Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2235 2022-06-21 10:41:29 |
2 format corrected. Meta information modification 2235 2022-06-21 11:06:22 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Barantsevich, N.;  Barantsevich, E. Treatment of Invasive Candidiasis. Encyclopedia. Available online: https://encyclopedia.pub/entry/24254 (accessed on 06 July 2024).
Barantsevich N,  Barantsevich E. Treatment of Invasive Candidiasis. Encyclopedia. Available at: https://encyclopedia.pub/entry/24254. Accessed July 06, 2024.
Barantsevich, Natalia, Elena Barantsevich. "Treatment of Invasive Candidiasis" Encyclopedia, https://encyclopedia.pub/entry/24254 (accessed July 06, 2024).
Barantsevich, N., & Barantsevich, E. (2022, June 21). Treatment of Invasive Candidiasis. In Encyclopedia. https://encyclopedia.pub/entry/24254
Barantsevich, Natalia and Elena Barantsevich. "Treatment of Invasive Candidiasis." Encyclopedia. Web. 21 June, 2022.
Treatment of Invasive Candidiasis
Edit

Candida species, belonging to commensal microbial communities in humans, cause opportunistic infections in individuals with impaired immunity. Pathogens encountered in more than 90% cases of invasive candidiasis include C. albicans, C. glabrata, C. krusei, C. tropicalis, and C. parapsilosis. The most frequently diagnosed invasive infection is candidemia. About 50% of candidemia cases result in deep-seated infection due to hematogenous spread. The sensitivity of blood cultures in autopsy-proven invasive candidiasis ranges from 21% to 71%. Non-cultural methods (beta-D-glucan, T2Candida assays), especially beta-D-glucan in combination with procalcitonin, appear promising in the exclusion of invasive candidiasis with high sensitivity (98%) and negative predictive value (95%). There is currently a clear deficiency in approved sensitive and precise diagnostic techniques. Omics technologies seem promising, though require further development and study. Therapeutic options for invasive candidiasis are generally limited to four classes of systemic antifungals (polyenes, antimetabolite 5-fluorocytosine, azoles, echinocandins) with the two latter being highly effective and well-tolerated and hence the most widely used.

Candida diagnostic tests beta-D-glucan T2Candida mannan procalcitonin echinocandins azoles flucytosine amphotericin B

1. Introduction

Candida species are yeasts and members of the commensal microbial community in humans [1][2][3]. They are present on skin and mucous membranes of the oral cavity and gastrointestinal and genitourinary tracts [1][2][4][5][6][7][8][9]. These fungi cause superficial or invasive infections in individuals with impaired immunity [10][11][12]. The term invasive candidiasis refers to candidemia and infections of other normally sterile sites [13][14][15][16][17].
Risk factors for invasive Candida infections relate to a wide variety of conditions (hematologic and solid organ malignancies, burns, major surgery) and treatment methods (stem cell and organ transplantation, use of immunosuppressive agents, antibiotics, chemotherapy, hemodialysis, intravenous nutrition) [15][16][17][18][19][20][21][22]. Prolonged hospital stay and admission to intensive care units are also recognized as major risk factors for invasive candidiasis [23][24][25].
The genus Candida includes numerous species [16][26][27][28][29]. The most common species to form normal microbiota and potentially cause invasive infections are C. albicans, C. glabrata, C. krusei, C. tropicalis, and C. parapsilosis. These five species are responsible for more than 90% of invasive infections [16][30][31][32][33]. Other Candida spp. have also been reported worldwide as causative agents of invasive candidiasis in patients, but to a lesser extent [16]. A novel Candida species—C. auris—has recently emerged as an etiologic agent of invasive candidiasis worldwide [34][35]. The fungus first identified in 2009 in Japan was later found on all continents except Antarctica [36][37][38]. This species is closely related to the C. haemulonii complex and has five geographically and genetically distant clades [39][40]. The rise in cases occurred simultaneously in different regions [41][42]. C. auris easily forms biofilms, persist on different surfaces [37][43][44], and has high potential for nosocomial transmission [45][46].
Candidemia, an infection of the bloodstream with Candida spp., is the most frequently detected type of invasive Candida infection. It is the fourth leading cause of nosocomial bloodstream infections in the United States of America (USA) and the seventh in Europe [47]. The overall mortality rate of candidemia is 22–75% [48]. Attributable mortality is rarely estimated due to contributing severe underlying conditions, and ranges from 10% to 47% [27][49]. The prevalence of candidemia differs in various geographical regions, with 0.32/1000 admissions in South-Eastern China and up to 2.49/1000 admissions in Brazil [48]. The distribution of certain Candida spp. as the cause of invasive infections may depend on the underlying conditions and antifungal preparations used. C. glabrata is more likely to be isolated in patients with malignancy and transplantation, and C. krusei in patients with haematologic malignancies receiving fluconazole as prophylaxis [23].

