkar et al. Puducherry1027-67,
M: 8
F: 2
Y: All,
DKA: 9
NConfirm, A: 10Y: 10NY: 5
N: 5
Confirm: 6, Suspect: 4Y: AllY: AllY: 1NNNDeath: 4,
Improved: 2,
Unchanged: 4
Mishra et al. Bangalore1037-78,
M: 9
F: 1
Y: 8
N: 2
NConfirm,
A: 10
Y: 6
N: 4
Y: 1
N: 9
Y: 6
N: 4
Confirm: AllY: AllY: 2NY: 1NNDeath: 4
Improved: 5
LFU: 1
Satish et al. Bangalore1130-74,
M: NR
F: NR
Y: MajorityY: 1 (Leukemia)Confirm,
A: 11
NNNConfirm: AllY: MajorityY: MajorityY: NRNNNDeath: 2
LAMA: 5
Improving: 4
Moorthy et al. Bangalore1739-73,
M: 15
F: 2
Y: 15
N: 2
NConfirm,
A: 4
R: 13
Y: 15
N: 2
NNConfirm: AllY: AllY: 11
N: 6
Y: 8
N: 9
Y: 14
N: 3
NNDeath: 7
Alive: 9
LFU: 1
Sharma et al. Jaipur 23 NR
M: 15
F: 8
Y: 21
N: 2
N Confirm,
A: 4
R: 19
Y: All N N Confirm: All Y: All Y: 10 Y: 2 N N N Death: 0
LFU: 2
Alive: 21
Hanley et al. UK122, MNNConfirm, ANRNRNRConfirm: AutopsyNNNNYNAutopsy report
Dallalzadeh et al. USA236, M
48, M
Y:2
DKA: 2
NConfirm,
A: 2
Y:2NY:2Confirm: 1
Suspected: 1
YYYNNNDeath: 1
Unchanged: 1
Werthman-E et al. USA133, FN, DKANConfirm, ANNNConfirmYYNNNNImproving
Placik et al. USA149, MNNConfirm, AYYYConfirmNNNNYNDeath
Mekkonen et al. USA160, M,T1DMNConfirm, AYNYConfirmYYNNNNDeath
Alekseyev et al. USA141, MT1DM, DKANConfirm, AYNNConfirmYNYNNNRecovered
Johnson et al. USA179, MYNConfirm, AYNYConfirm,
AF +
NNNNYNImproving
Kanwar et al. USA156, MNNConfirm, AYYNConfirmNNNNYNDeath
Khatri et al. USA168, MYN, (HT)Confirm, RYNNConfirmNNNN,
Skin
NNDeath
Monte Junior et al. Brazil186, MNNConfirm, ANNNConfirmNNNNNYDeath
Pasero et al. Italy166, MNNConfirm, ANNNConfirmYNNNYNDeath
Bellanger et al. France155, MNY, (Lymphoma)Confirm, ANNNConfirm,
AF +
NNNNYNDeath
Karimi-G et al. Iran161, MN, NODNConfirm, RYNYConfirmYYNNNNImproving
Veisi et al. Iran240, F:1;
54, M:1
N: 1
Y:1
NConfirm,
A: 2
Y: 2N: 2Y: 2Confirm, AllY: 2Y: 2Y: 1
N: 1
NNNDeath: 1
Recovered: 1
Sargin et al. Turkey156, FY, DKANConfirm, RYNNConfirmYYYNNNDeath
Waizel-H et al. Mexico124, FN, DKANConfirm, ANNNConfirmYYNNNNDeath
Zurl et al. Austria153, MNY, (Leukemia)Confirm, ANNNConfirm, AutopsyNNNNYNDeath

DM: Diabetes mellitus, CNS: Central nervous system, GIT: Gastro-intestinal tract, M: Male, F: Female, T1DM: Type 1 diabetes mellitus, DKA: Diabetic ketoacidosis, NOD: New-onset diabetes, NDD: Newly detected diabetes, A: Active COVID-19, R: Recovered COVID-19, Y: Yes, N: No, HT: Heart transplant, AF: Aspergillosis fungi, LFU: Lost to follow-up, LAMA: Left against medical advice.

