Targeted Delivery of Antifungal Liposomes to Rhizopus delemar: Comparison
Please note this is a comparison between Version 2 by Beatrix Zheng and Version 1 by Quanita J. Choudhury.

Mucormycosis (a.k.a. zygomycosis) is an often-life-threatening disease caused by fungi from the ancient fungal division Mucoromycota. Globally, there are nearly a million people with the disease. Rhizopus spp., and R. delemar (R. oryzae, R. arrhizus) in particular, are responsible for most of the diagnosed cases. Pulmonary, rhino-orbito-cerebral, and invasive mucormycosis are most effectively treated with amphotericin B (AmB) and particularly with liposomal formulations (e.g., AmBisome®). However, even after antifungal therapy, there is still a 50% mortality rate. Hence, there is a critical need to improve therapeutics for mucormycosis. Targeting AmB-loaded liposomes (AmB-LLs) with the pathogen receptor Dectin-1 (DEC1-AmB-LLs) to the beta-glucans expressed on the surface of Aspergillus fumigatus and Candida albicans lowers the effective dose required to kill cells relative to untargeted AmB-LLs. Because Dectin-1 is an immune receptor for R. delemar infections and may bind it directly, wthe researchers explored the Dectin-1-mediated delivery of liposomal AmB to R. delemar. DEC1-AmB-LLs bound 100- to 1000-fold more efficiently to the exopolysaccharide matrix of R. delemar germlings and mature hyphae relative to AmB-LLs. DEC1-AmB-LLs delivering sub-micromolar concentrations of AmB were an order of magnitude more efficient at inhibiting and/or killing R. delemar than AmB-LLs. Targeted antifungal drug-loaded liposomes have the potential to improve the treatment of mucormycosis. 

  • mucormycosis
  • Rhizopus delemar
  • C-type lectin receptors
  • Dectin-1
  • amphotericin B
  • liposomes
  • DectiSomes
  • oligoglucans
  • beta-glucan

1. Introduction

Globally, there are approximately 900,000 individuals with mucormycosis, mostly in India [1,2]. Among those at particular risk are patients with lung diseases; neutropenic patients, such as those receiving prolonged immunosuppression for hematopoietic stem cell transplants; patients receiving long-term treatment for inflammatory diseases; and patients with diabetic ketoacidosis, COVID-19, or AIDS [3,4,5,6,7,8,9,10]. The number of reported cases of mucormycosis has increased 6- to 7-fold in the last four decades [7], paralleling the increasing numbers of individuals on immunosuppressants and very recently COVID-19. Among the diverse Mucoromycota [11], the genus Rhizopus and one species in particular, Rhizopus delemar (R. oryzae, R. arrhizus), are responsible for 50% or more of all diagnosed cases [7,12,13]. R. delemar is an opportunistic pathogen living in soil on rotting vegetation. The primary infection route is via inhalation of its sporangiospores, which leads most commonly to pulmonary and rhino-orbito-cerebral infections [14]. Liposomal amphotericin B (AmB) followed by isavuconazole (ISZ) and/or posaconazole (POS) are the most commonly prescribed antifungals [15]. The surgical removal of infected tissue prior to antifungal therapy significantly improves the outcome [16,17]. However, even with antifungal therapy and surgery, there is still approximately a 50% to 99% mortality rate within several months of diagnosis depending upon the level of dissemination at the time of accurate diagnosis and treatment [7,14,16,18]. Clearly, there is a critical need for improved antifungal therapies for mucormycosis.