2. Treatment of Invasive Candidiasis

2.1. Principles of Therapy

The outcomes in patients with invasive candidiasis are generally dependent on mycological cure, the severity of the underlying conditions, and the time frame of therapy. The delay in the introduction of antifungal treatment for each 12–24 h may result in increases in mortality rate of up to 100% [49].
There are three major points that affect the outcome and duration of invasive candidiasis. First is the early diagnosis of infection. This requires the analysis of risk factors and clinical manifestations and the prompt use of all available approved cultural and non-cultural diagnostic methods [27][50]. Second is the search for the possible source of infection and its removal. It is very important to eliminate all blood and urine catheters. often encountered as sources of infection. as well as prosthetic devices where possible. All catheters and devices should be cultured upon removal. The surgical debridement of the site of infection has to be performed or, alternatively, the drainage of abscesses or infected peritoneal or pleural fluids should be performed [21][27][51][52][53]. Third, early effective systemic antifungal therapy has to be administered [51][52]. The delay in the administration of antifungal therapy, inappropriate formulations, and inadequate dosages result in higher mortality rates [54][55][56]. It is important to consider both the susceptibility of the yeast and drug–drug interactions along with the PK/PD data which have a high impact on certain loci of infections (e.g., in Candida endocarditis, Candida endophthalmitis, the central nervous system, or bone tissue involvement) [27].
The deficiency in reliable and sensitive methods for the diagnosis of invasive candidiasis and the long time (2–4 days) necessary for the isolation of fungus and susceptibility testing makes the early treatment that is crucial for a positive outcome challenging. The major aim is to start therapy as early as possible and use the effective regiment, but this is not easily feasible. There are several regularly updated detailed guidelines for the treatment of invasive candidiasis that can be helpful to healthcare professionals [56][57].

2.2. Antifungal Preparations

Systemic antifungals are used in the treatment of invasive candidiasis. There are four classes of preparations that differ in their mechanisms of action. One of the earliest systemic antifungal preparations introduced to clinical practice in 1959 was polyene amphotericin B [58]. It has been successfully used in the treatment of different invasive fungal infections, including candidiasis, since that time. Polyenes bind to ergosterol, the major component of the fungal cell membrane, creating pores and subsequent cell death [59][60]. These agents have a broad spectrum and a potent fungicidal effect. Most Candida species retain susceptibility to systemic polyenes. However, resistance is frequently detected in C. lusitaniae [61]. Amphotericin B and its lipid formulations are the only systemic polyenes available with lipid formulations less toxic and better tolerated by patients [62]. The use of conventional amphotericin B is limited by often-encountered individual intolerance and nephrotoxicity [63][64][65].
5-fluorocytosine (flucytosine) was developed as an antimetabolite in 1957. It inhibits fungal protein synthesis after being converted to 5-fluorouracil by cytosine deaminase and incorporated into fungal RNA, replacing uridylic acid. It is also a potent inhibitor of fungal DNA synthesis through the inhibition of thymidylate synthetase [66][67]. The drug is effective against a wide range of Candida spp., is well tolerated, and has a synergistic effect with amphotericin B. Flucytosine is used in combination with amphotericin B in invasive candidiasis in neonates with Candida meningitis due to its superb penetration into the cerebrospinal fluid. The use of flucytosine monotherapy is limited due to easily emerging resistance [68][69][70][71]. Hematologic and hepatic toxicities are associated with flucytosine administration. The careful monitoring of blood cell count is recommended. The adjustment of dose is advocated in patients with renal disfunction: serum concentration monitoring is helpful [70].
Systemic azoles and currently widely used triasoles have several antifungal formulations, including fluconazole, itraconazole, voriconazole, posaconazole, ravuconazole, and isavuconazole. Azoles inhibit lanosterol 14α demethylase, a key enzyme of ergosterol biosynthesis. Azoles generally demonstrate fungistatic activity against Candida spp. Most of them are active against fungi, causing invasive candidiasis with some agents demonstrating decreased activity against certain species. Resistance to fluconazole is most common in C. auris, C. glabrata, and C. parapsilosis; C. krusei has intrinsic resistance to fluconazole. Resistance to other azoles is rarely encountered, though increasing in frequency [72][73][74][75].
Echinocandins (micafungin, anidulafungin, caspofungin) present the newest class of antifungals and feature a fungicidal effect in Candida species. Their mechanism of action is the inhibition of β-D-glucan synthase, the important enzyme in cell wall synthesis. All echinocandins have high efficiency in invasive infections and an exceptional safety profile. A novel echinocandin rezafungin with once-weekly dosing and activity against Candida spp., including subsets of echinocandin-resistant Candida auris, has been recently developed and is currently in phase III trials [76][77][78][79][80][81][82]. Echinocandins demonstrate wide distribution in different organs and tissues, except the brain and eyes [83][84].