Table 2

Characteristics of 101 patients of mucormycosis with COVID-19.

Confirmed mucormycosis, N = 101n, (%)Remarks and limitations
Country reported (Published)India82 (81.2)Highest cases reported from India. ≈ denotes nearest rounded of value.
USA9 (8.9)
Iran3 (≈3.0)
UK1 (≈1.0)
France1 (≈1.0)
Italy1 (≈1.0)
Brazil1 (≈1.0)
Turkey1 (≈1.0)
Mexico1 (≈1.0)
Austria 1 (≈1.0)
Age (Years) Range 22-86
SexMale71/90 (78.9)More commonly observed in males.
Female 19/90 (21.1)
COVID-19 statusActive60/101 (59.4)Exact definition of active and recovered cases of COVID-19 was different and not unanimous.
Recovered 41/101 (40.6)
Risk factorsHyperglycemia at presentation75/90 (83.3)No unanimous definition of hyperglycemia.
Malignancy3/101 (3.0)2 Leukemia, 1 Lymphoma
Post-transplant 1/101 (1.0) 1 Heart transplant
Hyperglycemia at presentationPre-existing DM72/90 (80.0)Unless reported as insulin-dependent or type 1 diabetes, all cases were assumed as type 2 diabetes. Lack of baseline HbA1c data and duration of diabetes for majority of DM patients.
Types of DM
Type 2 diabetes70/72 (97.2)
Type 1 diabetes2/72 (2.8)
New-onset DM/hyperglycemia2/90 (2.2)
Presented with DKA 15/101 (14.9)
Treatment history of COVID-19Steroid74/97 (76.3)Few cases were received all 3 drugs for COVID-19.
Tocilizumab4/97 (4.1)
Remdesivir 20/97 (20.6)
MucormycosisConfirmed95/101 (94.1)Confirmed denotes microbiological or histopathological diagnosis.
Suspected 6/101 (5.9)
Location of mucormycosisNasal/Sinus80/90 (88.9)There appears to have an overlap between Nasal/Sinus only and Rhino-orbital variety.
Rhino-orbital51/90 (56.7)
Rhino-orbito-cerebral20/90 (22.2)
Bone involvement15/101 (14.9)
Pulmonary8/101 (7.9)
Gastrointestinal1/101 (1.0)
Cutaneous 1/101 (1.0)
OutcomesAlive (Improved/Improving)56/101 (55.4)Outcomes is difficult to assess considering that several cases were still under in-hospital treatment and their final outcome are not yet known.
Unchanged5/101 (5.0)
Death31/101 (30.7)
Status unknown (LFU, LAMA)9/101 (8.9)

DM: Diabetes mellitus, DKA: Diabetic ketoacidosis, LFU: Lost to follow-up, LAMA: Left against medical advice.

4. Discussion

Although mucormycosis is an extremely rare in healthy individuals but several immunocompromised conditions predispose it. This includes uncontrolled DM with or without DKA, hematological and other malignancies, organ transplantation, prolonged neutropenia, immunosuppressive and corticosteroid therapy, iron overload or hemochromatosis, deferoxamine or desferrioxamine therapy, voriconazole prohylaxis for transplant recipients, severe burns, acquired immunodeficiency syndrome (AIDS), intravenous drug abusers, malnutrition and open wound following trauma [ 45 ]. Mucormycosis can involve nose, sinuses, orbit, central nervous system (CNS), lung (pulmonary), gastrointestinal tract (GIT), skin, jaw bones, joints, heart, kidney, and mediastinum (invasive type), but ROCM is the commonest variety seen in clinical practice world-wide [ 45 ]. It should be noted that term ROCM refers to the entire spectrum ranging from limited sino-nasal disease (sino-nasal tissue invasion), limited rhino-orbital disease (progression to orbits) to rhino-orbital-cerebral disease (CNS involvement) [ 46 ]. The area of involvement may differ due to underlying condition. For example, ROCM is frequently observed in association with uncontrolled diabetes and DKA, whereas pulmonary involvement is often observed in patients having neutropenia, bone marrow and organ transplant, and hematological malignancies, while GIT gets involved more in malnourished individuals. Giant cell invasion, thrombosis and eosinophilic necrosis of the underlying tissue is the pathological hallmark of mucormycosis. Microbiological identification of the hyphae based on diameter, presence or absence of septa, branching angle (right or acute branching), and pigmentation, differentiates it from other fungal infections. The 1950 Smith and Krichner [ 47 ] criteria for the clinical diagnosis of mucormycosis are still considered to be gold standard and include:

  • (i) Black, necrotic turbinate's easily mistaken for dried, crusted blood,
  • (ii) Blood-tinged nasal discharge and facial pain, both on the same side,
  • (iii) Soft peri-orbital or peri-nasal swelling with discoloration and induration,
  • (iv) Ptosis of the eyelid, proptosis of the eyeball and complete ophthalmoplegia and, ( v ) Multiple cranial nerve palsies unrelated to documented lesions.

A 2019 nationwide multi-center study of 388 confirmed or suspected cases of mucormycosis in India prior to COVID-19, Prakash et al. found that 18% had DKA and 57% of patients had uncontrolled DM [ 48 ]. Similarly, in a data of 465 cases of mucormycosis without COVID-19 in India, Patel et al. [ 49 ] has shown that rhino-orbital presentation was the most common (67.7%), followed by pulmonary (13.3%) and cutaneous type (10.5%). The predisposing factors associated with mucormycosis in Indians include DM (73.5%), malignancy (9.0%) and organ transplantation (7.7%) [ 49 ]. Presence of DM significantly increases the odds of contracting ROCM by 7.5-fold (Odds ratio 7.55, P = 0.001) as shown in a prospective Indian study, prior to COVID-19 pandemic [ 50 ]. In a recent systematic review conducted until April 9, 2021 by John et al. [ 51 ] that reported the findings of 41 confirmed mucormycosis cases in people with COVID-19, DM was reported in 93% of cases, while 88% were receiving corticosteroids. These findings are consistent with our findings of even larger case series of 101 mucormycosis cases (95 confirmed and 6 suspected) in Covid-19, where 80% cases had DM, and more than two-third (76.3%) received a course of corticosteroids. Collectively, these findings suggest a familiar connection of mucormycosis, diabetes and steroid, in people with COVID-19.

Since there are no studies that compared patients of mucormycosis in non-diabetic COVID-19 who did not receive steroids versus COVID-19 patients who received steroids and developed mucormycosis, it is difficult to establish a causal effect relationship between COVID-19 and mucormycosis in relation to corticosteroids. Nonetheless, there appears to be a number of triggers that may precipitate mucormycosis in people with COVID-19 in relation to corticosteroids:

  • (i) Presence of DM with or without DKA increases the risk of contracting mucormycosis and DM is often associated with an increased severity of COVID-19,
  • (ii) Uncontrolled hyperglycemia and precipitation of DKA is often observed due to corticosteroid intake. Low pH due to acidosis is a fertile media for mucor spores to germinate. Moreover, steroid use reduces the phagocytic activity of WBC (both first line and second line defense mechanism), causes impairment of bronchoalveolar macrophages migration, ingestion, and phagolysosome fusion, making a diabetic patient exceptionally vulnerable to mucormycosis.
  • (iii) COVID-19 often causes endothelialitis, endothelial damage, thrombosis, lymphopenia, and reduction in CD4 + and CD8 + T-cell level and thus predisposes to secondary or opportunistic fungal infection,
  • (iv) Free available iron is an ideal resource for mucormycosis. Hyperglycemia causes glycosylation of transferrin and ferritin, and reduces iron binding allowing increased free iron. Moreover, increase in cytokines in patients with COVID-19 especially interleukin-6, increases free iron by increasing ferritin levels due to increased synthesis and decreased iron transport. Furthermore, concomitant acidosis increases free iron by the same mechanism and additionally by reducing the ability of transferrin to chelate iron,
  • (v) High glucose, low pH, free iron, and ketones in presence of decreased phagocytic activity of WBC, enhances the growth of mucor. In addition, it enhances the expression of glucose-regulator protein 78 (GRP-78) of endothelium cells and fungal ligand spore coating homolog (CotH) protein, enabling angio-invasion, hematogenous dissemination and tissue necrosis [ 52 ].