The immune response to infections caused by Rhizopus spp. is mediated by signaling from the C-type lectin pathogen receptor Dectin-1 (CLEC7A) [12]. Dectin-1 is expressed on the surface of some classes of leukocytes, including dendritic cells and neutrophils. Indirect evidence suggests Dectin-1 may bind directly to oligoglucans expressed by Rhizopus [19,20]. Two Dectin-1 monomers float together such that their extracellular carbohydrate recognition domains (CRDs) form homo-dimers that bind with high affinity to beta-glucans in the cell wall and/or the exopolysaccharide matrices of pathogens [21]. WThe hresearchers have been developing DectiSomes as anti-infective agents, using C-type lectin pathogen receptors to target liposomes loaded with antifungal drugs to pathogenic fungi [22,23,24]. The Weresearchers have shown that the CRD and stalk region of Dectin-1 may be tethered to liposomes loaded with antifungal drugs, targeting these liposomes specifically to beta-glucans on the surface of fungal pathogens [23,25]. As designed, Dectin-1 CRD monomers float in the liposomal membrane and form the functional homo-dimers necessary for beta-glucan binding. Dectin-1-coated, AmB-loaded liposomes (DEC1-AmB-LLs) bind to the cell walls and exopolysaccharides of Aspergillus fumigatus and Candida albicans orders of magnitude more strongly than untargeted AmBisome®-like AmB-LLs. DEC1-AmB-LLs also inhibit and/or kill in vitro-grown A. fumigatus 100-fold more efficiently than AmB-LLs, reducing the in vitro effective dose for 90% killing more than 10-fold. Considering that Dectin-1 might bind directly to R. delemar, wthe researchers explored the binding of DEC1-AmB-LLs to R. delemar and their potential to enhance the efficacy of antifungal liposome treatment.

2. DiscCusrrent Insightsion

Dectin-1 recognizes beta-glucans that are present in fungal cell walls and exopolysaccharide matrices but are sometimes masked by other molecular components. WThe researchers showed that Dectin-1 was extremely efficient at targeting AmB-loaded liposomes, DEC1-AmB-LLs, to R. delemar swollen sporangiospores, germlings, and mature hyphae. WThe researchers observed DEC1-AmB-LLs bound primarily to the exopolysaccharide matrix and less so to the cell wall or to exopolysaccharide deposited close to the cell wall. Rhodamine-tagged Dectin-1 protein bound with the same specificity to R. delemar’s exopolysaccharide. Hence, it appears that the 100 nm-size of DEC1-AmB-LLs did not significantly limit liposome access to its cognate ligands. DEC1-AmB-LLs were significantly and dramatically more effective at inhibiting and/or killing Rhizopus in vitro than untargeted AmB-LLs or BSA-AmB-LLs. Using both cell growth and metabolic assays, wthe researchers observed that DEC1-AmB-LLs delivering sub-micromolar concentrations of AmB were significantly more efficient at inhibiting and/or killing R. delemar than untargeted AmB-LLs. WThe researchers were able to detect significant loss of metabolic activity within three hours of treatment.
AmB has several partially validated antifungal activities related to its affinity for ergosterol (Erg) in the fungal bilipid membrane, including opening ion channels in the membrane to cause lethal ion leakage and extracting Erg from the lipid bilayer to the membrane surface, which also compromises the membrane [32]. OurThe results do not distinguish among the various mechanisms of AmB’s activity. Yet, ourthe data robustly demonstrate that that Dectin-1-targeted DEC1-AmB-LLs were more efficiently associated with R. delemar’s exopolysaccharides and had greater antifungal activity than either AmB delivered in AmB-LLs or ourthe protein-coated control BSA-AmB-LLs. Therefore, it does not appear that AmB itself plays a measurable role in the enhanced efficacy of targeted liposomes.
Each DEC1-AmB-LL DectiSome contains several thousand molecules of rhodamine B that enhance signal intensity and more than a thousand Dectin-1 receptor molecules on its surface, enabling multimer formation that enhances the avidity of binding to cognate oligoglycans [25]. If a C-type lectin receptor protein was used alone in a fungal cell binding study and assayed by immunofluorescence, signal intensities might be reduced by orders of magnitude relative to that achieved by a fluorescent DectiSome. This makes DectiSomes excellent reagents for examining the direct binding of different C-type lectins to various pathogens [22,23,24,25,28].