2.3. Choice of Antifungal Preparations and Duration of Treatment

Early diagnosis and early efficient initial therapy play a crucial role in the outcome of infection: a 24 h delay in obtaining positive blood cultures is associated with an almost two-fold increase in mortality in cancer patients [85]. Echinocandins are considered the drugs of choice for initial therapy in most cases of invasive candidiasis [86][87][88], though it depends on the severity of infection, data on the effectiveness of the antifungal treatment in previous episodes and intolerance to antifungals, the involvement of organs that demand specific permeability, and the tissue distribution of the agent (central nervous system, valves, bones, articular tissues, etc). Data on the dominant pathogen and its antifungal susceptibility in certain hospital settings, such as wards and departments, especially in non-neutropenic patients when an exogenous source of infection is suspected, are also important (Table 1).
Table 1. Therapy of invasive candidiasis and preferred medications.
Etiologic Agent
of Invasive
Candidiasis
Therapy
C. albicans, C. parapsilosis,
C. tropicalis
C. krusei,
C. glabrata
C. auris
First-line therapy * Echinocandin Echinocandin Echinocandin
Alternative first-line therapy Fluconazole Amphotericin B lipid formulations Amphotericin B lipid formulations
Step-down therapy ** Fluconazole Voriconazole Susceptibility data required
Notes: * Liposomal amphotericin B and flucytosine are used in the central nervous system or for eye infections. ** Step-down therapy is based on MIC assessment in individual cases.
Echinocandins were effective as initial treatment in 70–75% of patients with invasive candidiasis, according to randomized clinical trials [27][87]. Lower than expected survival rate might be due to the insusceptibility of the fungus to the agent used, or other circumstances (severe underlying condition or infection, delay in diagnosis or the initiation of therapy, non-performed debridement, or poor permeability of the drug into the infected site) [89][90][91][92][93]. An analysis of seven randomized clinical trials in roughly 2000 patients with invasive candidiasis showed that choice of an echinocandin as the initial treatment was associated with a significantly lower 30-day mortality rate compared with azoles or amphotericin B [56]. Worse outcomes were associated with C. tropicalis infections, higher APACHE II score, and older age [56]. C. parapsilosis usually demonstrates higher MIC to echinocandins than other Candida spp. Nevertheless, data from several clinical trials have shown that initial therapy with any echinocandin is appropriate for patients with C. parapsilosis infection, contrary to infections caused by other Candida spp. with acquired resistance to echinocandins [94][95][96]. Two randomized trials compared an echinocandin with an azole as a first-line therapy of candidaemia. One of them demonstrated that overall response rates were lower with fluconazole (60%) examined against anidulafungin (76%); another provided similar data for isavuconazole (60%) compared with caspofungin (71%) [89].
De-escalation or step-down therapy with changes from echinocandins to azoles provides favorable results in invasive candidiasis with different organ involvement (Table 1). The step-down therapy is normally started after 3–7 days of initial treatment and is administered in accordance with the results of susceptibility testing. Fluconazole could be the perfect choice for infections caused by susceptible strains with high efficiency, well-tolerability, and oral formulations with absorption from the gastrointestinal tract above 90% (Table 1). Several clinical trials did not reveal any differences in the 30-day survival or mycological cure rates of patients who received echinocandin therapy and those who were transferred to de-escalation therapy with oral azoles, according to susceptibility testing after 5 days of treatment with echinocandin [91][97]. Another option for step-down therapy with azoles in invasive candidiasis is voriconazole active against C. guilliermondii, C. glabrata and C. krusei with reduced susceptibility or resistance to fluconazole. De-escalation therapy significantly decreases the financial burden of invasive candidiasis for healthcare systems, with reduced hospital costs and shortened stay, as azoles, unlike echinocandins, can be successfully administered as oral formulations.
Azoles have some advantages compared to echinocandins as a first-line therapy treatment for the involvement of certain organs. Contrary to echinocandins, azoles easily cross the blood–brain barrier, making them the drug of choice for initial treatment in cases with central nervous system involvement. Fluconazole is widely used in low-income countries with a low prevalence of azole resistance as the first therapeutic choice in all types of invasive Candida infections. Patients with invasive candidiasis and no previous exposure to azoles may receive azoles as initial therapy if they are stable hemodynamically and do not have an increased risk of C. glabrata infection. The group of patients at high risk of C. glabrata includes patients with cancer or diabetes mellitus, and the elderly [51]. There are trials of newer azoles (posaconazole, ravuconazole, isavuconazole) demonstrating excellent in vitro activity against C. krusei, C. guilliermondii and C. glabrata. They might be alternatives to echinocandins as a first-line therapy [27][98].