Fig. 1 depicts the postulated mechanism of increased propensity of having mucormycosis infection in COVID-19 patients.

Fig. 1

Postulated interaction of diabetes, corticosteroid and COVID-19 with mucormycosis.

There are certain limitations to conduct this kind of systematic review based on case reports/series subject to publication biases and considerable heterogeneity in reporting cases. It is highly likely that reported cases of mucormycosis may be an underrepresentation of the real burden owing to difficulty in making a microbiological or histopathological diagnosis especially in a raging pandemic setting. While some case reports had every minute detail, other did not report important parameter, for example – duration of DM, lack of baseline HbA1c data in majority of cases. Secondly, the lack of a denominator value may not allow the true estimation of mucormycosis incidence in people with COVID-19 compounded by the lack of control. Thirdly, defining active and recovered COVID-19 and its relation to the onset of mucormycosis could be difficult considering the lower sensitivity of confirmatory RT-PCR. Finally, evaluating the outcomes in people with mucormycosis and COVID-19 could be difficult at the moment because these case reports have been published while many of these patients are still under treatment. Other minor limitations have been highlighted in Table 2 .

5. Conclusions

Increase in mucormycosis in Indian context appears to be an unholy intersection of trinity of diabetes (high prevalence genetically), rampant use of corticosteroid (increases blood glucose and opportunistic fungal infection) and COVID-19 (cytokine storm, lymphopenia, endothelial damage). All efforts should be made to maintain optimal hyperglycemia and only judicious evidence-based use of corticosteroids in patients with COVID-19 is recommended in order to reduce the burden of fatal mucormycosis.

No funding.

Author's contribution

AKS conceptualized, searched the literature and wrote first draft; RS made the tables, analyzed the data and revised the first draft, SRJ and AM edited the final draft. All authors agreed mutually to submit for publication.

All authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship and take responsibility for the integrity of the work. They confirm that this paper will not be published elsewhere in the same form, in English or in any other language, including electronically.

Declaration of competing interest

We hereby declare that we have no conflict of interest, related to this article titled “Mucormycosis in COVID-19: A Systematic Review of Cases Reported Worldwide and in India”.

IMAGES

  1. (PDF) Mucormycosis in COVID Times

    mucormycosis research paper pdf

  2. (PDF) Therapy of Mucormycosis

    mucormycosis research paper pdf

  3. (PDF) Section A-Research paper Prosthetic Rehabilitation of

    mucormycosis research paper pdf

  4. (PDF) "Mucormycosis" The Emerging Global Threat -A Complete Review

    mucormycosis research paper pdf

  5. (PDF) Mucormycosis in immunochallenged patients

    mucormycosis research paper pdf

  6. (PDF) Open Access Journal of Clinical Research Case Report Prosthetic

    mucormycosis research paper pdf

VIDEO

  1. Mucormycosis: Clinical Management & Case Presentation(IMA UP)

  2. How to Write a Scientific Research Paper

  3. How to Summarize PDF for Free with PDF Flex

  4. Mucormycosis Death : म्युकोरमायकोसिसमुळे कल्याण शहरासह ग्रामीण भागात दोन रुग्णांचा मृत्यू

  5. How to Write Good Elsevier Scientific Paper

  6. Training on updates in diagnosis and management of mucormycosis

COMMENTS

  1. Epidemiology and Diagnosis of Mucormycosis: An Update

    2.1. Incidence of Mucormycosis. The incidence of mucormycosis is increasing [3,5,15,16,17].The exact incidence/prevalence is not known because there are few population-based studies [3,15,18], and they differ in capture periods, populations and definition or diagnostic procedures.In the study performed in the San Francisco Bay area, from 1992 to 1993, the annual incidence of mucormycosis was ...