The Mucoromycota is an ancient division of the fungal kingdom that contains a large number of morphologically diverse human pathogens that cause mucormycosis [33,34]. They are estimated to have diverged from a common ancestor in the fungal tree of life nearly 1.3 billion years ago [35,36]. Hence, it is not surprising that the glycan composition of the Mucoromycota cell wall and exopolysaccharide matrix [19,37,38,39] appear to be distinct from other pathogenic fungi [30,40,41,42]. The sporangiospore and hyphal cell wall [38] and the exopolysaccharide matrix [39] each are composed of approximately 43% glucose; other components include lower amounts of N-acetyl-glucosamine, mannose, fructose, lipids, proteins, and phosphate [38,39]. Considering that Dectin-1 recognizes oligo-beta-glucans, it is not surprising that Dectin-1-targeted liposomes bound to Rhizopus. The weaker binding wthe researchers observed to the cell wall relative to the exopolysaccharide of Rhizopus suggests that most of the cell wall oligoglucans were masked from DEC1-AmB-LL binding. Experiments with DectiSomes targeted by the oligo-mannan-specific C-type lectin Dectin-2 are ongoing.
While liposomal AmB formulations such as AmBisome® delivering as much as 10 mg/kg/day are significantly less toxic than alternative AmB therapies, the several-months-long therapies needed to clear mucormycosis still result in infusion-related reactions and nephrotoxicity [43,44,45]. If DEC1-AmB-LLs can reduce the effective dose of liposomal AmB and/or reduce the duration of treatment in the clinic, this should reduce the risk of patients developing toxic effects from AmB. Salvage therapies after patients become intolerant to AmB include very high doses of posaconazole (POS) or isavuconazole (ISZ) on the order of hundreds of mg/kg/day [46,47,48,49,50]. Even if Dectin-1-targeted liposomes improve the performance of POS or ISZ by 10-fold in the clinic, it may not be cost-effective to prepare targeted liposomes that deliver tens of mg/kg/day doses of these drugs. However, ourthe data also suggest that DEC1-AmB-LLs kill Rhizopus faster than untargeted therapies. Enhanced speed of killing may enable patients to clear Rhizopus infections with drug regimens of shorter duration or with fewer treatments, reducing the risk of AmB toxicity.
Immunoliposomes have been used in the clinic for some time to target anti-cancer drugs to cancer cells and tumors. They generally improve drug efficacy several-fold over untargeted drugs [51,52,53]. Although conceptually DectiSomes function similarly to immunoliposomes by targeting drugs to pathogenic cells, DectiSomes have some distinct advantages [24]. C-type lectin receptors such as Dectin-1 generally recognize a much wider variety of target ligands than monoclonal antibodies, which supports their development as pan-antifungal reagents. Dectin-1 in particular recognizes the beta-glucans produced by nineteen of the twenty genera of pathogenic fungi [12]. Once developed for one fungal pathogen in the clinic, it should not be difficult to broaden their application to other pathogens. In addition, C-type lectins are much less expensive to produce than monoclonal antibodies, which will favor their development as reagents to treat fungal diseases in less wealthy countries [54,55,56,57]. Finally, low production costs may encourage the pharmaceutical industry to expend the large amounts of capital needed to develop DectiSomes.
In conclusion, there is a pressing demand for more effective therapeutics to treat mucormycosis because even after surgery and drug treatment, there is still a high mortality rate. WThe researchers have shown order of magnitude improvements in the in vitro performance of AmB against R. delemar when delivered by Dectin-1-targeted liposomes. It appears that targeting liposomal AmB to the exopolysaccharide matrix of Rhizopus is sufficient to significantly improve liposomal drug performance. Future experiments will focus on mouse models of mucormycosis, including determining if Dectin-1-targeted liposomes bind to R. delemar at infection sites in the lung, reduce fungal burden in the lungs, and improve mouse survival. WThe researchers also will need to confirm that Dectin-1-targeted liposomes work effectively against other clinically relevant members of Mucoromycota [58] in light of their ancient diversity [34].