Amphotericin B deoxycholate or, preferably, lipid formulations may be considered in infections refractory to other systemic antifungals in Candida endocarditis or Candida endophthalmitis. Intolerance of other classes of antifungals in individuals may also favor the use of amphotericin B lipid formulations. Multidrug-resistant strains with resistance to azoles and echinocandins are encountered with increasing frequency in C. glabrata and C. auris, with amphotericin B lipid formulations successfully used in these cases. Pan-drug-resistant strains of C. auris with no available treatment options have already been reported [99].
The recommended duration of systemic antifungal therapy for candidemia is at least 14 days after the eradication of Candida spp. from the blood and the resolution of all symptoms and signs of infection [51][57]. In deep-seated invasive candidiasis, e.g., chronic hepatosplenic candidiasis or intra-abdominal candidiasis, the duration of treatment normally differs from several weeks to 6–12 months and is individually adjusted. It is guided by the rate of lesion resolution, physicians’ personal experience, and scarce and typically non-randomized studies [27].

References

  1. Romo, J.A.; Kumamoto, C.A. On commensalism of Candida. J. Fungi 2020, 6, 16.
  2. Kumamoto, C.A.; Gresnigt, M.S.; Hube, B. The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine. Curr. Opin. Microbiol. 2020, 56, 7–15.
  3. Pérez, J.C. Fungi of the human gut microbiota: Roles and significance. Int. J. Med. Microbiol. 2021, 311, 151490.
  4. Bacali, C.; Vulturar, R.; Buduru, S.; Cozma, A.; Fodor, A.; Chis, A.; Lucaciu, O.; Damian, L.; Moldovan, M.L. Oral microbiome: Metting to now and befriend neighbours, a biological approach. Biomedicines 2022, 10, 671.
  5. Hameed, S.; Hans, S.; Monasky, R.; Thangamani, S.; Fatima, Z. Understanding human microbiota offers novel and promising therapeutic options against Candida infections. Pathogens 2021, 10, 183.
  6. Boxberger, M.; Cenizo, V.; Cassir, N.; La Scola, B. Challenges in exploring and manipulating the human skin microbiome. Microbiome 2021, 9, 125.
  7. Tortelli, B.A.; Lewis, W.G.; Allsworth, J.E.; Member-Meneh, N.; Foster, L.R.; Reno, H.E.; Peipert, J.F.; Fay, J.C.; Lewis, A.L. Associations between the vaginal microbiome and Candida colonization in women of reproductive age. Am. J. Obstet. Gynecol. 2020, 222, 471.e1–471.e9.
  8. Chee, W.J.Y.; Chew, S.Y.; Than, L.T.L. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb. Cell Fact. 2020, 19, 203.
  9. Kalia, N.; Singh, J.; Kaur, M. Microbiota in vaginal health and pathogenesis of recurrent vulvovaginal infections: A critical review. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 5.
  10. Pellon, A.; Sadeghi Nasab, S.D.; Moyes, D.L. New insights in Candida albicans innate immunity at the mucosa: Toxins, epithelium, metabolism, and beyond. Front. Cell. Infect. Microbiol. 2020, 10, 81.
  11. Lanternier, F.; Cypowyj, S.; Picard, C.; Bustamante, J.; Lortholary, O.; Casanova, J.L.; Puel, A. Primary immunodeficiencies underlying fungal infections. Curr. Opin. Pediatrics 2013, 25, 736–747.
  12. Abd Elaziz, D.; Abd El-Ghany, M.; Meshaal, S.; El Hawary, R.; Lotfy, S.; Galal, N.; Ouf, S.A.; Elmarsafy, A. Fungal infections in primary immunodeficiency diseases. Clin. Immunol. 2020, 219, 108553.
  13. Festekjian, A.; Neely, M. Incidence and predictors of invasive candidiasis associated with candidaemia in children. Mycoses 2011, 54, 146–153.
  14. St-Germain, G.; Laverdière, M.; Pelletier, R.; Bourgault, A.M.; Libman, M.; Lemieux, C.; Noël, G. Prevalence and antifungal susceptibility of 442 Candida isolates from blood and other normally sterile sites: Results of a 2-year (1996 to 1998) multicenter surveillance study in Quebec, Canada. J. Clin. Microbiol. 2001, 39, 949–953.
  15. 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.
  16. Spampinato, C.; Leonardi, D. Candida infections, causes, targets, and resistance mechanisms: Traditional and alternative antifungal agents. Biomed Res. Int. 2013, 2013, 204237.
  17. Ghanem-Zoubi, N.; Khoury, J.; Arnon, M.; Zorbavel, D.; Geffen, Y.; Paul, M. Risk factors for non-albicans candidemia focusing on prior antifungal and immunosuppressive therapy. Isr. Med. Assoc. J. 2019, 21, 303–307.