  2. Mucormycosis: risk factors, diagnosis, treatments, and challenges

    Introduction. Mucormycosis is a fungal infection generally caused by the filamentous molds, and it belongs to the mucorals and entomophthorales order (Chakrabarti et al. 2006). Mucorales can be found in a variety of habitats including dirt, rotting plant matter, bread, and dust (Lazar et al. 2014). Mucorales infections can be contracted by inhalation of spores, absorption of infected meals, or ...

  3. Global guideline for the diagnosis and management of mucormycosis: an

    Mucormycosis is a difficult to diagnose rare disease with high morbidity and mortality. Diagnosis is often delayed, and disease tends to progress rapidly. Urgent surgical and medical intervention is lifesaving. Guidance on the complex multidisciplinary management has potential to improve prognosis, but approaches differ between health-care settings. From January, 2018, authors from 33 ...

  4. Clinical Review of Mucormycosis

    Epidemiology and host factors. Mucorales are ubiquitous fungi usually found in soil, decaying organic matter, compost and contaminated foods. Mucormycosis is considered a rare infection - diabetes remains the most prominent underlying medical comorbidity in infected patients, and was identified as an independent risk factor for rhino-orbital-cerebral mucormycosis in a meta-analysis of 851 ...

  5. (PDF) Mucormycosis

    Mucormycosis frequently infects the sinuses, brain, or lungs. While infection of. the oral cavity or brain are the most common forms of mucormycosis, the. fungus can also infect other areas of the ...

  6. Mucormycosis in 2023: an update on pathogenesis and management

    3 Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States. Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus, Mucor, and Lichtheimia, accounting for > 90% of all cases. MCR is seen in patients with severe immunosuppression such as those ...

  7. [PDF] Mucormycosis in 2023: an update on pathogenesis and management

    Therapy of MCR is multidisciplinary and requires a high index of suspicion for initiation of early Mucorales-active antifungals, but promise is brought by RNAi and CRISPR/Cas9 approaches. Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus, Mucor, and Lichtheimia, accounting for > 90% of all cases. MCR is seen in patients with ...

  8. Epidemiology and Diagnosis of Mucormycosis: An Update

    Mucormycosis is an angioinvasive fungal infection, due to fungi of the order Mucorales. Its incidence cannot be measured exactly, since there are few population-based studies, but multiple studies have shown that it is increasing. The prevalence of mucormycosis in India is about 80 times the prevalence in developed countries, being approximately 0.14 cases per 1000 population.

  9. Mucormycosis treatment: Recommendations, latest advances, and

    This review provides an update on mucormycosis management. The latest recommendations strongly recommend as first-line therapy the use of liposomal amphotericin B (≥5mg/kg) combined with surgery whenever possible. Isavuconazole and intravenous or delayed-release tablet forms of posaconazole have remained second-line.

  10. Mucormycosis: risk factors, diagnosis, treatments, and challenges

    Abstract. Mucormycosis is a deadly opportunistic disease caused by a group of fungus named mucormycetes. Fungal spores are normally present in the environment and the immune system of the body prevents them from causing disease in a healthy immunocompetent individual. But when the defense mechanism of the body is compromised such as in the ...

  11. Mucormycosis: time to address this deadly fungal infection

    Mucormycosis is caused by ubiquitous environmental moulds with a global distribution, including the Rhizopus, Apophysomyces, Mucor, and Lichtheimia species. Although generally harmless to an immunocompetent host, the infection can be deadly in patients with an impaired immune system, such as in those with haematological malignancies or poorly controlled diabetes, or in individuals receiving ...

  12. Global guideline for the diagnosis and management of mucormycosis: an

    Mucormycosis is a difficult to diagnose rare disease with high morbidity and mortality. Diagnosis is often delayed, and disease tends to progress rapidly. Urgent surgical and medical intervention is lifesaving. Guidance on the complex multidisciplinary management has potential to improve prognosis, …

  13. PDF Global guideline for the diagnosis and management of mucormycosis: an

    settings. Management of mucormycosis depends on recognising disease patterns and on early diagnosis. Limited availability of contemporary treatments burdens patients in low and middle income settings. Areas of uncertainty were identified and future research directions specified. Introduction Suspected mucormycosis requires urgent intervention,

  14. (PDF) Mucormycosis: A Brief Review

    Mucormycosis is an angio invasive infection that occurs due to the fungi mucorales. It is a rare disease but increasingly recognized in immunocompromised patients. It can be categorized into rhino ...