References

  1. Prakash, H.; Chakrabarti, A. Global Epidemiology of Mucormycosis. J. Fungi 2019, 5, 26. [CrossRef] [PubMed]
  2. Bongomin, F.; Gago, S.; Oladele, R.O.; Denning, D.W. Global and Multi-National Prevalence of Fungal Diseases—Estimate Precision. J. Fungi 2017, 3, 57. [CrossRef] [PubMed]
  3. Kontoyiannis, D.P.; Lewis, R. Invasive Zygomycosis: Update on Pathogenesis, Clinical Manifestations, and Management. Infect. Dis. Clin. N. Am. 2006, 20, 581–607. [CrossRef] [PubMed]
  4. Pyrgos, V.; Shoham, S.; Walsh, T. Pulmonary Zygomycosis. Semin. Respir. Crit. Care Med. 2008, 29, 111–120. [CrossRef] [PubMed]
  5. Hassan, M.A.; Voigt, K. Pathogenicity patterns of mucormycosis: Epidemiology, interaction with immune cells and virulence factors. Med. Mycol. 2019, 57, S245–S256. [CrossRef] [PubMed]
  6. Jeong, W.; Keighley, C.; Wolfe, R.; Lee, W.L.; Slavin, M.A.; Kong, D.C.M.; Chen, S.C.-A. The epidemiology and clinical manifestations of mucormycosis: A systematic review and meta-analysis of case reports. Clin. Microbiol. Infect. 2019, 25, 26–34. [CrossRef]
  7. Song, Y.; Qiao, J.; Giovanni, G.; Liu, G.; Yang, H.; Wu, J.; Chen, J. Mucormycosis in renal transplant recipients: Review of 174 reported cases. BMC Infect. Dis. 2017, 17, 283. [CrossRef]
  8. Banerjee, I.; Robinson, J.; Asim, M.; Sathian, B.; Banerjee, I. Mucormycosis and COVID-19 an epidemic in a pandemic? Nepal J. Epidemiol. 2021, 11, 1034–1039. [CrossRef]
  9. Ibrahim, A.S.; Spellberg, B.; Walsh, T.J.; Kontoyiannis, D.P. Pathogenesis of Mucormycosis. Clin. Infect. Dis. 2012, 54, S16–S22. [CrossRef]
  10. Prakash, H.; Skiada, A.; Paul, R.; Chakrabarti, A.; Rudramurthy, S. Connecting the Dots: Interplay of Pathogenic Mechanisms between COVID-19 Disease and Mucormycosis. J. Fungi 2021, 7, 616. [CrossRef]
  11. Garre, V. Recent Advances and Future Directions in the Understanding of Mucormycosis. Front. Cell. Infect. Microbiol. 2022, 12, 850581. [CrossRef] [PubMed]
  12. Goyal, S.; Castrillón-Betancur, J.C.; Klaile, E.; Slevogt, H. The Interaction of Human Pathogenic Fungi with C-Type Lectin Receptors. Front. Immunol. 2018, 9, 1261. [CrossRef] [PubMed]
  13. Howard, D. (Ed.) Fungi Pathogenic for Humans and Animals; Dekker Incorporated, Marcel: New York, NY, USA, 1983; p. 652.
  14. Klimko, N.; Khostelidi, S.; Shadrivova, O.; Volkova, A.; Popova, M.; Uspenskaya, O.; Shneyder, T.; Bogomolova, T.; Ignatyeva, S.; Zubarovskaya, L.; et al. Contrasts between mucormycosis and aspergillosis in oncohematological patients. Med. Mycol. 2019, 57, S138–S144. [CrossRef]
  15. Cornely, O.A.; Alastruey-Izquierdo, A.; Arenz, D.; Chen, S.C.A.; Dannaoui, E.; Hochhegger, B.; Hoenigl, M.; Jensen, H.E.; Lagrou, K.; Lewis, R.E.; et al. Global guideline for the diagnosis and management of mucormycosis: An initiative of the European Confederation of Medical Mycology in cooperation with the Mycoses Study Group Education and Research Consortium. Lancet Infect. Dis. 2019, 19, e405–e421. [CrossRef]