  18. Yapar, N. Epidemiology and risk factors for invasive candidiasis. Ther. Clin. Risk Manag. 2014, 10, 95–105.
  19. Thomas-Rüddel, D.O.; Schlattmann, P.; Pletz, M.; Kurzai, O.; Bloos, F. Risk factors for invasive Candida infection in critically ill patients: A systematic review and meta-analysis. Chest 2022, 161, 345–355.
  20. Pyrgos, V.; Ratanavanich, K.; Donegan, N.; Veis, J.; Walsh, T.J.; Shoham, S. Candida bloodstream infections in hemodialysis recipients. Med. Mycol. 2009, 47, 463–467.
  21. Girmenia, C.; Finolezzi, E.; Federico, V.; Santopietro, M.; Perrone, S. Invasive Candida infections in patients with haematological malignancies and hematopoietic stem cell transplant recipients: Current epidemiology and therapeutic options. Mediterr. J. Hematol. Infect. Dis. 2011, 3, e2011013.
  22. Lin, Y.-L.; Chen, I.-C.; Yen, J.-H.; Lai, C.-S.; Tsai, Y.-C.; Lu, C.-T.; Wu, C.-Y.; Lin, W.-S.; Lin, C.-H.; Huang, Y.-C. Invasive Candidiasis in Hospitalized Patients with Major Burns. J. Pers. Med. 2022, 12, 47.
  23. McCarty, T.P.; White, C.M.; Pappas, P.G. Candidemia and Invasive Candidiasis. Infect Dis. Clin. 2021, 35, 389–413.
  24. Zhang, Z.; Zhu, R.; Luan, Z.; Ma, X. Risk of invasive candidiasis with prolonged duration of ICU stay: A systematic review and meta-analysis. BMJ Open 2020, 10, e036452.
  25. Delaloye, J.; Calandra, T. Invasive candidiasis as a cause of sepsis in the critically ill patient. Virulence 2014, 5, 161–169.
  26. Martin-Loeches, I.; Antonelli, M.; Cuenca-Estrella, M.; Dimopoulos, G.; Einav, S.; De Waele, J.J.; Garnacho-Montero, J.; Kanj, S.S.; Machado, F.R.; Montravers, P.; et al. ESICM/ESCMID task force on practical management of invasive candidiasis in critically ill patients. Intensive Care Med. 2019, 45, 789–805.
  27. Pappas, P.; Lionakis, M.; Arendrup, M.; Ostrosky-Zeichner, L.; Kullberg, B.J. Invasive candidiasis. Nat. Rev. Dis. Prim. 2018, 4, 18026.
  28. Turner, S.A.; Butler, G. The Candida pathogenic species complex. Cold Spring Harb. Perspect. Med. 2014, 4, a019778.
  29. Papon, N.; Courdavault, V.; Clastre, M.; Bennett, R.J. Emerging and emerged pathogenic Candida species: Beyond the Candida albicans paradigm. PLoS Pathog. 2013, 9, e1003550.
  30. Singh, D.K.; Tóth, R.; Gácser, A. Mechanisms of pathogenic Candida species to evade the host complement attack. Front. Cell. Infect. Microbiol. 2020, 10, 94.
  31. Xiao, Z.; Wang, Q.; Zhu, F.; An, Y. Epidemiology, species distribution, antifungal susceptibility and mortality risk factors of candidemia among critically ill patients: A retrospective study from 2011 to 2017 in a teaching hospital in China. Antimicrob. Resist. Infect. Control 2019, 8, 89.
  32. Pappas, P.G. Antifungal clinical trials and guidelines: What we know and do not know. Cold Spring Harb. Perspect. Med. 2014, 4, a019745.
  33. Pfaller, M.A.; Diekema, D.J. Epidemiology of invasive candidiasis: A persistent public health problem. Clin. Microbiol. Rev. 2007, 20, 133–163.
  34. Rossato, L.; Colombo, A.L. Candida auris: What have we learned about its mechanisms of pathogenicity? Front. Microbiol. 2018, 9, 3081.
  35. Du, H.; Bing, J.; Hu, T.; Ennis, C.L.; Nobile, C.J.; Huang, G. Candida auris: Epidemiology, biology, antifungal resistance, and virulence. PLoS Pathog. 2020, 16, e1008921.
  36. Murphy, S.E.; Bicanic, T. Drug resistance and novel therapeutic approaches in invasive candidiasis. Front. Cell. Infect. Microbiol. 2021, 11, 759408.
  37. Keighley, C.; Garnham, K.; Harch, S.A.J.; Robertson, M.; Chaw, K.; Teng, J.C.; Chen, S.C.-A. Candida auris: Diagnostic challenges and emerging opportunities for the clinical microbiology laboratory. Curr. Fungal Infect. Rep. 2021, 15, 116–126.