  15. A Systematic Review of the Therapeutic Outcome of Mucormycosis

    Mucormycosis is a potentially lethal mycosis caused by filamentous fungi of the subphylum Mucoromycotina [1, 2].Fungi in the subphylum Mucormycotina are saprophytic fungi that are ubiquitous in nature [2, 3].They are frequently isolated from decaying organic material, soil, and compost piles [].Human infection follows the inhalation of fungal spores, traumatic inoculation, and consumption of ...

  16. (PDF) MUCORMYCOSI (BLACK FUNGUS): A REVIEW Review Article

    Mucormycosis is a new angioinvasive infection cau sed by the ubiquitous filamentous fungus of th e Mucorales order of the Zygomycete class. Mucormycosis. has emerged as the third most prevalent ...

  17. PDF ORIGINAL RESEARCH Mucormycosis Research: A global outlook through

    Bibliometric research evidence on mucormycosis can be found in two recent publications. Dubey and Sharma (2021) presented a post-covid mucormycosis research status by analyzing only forty-one documents. The limitation of the study can be observed in less publications, and the study is limited to mucormycosis associated with COVID-19.

  18. (PDF) A Review: Mucormycosis

    Mucormycosis is an angioinvasive disease that is characterized by tissue infarction and necrosis. It is an insidious fungal infection caused by members of the zygomycotic and Mucorales species. It ...

  19. PDF Mucormycosis

    IJCRT2208050 International Journal of Creative Research Thoughts (IJCRT) www.ijcrt.org a401 Mucormycosis Mrs. Dhivya.R, M.sc (Nursing), Sree balaji college of nursing, Bharath university,Chrompet, chennai, Tamilnadu, India ABSTRACT Mucormycosis is an angio invasive infection that occurs due to the fungi mucorales. It is a rare disease

  20. Pathogenesis of Mucormycosis

    Mucormycosis is an infection caused by fungi belonging to the order Mucorales [].Rhizopus oryzae is the most common organism isolated from patients with mucormycosis and is responsible for ∼70% of all cases of mucormycosis [2-4].The major risk factors for mucormycosis include uncontrolled diabetes mellitus in ketoacidosis, other forms of metabolic acidosis, treatment with corticosteroids ...

  21. Generative AI Can Harm Learning

    Keywords: Generative AI, Human Capital Development, Education, Human-AI Collaboration, Large Language Models

  22. (PDF) Mucormycosis Research: A global outlook through bibliometric

    Through this paper, it is quite clear that the appearance of research contribution on mucormycosis coupled with COVID-19 has opened a new direction of research that clinical researchers take up in ...

  23. Pathogenicity and transmissibility of bovine H5N1 influenza virus

    Download PDF. Similar content being viewed by others. ... The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.

  24. King's Speech 2024: background briefing notes

    Research and statistics. Reports, analysis and official statistics. ... Policy paper King's Speech 2024: background briefing notes ... PDF, 556 KB, 104 pages.

  25. COVID-19-associated mucormycosis: Case report and systematic review

    Papers published in any language between December 1, 2019, to June 1, 2021, were included. The literature was searched using keywords of [(COVID 19 OR Coronavirus OR corona) AND (mucormycosis OR mucor)]. The EndNote database was used from importing and managing abstracts and full texts. After first evaluation of the paper, duplicates were removed.

  26. Mucormycosis in COVID-19: A systematic review of cases reported

    Results. Overall, 101 cases of mucormycosis in people with COVID-19 have been reported, of which 82 cases were from India and 19 from the rest of the world. Mucormycosis was predominantly seen in males (78.9%), both in people who were active (59.4%) or recovered (40.6%) from COVID-19. Pre-existing diabetes mellitus (DM) was present in 80% of ...