  16. Patel, A.; Kaur, H.; Xess, I.; Michael, J.; Savio, J.; Rudramurthy, S.; Singh, R.; Shastri, P.; Umabala, P.; Sardana, R.; et al. A multicentre observational study on the epidemiology, risk factors, management and outcomes of mucormycosis in India. Clin. Microbiol. Infect. 2019, 26, 944.e9–944.e15. [CrossRef]
  17. Hamilos, G.; Samonis, G.; Kontoyiannis, D.P. Pulmonary Mucormycosis. Semin. Respir. Crit. Care Med. 2011, 32, 693–702. [CrossRef]
  18. Hong, H.-L.; Lee, Y.-M.; Kim, T.; Lee, J.-Y.; Chung, Y.-S.; Kim, M.-N.; Kim, S.-H.; Choi, S.-H.; Kim, Y.S.; Woo, J.H.; et al. Risk Factors for Mortality in Patients with Invasive Mucormycosis. Infect. Chemother. 2013, 45, 292–298. [CrossRef]
  19. Chamilos, G.; Ganguly, D.; Lande, R.; Gregorio, J.; Meller, S.; Goldman, W.E.; Gilliet, M.; Kontoyiannis, D.P. Generation of IL-23 Producing Dendritic Cells (DCs) by Airborne Fungi Regulates Fungal Pathogenicity via the Induction of TH-17 Responses. PLoS ONE 2010, 5, e12955. [CrossRef]
  20. Camargo, J.F.; Bhimji, A.; Kumar, D.; Kaul, R.; Pavan, R.; Schuh, A.; Seftel, M.; Lipton, J.H.; Gupta, V.; Humar, A.; et al. Impaired T Cell Responsiveness to Interleukin-6 in Hematological Patients with Invasive Aspergillosis. PLoS ONE 2015, 10, e0123171. [CrossRef]
  21. Brown, J.; O’Callaghan, C.A.; Marshall, A.; Gilbert, R.; Siebold, C.; Gordon, S.; Brown, G.; Jones, E.Y. Structure of the fungal β-glucan-binding immune receptor dectin-1: Implications for function. Protein Sci. 2007, 16, 1042–1052. [CrossRef]
  22. Ambati, S.; Pham, T.; Lewis, Z.A.; Lin, X.; Meagher, R.B. DC-SIGN targets amphotericin B-loaded liposomes to diverse pathogenic fungi. Fungal Biol. Biotechnol. 2021, 8, 22. [CrossRef] [PubMed]
  23. Ambati, S.; Pham, T.; Lewis, Z.A.; Lin, X.; Meagher, R.B. DectiSomes: Glycan Targeting of Liposomal Drugs Improves the Treatment of Disseminated Candidiasis. Antimicrob. Agents Chemother. 2022, 66, e01467-21. [CrossRef]
  24. Meagher, R.B.; Lewis, Z.A.; Ambati, S.; Lin, X. Aiming for a bull’s-eye: Targeting antifungals to fungi with dectin-decorated liposomes. PLoS Pathog. 2021, 17, e1009699. [CrossRef] [PubMed]
  25. Ambati, S.; Ferarro, A.R.; Khang, S.E.; Lin, J.; Lin, X.; Momany, M.; Lewis, Z.A.; Meagher, R.B. Dectin-1-Targeted Antifungal Liposomes Exhibit Enhanced Efficacy. mSphere 2019, 4, e00025-19. [CrossRef] [PubMed]
  26. Ramana, L.N.; Sharma, S.; Sethuraman, S.; Ranga, U.; Krishnan, U.M. Stealth anti-CD4 conjugated immunoliposomes with dual antiretroviral drugs—Modern Trojan horses to combat HIV. Eur. J. Pharm. Biopharm. 2015, 89, 300–311. [CrossRef]
  27. Tenchov, R. Understanding the Nanotechnology in COVID-19 Vaccines. CAS. 18 February 2021, pp. 1–15. Available online: https://www.cas.org/resource/blog/understanding-nanotechnology-covid-19-vaccines (accessed on 18 February 2021).