  38. Lone, S.A.; Ahmad, A. Candida auris—The growing menace to global health. Mycoses 2019, 62, 620–637.
  39. Garcia-Bustos, V.; Cabanero -Navalon, M.D.; Ruiz-Saurí, A.; Ruiz-Gaitán, A.C.; Salavert, M.; Tormo, M.Á.; Pemán, J. What do we know about Candida auris? State of the art, knowledge gaps, and future directions. Microorganisms 2021, 9, 2177.
  40. Chatzimoschou, A.; Giampani, A.; Meis, J.F.; Roilides, E. Activities of nine antifungal agents against Candida auris biofilms. Mycoses 2021, 64, 381–384.
  41. Forsberg, K.; Woodworth, K.; Walters, M.; Berkow, E.L.; Jackson, B.; Chiller, T.; Vallabhaneni, S. Candida auris: The recent emergence of a multidrug-resistant fungal pathogen. Med. Mycol. 2019, 57, 1–12.
  42. Lockhart, S.R.; Etienne, K.A.; Vallabhaneni, S.; Farooqi, J.; Chowdhary, A.; Govender, N.P.; Colombo, A.L.; Calvo, B.; Cuomo, C.; Desjardins, C.A.; et al. Simultaneous emergence of multidrug-resistant Candida auris on 3 continents confirmed by whole-genome sequencing and epidemiological analyses. Clin. Infect. Dis. 2017, 64, 134–140.
  43. Horton, M.V.; Nett, J.E. Candida auris infection and biofilm formation: Going beyond the surface. Curr. Clin. Microbiol. Rep. 2020, 7, 51–56.
  44. Lara, H.H.; Ixtepan-Turrent, L.; Jose Yacaman, M.; Lopez-Ribot, J. Inhibition of Candida auris biofilm formation on medical and environmental surfaces by silver nanoparticles. ACS Appl. Mater. Interfaces 2020, 12, 21183–21191.
  45. Rodrigues, L.S.; Gazara, R.K.; Passarelli-Araujo, H.; Valengo, A.E.; Pontes, P.V.M.; Nunes-da-Fonseca, R.; de Souza, R.F.; Venancio, T.M.; Dalla-Costa, L.M. First genome sequences of two multidrug-resistant Candida haemulonii var. vulnera isolates from pediatric patients with candidemia. Front. Microbiol. 2020, 11, 1535.
  46. Belkin, A.; Gazit, Z.; Keller, N.; Ben-Ami, R.; Wieder-Finesod, A.; Novikov, A.; Rahav, G.; Brosh-Nissimov, T. Candida auris infection leading to nosocomial transmission, Israel, 2017. Emerg. Infect. Dis. 2018, 24, 801–804.
  47. Cuervo, G.; Garcia-Vidal, C.; Puig-Asensio, M.; Merino, P.; Vena, A.; Martín-Peña, A.; Montejo, J.M.; Ruiz, A.; Lázaro-Perona, F.; Fortún, J.; et al. Usefulness of guideline recommendations for prognosis in patients with candidemia. Med. Mycol. 2019, 57, 659–667.
  48. Liu, F.; Zhong, L.; Zhou, F.; Zheng, C.; Zhang, K.; Cai, J.; Zhou, H.; Tang, K.; Dong, Z.; Cui, W.; et al. Clinical features, strain distribution, antifungal resistance and prognosis of patients with non-albicans candidemia: A retrospective observational study. Infect. Drug Resist. 2021, 14, 3233–3246.
  49. Díez, A.; Carrano, G.; Bregón-Villahoz, M.; Cuétara, M.S.; García-Ruiz, J.C.; Fernandez-de-Larrinoa, I.; Moragues, M.D. Biomarkers for the diagnosis of invasive candidiasis in immunocompetent and immunocompromised patients. Diagn. Microbiol. Infect. Dis. 2021, 101, 115509.
  50. Calandra, T.; Roberts, J.A.; Antonelli, M.; Bassetti, M.; Vincent, J.L. Diagnosis and management of invasive candidiasis in the ICU: An updated approach to an old enemy. Crit. Care 2016, 20, 125.
  51. Pappas, P.G.; Kauffman, C.A.; Andes, D.R.; Clancy, C.J.; Marr, K.A.; Ostrosky-Zeichner, L.; Reboli, A.C.; Schuster, M.G.; Vazquez, J.A.; Walsh, T.J.; et al. Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 62, e1–e50.
  52. Krifors, A.; Ullberg, M.; Castegren, M.; Petersson, J.; Sparrelid, E.; Hammarström, H.; Sjölin, J.; Özenci, V.; Blennow, O. T2Candida assay in the diagnosis of intraabdominal candidiasis: A prospective multicenter study. J. Fungi 2022, 8, 86.
  53. Jung, P.; Mischo, C.E.; Gunaratnam, G.; Spengler, C.; Becker, S.L.; Hube, B.; Jacobs, K.; Bischoff, M. Candida albicans adhesion to central venous catheters: Impact of blood plasma-driven germ tube formation and pathogen-derived adhesins. Virulence 2020, 11, 1453–1465.
  54. Kollef, M.; Micek, S.; Hampton, N.; Doherty, J.A.; Kumar, A. Septic shock attributed to Candida infection: Importance of empiric therapy and source control. Clin. Infect. Dis. 2012, 54, 1739–1746.
  55. Vergidis, P.; Clancy, C.J.; Shields, R.K.; Park, S.Y.; Wildfeuer, B.N.; Simmons, R.L.; Nguyen, M.H. Intra-abdominal candidiasis: The importance of early source control and antifungal treatment. PLoS ONE 2016, 11, e0153247.
  56. Andes, D.R.; Safdar, N.; Baddley, J.W.; Playford, G.; Reboli, A.C.; Rex, J.H.; Sobel, J.D.; Pappas, P.G.; Kullberg, B.J.; Mycoses Study Group. Impact of treatment strategy on outcomes in patients with candidemia and other forms of invasive candidiasis: A patient-level quantitative review of randomized trials. Clin. Infect. Dis. 2012, 54, 1110–1122.
  57. Cornely, O.A.; Bassetti, M.; Calandra, T.; Garbino, J.; Kullberg, B.J.; Lortholary, O.; Meersseman, W.; Akova, M.; Arendrup, M.C.; Arikan-Akdagli, S.; et al. ESCMID* guideline for the diagnosis and management of Candida diseases 2012: Non-neutropenic adult patients. Clin. Microbiol. Infect. 2012, 18, 19–37.
  58. Cavassin, F.B.; Baú-Carneiro, J.L.; Vilas-Boas, R.R.; Queiroz-Telles, F. Sixty years of amphotericin B: An overview of the main antifungal agent used to treat invasive fungal infections. Infect. Dis. Ther. 2021, 10, 115–147.
  59. Anderson, T.M.; Clay, M.C.; Cioffi, A.G.; Diaz, K.A.; Hisao, G.S.; Tuttle, M.D.; Nieuwkoop, A.J.; Comellas, G.; Maryum, N.; Wang, S.; et al. Amphotericin forms an extramembranous and fungicidal sterol sponge. Nat. Chem. Biol. 2014, 10, 400–406.
  60. Robbins, N.; Caplan, T.; Cowen, L.E. Molecular evolution of antifungal drug resistance. Annu. Rev. Microbiol. 2017, 71, 753–775.
  61. Apsemidou, A.; Füller, M.A.; Idelevich, E.A.; Kurzai, O.; Tragiannidis, A.; Groll, A.H. Candida lusitaniae breakthrough fungemia in an immuno-compromised adolescent: Case report and review of the literature. J. Fungi 2020, 6, 380.
  62. Faustino, C.; Pinheiro, L. Lipid systems for the delivery of amphotericin B in antifungal therapy. Pharmaceutics 2020, 12, 29.
  63. Abdel-Hafez, Y.; Siaj, H.; Janajri, M.; Abu-Baker, Y.; Nazzal, Z.; Hamdan, Z.; Adwan, R.; Aiesh, B.M.; Anaya, A.I. Tolerability and epidemiology of nephrotoxicity associated with conventional amphotericin B therapy: A retrospective study in tertiary care centers in Palestine. BMC Nephrol. 2022, 23, 132.
  64. Caputo, R.; Asprea, M.; Giovannetti, L.; Messori, A. Nephrotoxicity of three formulations of amphotericin B: Trial sequential analysis. Arch. Med. Sci. 2020, 16, 1493–1495.
  65. Zhang, H.; Qu, W.; Nazzal, M.; Ortiz, J. Burn patients with history of kidney transplant experience increased incidence of wound infection. Burns 2020, 46, 609–615.
  66. Peyclit, L.; Yousfi, H.; Rolain, J.-M.; Bittar, F. Drug repurposing in medical mycology: Identification of compounds as potential antifungals to overcome the emergence of multidrug-resistant fungi. Pharmaceuticals 2021, 14, 488.
  67. Delma, F.Z.; Al-Hatmi, A.M.S.; Brüggemann, R.J.M.; Melchers, W.J.G.; de Hoog, S.; Verweij, P.E.; Buil, J.B. Molecular mechanisms of 5-Fluorocytosine resistance in yeasts and filamentous fungi. J. Fungi 2021, 7, 909.
  68. Botero-Calderon, L.; Benjamin, D.K., Jr.; Cohen-Wolkowiez, M. Advances in the treatment of invasive neonatal candidiasis. Expert Opin. Pharmacother. 2015, 16, 1035–1048.