  28. Ambati, S.; Ellis, E.C.; Lin, J.; Lin, X.; Lewis, Z.A.; Meagher, R.B. Dectin-2-Targeted Antifungal Liposomes Exhibit Enhanced Efficacy. mSphere 2019, 4, e00715-19. [CrossRef]
  29. Andrianaki, A.M.; Kyrmizi, I.; Thanopoulou, K.; Baldin, C.; Drakos, E.; Soliman, S.S.M.; Shetty, A.C.; McCracken, C.; Akoumianaki, T.; Stylianou, K.; et al. Iron restriction inside macrophages regulates pulmonary host defense against Rhizopus species. Nat. Commun. 2018, 9, 3333. [CrossRef]
  30. Hamidi, M.; Okoro, O.V.; Milan, P.B.; Khalili, M.R.; Samadian, H.; Nie, L.; Shavandi, A. Fungal exopolysaccharides: Properties, sources, modifications, and biomedical applications. Carbohydr. Polym. 2022, 284, 119152. [CrossRef]
  31. Adams, E.L.; Rice, P.J.; Graves, B.; Ensley, H.E.; Yu, H.; Brown, G.D.; Gordon, S.; Monteiro, M.A.; Papp-Szabo, E.; Lowman, D.W.; et al. Differential High-Affinity Interaction of Dectin-1 with Natural or Synthetic Glucans Is Dependent upon Primary Structure and Is Influenced by Polymer Chain Length and Side-Chain Branching. J. Pharmacol. Exp. Ther. 2008, 325, 115–123. [CrossRef]
  32. Anderson, T.M.; Clay, M.C.; Cioffi, A.G.; Diaz, K.A.; Hisao, G.S.; Tuttle, M.D.; Nieuwkoop, A.; Comellas, G.; Maryum, N.; Wang, S.; et al. Amphotericin forms an extramembranous and fungicidal sterol sponge. Nat. Chem. Biol. 2014, 10, 400–406. [CrossRef]
  33. Ribes, J.A.; Vanover-Sams, C.L.; Baker, D.J. Zygomycetes in Human Disease. Clin. Microbiol. Rev. 2000, 13, 236–301. [CrossRef] [PubMed]
  34. Spatafora, J.W.; Chang, Y.; Benny, G.L.; Lazarus, K.; Smith, M.E.; Berbee, M.L.; Bonito, G.; Corradi, N.; Grigoriev, I.; Gryganskyi, A.; et al. A phylum-level phylogenetic classification of zygomycete fungi based on genome-scale data. Mycologia 2016, 108, 1028–1046. [CrossRef]
  35. Spatafora, J.W.; Aime, M.C.; Grigoriev, I.V.; Martin, F.; Stajich, J.E.; Blackwell, M. The Fungal Tree of Life: From Molecular Systematics to Genome-Scale Phylogenies. Microbiol. Spectr. 2017, 5, 3–34. [CrossRef] [PubMed]
  36. Padovan, A.C.B.; Sanson, G.F.; Brunstein, A.; Briones, M.R. Fungi Evolution Revisited: Application of the Penalized Likelihood Method to a Bayesian Fungal Phylogeny Provides a New Perspective on Phylogenetic Relationships and Divergence Dates of Ascomycota Groups. J. Mol. Evol. 2005, 60, 726–735. [CrossRef] [PubMed]
  37. Bartnicki-Garcia, S.; Reyes, E. Chemistry of spore wall differentiation in Mucor rouxii. Arch. Biochem. Biophys. 1964, 108, 125–133. [CrossRef]
  38. Lecointe, K.; Cornu, M.; Leroy, J.; Coulon, P.; Sendid, B. Polysaccharides Cell Wall Architecture of Mucorales. Front. Microbiol. 2019, 10, 469. [CrossRef]
  39. Yu, W.; Chen, G.; Zhang, P.; Chen, K. Purification, partial characterization and antitumor effect of an exopolysaccharide from Rhizopus nigricans. Int. J. Biol. Macromol. 2016, 82, 299–307. [CrossRef]
  40. Gow, N.A.R.; Latge, J.-P.; Munro, C.A. The Fungal Cell Wall: Structure, Biosynthesis, and Function. Microbiol. Spectr. 2017, 5, 28513415. [CrossRef]
  41. Reichhardt, C.; A Stevens, D.; Cegelski, L. Fungal biofilm composition and opportunities in drug discovery. Future Med. Chem. 2016, 8, 1455–1468. [CrossRef]
  42. Lagree, K.; Mitchell, A.P. Fungal Biofilms: Inside Out. Microbiol. Spectr. 2017, 5, 873–886. [CrossRef]
  43. Hamill, R.J. Amphotericin B Formulations: A Comparative Review of Efficacy and Toxicity. Drugs 2013, 73, 919–934. [CrossRef] [PubMed]
  44. Deray, G. Amphotericin B nephrotoxicity. J. Antimicrob. Chemother. 2002, 49, 37–41. [CrossRef] [PubMed]
  45. Stanzani, M.; Vianelli, N.; Cavo, M.; Maritati, A.; Morotti, M.; Lewis, R.E. Retrospective Cohort Analysis of Liposomal Am- photericin B Nephrotoxicity in Patients with Hematological Malignancies. Antimicrob. Agents Chemother. 2017, 61, e02651-16. [CrossRef] [PubMed]
  46. Jeong, W.; Keighley, C.; Wolfe, R.; Lee, W.L.; Slavin, M.A.; Chen, S.C.-A.; Kong, D.C. Contemporary management and clinical outcomes of mucormycosis: A systematic review and meta-analysis of case reports. Int. J. Antimicrob. Agents 2019, 53, 589–597. [CrossRef] [PubMed]
  47. Floros, L.; Kuessner, D.; Posthumus, J.; Bagshaw, E.; Sjölin, J. Cost-effectiveness analysis of isavuconazole versus voriconazole for the treatment of patients with possible invasive aspergillosis in Sweden. BMC Infect. Dis. 2019, 19, 134. [CrossRef] [PubMed]
  48. Skiada, A.; Lass-Floerl, C.; Klimko, N.; Ibrahim, A.; Roilides, E.; Petrikkos, G. Challenges in the diagnosis and treatment of mucormycosis. Med. Mycol. 2018, 56, S93–S101. [CrossRef]
  49. Dannaoui, E. Antifungal resistance in mucorales. Int. J. Antimicrob. Agents 2017, 50, 617–621. [CrossRef]
  50. Riley, T.T.; Muzny, C.; Swiatlo, E.; Legendre, D.P. Breaking the Mold: A Review of Mucormycosis and Current Pharmacological Treatment Options. Ann. Pharmacother. 2016, 50, 747–757. [CrossRef]
  51. Merino, M.; Zalba, S.; Garrido, M.J. Immunoliposomes in clinical oncology: State of the art and future perspectives. J. Control. Release 2018, 275, 162–176. [CrossRef]
  52. Eloy, J.O.; Petrilli, R.; Trevizan, L.N.F.; Chorilli, M. Immunoliposomes: A review on functionalization strategies and targets for drug delivery. Colloids Surf. B Biointerfaces 2017, 159, 454–467. [CrossRef]
  53. Wang, D.; Sun, Y.; Liu, Y.; Meng, F.; Lee, R.J. Clinical translation of immunoliposomes for cancer therapy: Recent perspectives. Expert Opin. Drug Deliv. 2018, 15, 893–903. [CrossRef] [PubMed]
  54. Suvvari, T.K.; Arigapudi, N.; Kandi, V.R.; Kutikuppala, L.S. Mucormycosis: A killer in the shadow of COVID-19. J. Mycol. Med. 2021, 31, 101161. [CrossRef] [PubMed]
  55. Selarka, L.; Sharma, S.; Saini, D.; Sharma, S.; Batra, A.; Waghmare, V.T.; Dileep, P.; Patel, S.; Shah, M.; Parikh, T.; et al. Mucormycosis and COVID-19: An epidemic within a pandemic in India. Mycoses 2021, 64, 1253–1260. [CrossRef] [PubMed]
  56. Pal, R.; Singh, B.; Bhadada, S.K.; Banerjee, M.; Bhogal, R.S.; Hage, N.; Kumar, A. COVID-19-associated mucormycosis: An updated systematic review of literature. Mycoses 2021, 64, 1452–1459. [CrossRef]
  57. Bhanuprasad, K.; Manesh, A.; Devasagayam, E.; Varghese, L.; Cherian, L.M.; Kurien, R.; Karthik, R.; Deodhar, D.; Vanjare, H.; Peter, J.; et al. Risk factors associated with the mucormycosis epidemic during the COVID-19 pandemic. Int. J. Infect. Dis. 2021, 111, 267–270. [CrossRef]
  58. Walther, G.; Wagner, L.; Kurzai, O. Updates on the Taxonomy of Mucorales with an Emphasis on Clinically Important Taxa. J. Fungi 2019, 5, 106. [CrossRef]
More