  69. Testoni, D.; Smith, P.B.; Benjamin, D.K., Jr. The use of antifungal therapy in neonatal intensive care. Clin. Perinatol. 2012, 39, 83–98.
  70. Vermes, A.; van Der Sijs, H.; Guchelaar, H.J. Flucytosine: Correlation between toxicity and pharmacokinetic parameters. Chemotherapy 2000, 46, 86–94.
  71. Vitale, R.G. Role of antifungal combinations in difficult to treat Candida infections. J. Fungi 2021, 7, 731.
  72. Hassanmoghadam, F.; Shokohi, T.; Hedayati, M.T.; Aslani, N.; Haghani, I.; Nabili, M.; Lotfali, E.; Davari, A.; Moazeni, M. High prevalence of itraconazole resistance among Candida parapsilosis isolated from Iran. Curr. Med. Mycol. 2019, 5, 43–46.
  73. Galia, L.; Pezzani, M.D.; Compri, M.; Callegari, A.; Rajendran, N.B.; Carrara, E.; Tacconelli, E.; The COMBACTE MAGNET EPI-Net Network. Surveillance of Antifungal Resistance in Candidemia Fails to Inform Antifungal Stewardship in European Countries. J. Fungi 2022, 8, 249.
  74. Berkow, E.L.; Lockhart, S.R. Fluconazole resistance in Candida species: A current perspective. Infect. Drug Resist. 2017, 10, 237–245.
  75. Whaley, S.G.; Berkow, E.L.; Rybak, J.M.; Nishimoto, A.T.; Barker, K.S.; Rogers, P.D. Azole antifungal resistance in Candida albicans and emerging non-albicans Candida species. Front. Microbiol. 2017, 7, 2173.
  76. Ham, Y.Y.; Lewis, J.S.; Thompson, G.R. Rezafungin: A novel antifungal for the treatment of invasive candidiasis. Future Microbiol. 2021, 16, 27–36.
  77. Miesel, L.; Lin, K.Y.; Ong, V. Rezafungin treatment in mouse models of invasive candidiasis and aspergillosis: Insights on the PK/PD pharmacometrics of rezafungin efficacy. Pharmacol. Res. Perspect. 2019, 7, e00546.
  78. Miesel, L.; Cushion, M.T.; Ashbaugh, A.; Lopez, S.R.; Ong, V. Efficacy of rezafungin in prophylactic mouse models of invasive Candidiasis, Aspergillosis, and Pneumocystis pneumonia. Antimicrob. Agents Chemother. 2021, 65, e01992-20.
  79. Lepak, A.J.; Zhao, M.; Andes, D.R. Determination of pharmacodynamic target exposures for rezafungin against Candida tropicalis and Candida dubliniensis in the neutropenic mouse disseminated candidiasis model. Antimicrob. Agents Chemother. 2019, 63, e01556-19.
  80. Pfaller, M.A.; Carvalhaes, C.; Messer, S.A.; Rhomberg, P.R.; Castanheira, M. Activity of a long-acting echinocandin, rezafungin, and comparator antifungal agents tested against contemporary unvasive fungal isolates (SENTRY program, 2016 to 2018). Antimicrob. Agents Chemother. 2020, 64, e00099-20.
  81. Farhadi, Z.; Farhadi, T.; Hashemian, S.M. Virtual screening for potential inhibitors of β(1,3)-D-glucan synthase as drug candidates against fungal cell wall. J. Drug Assess. 2020, 9, 52–59.
  82. Szymański, M.; Chmielewska, S.; Czyżewska, U.; Malinowska, M.; Tylicki, A. Echinocandins—Structure, mechanism of action and use in antifungal therapy. J. Enzym. Inhib. Med. Chem. 2022, 37, 876–894.
  83. Sakai, D.; Matsumiya, W.; Kusuhara, S.; Nakamura, M. Factors associated with the development of ocular candidiasis and ocular prognosis with echinocandin therapy for candidemia. J. Ophthal. Inflamm. Infect. 2021, 11, 17.
  84. Hautala, N.; Köykkä, H.; Siiskonen, M.; Saari, J.; Kauranen, J.; Hautala, T. Effect of first-line antifungal treatment on ocular complication risk in Candida or yeast blood stream infection. BMJ Open Ophthalmol. 2021, 6, e000837.
  85. Taur, Y.; Cohen, N.; Dubnow, S.; Paskovaty, A.; Seo, S.K. Effect of antifungal therapy timing on mortality in cancer patients with candidemia. Antimicrob Agents Chemother. 2010, 54, 184–190.
  86. Mroczyńska, M.; Brillowska-Dąbrowska, A. Review on current status of echinocandins use. Antibiotic 2020, 9, 227.
  87. Bretagne, S.; Desnos-Ollivier, M.; Sitbon, K.; Lortholary, O.; Che, D.; Dromer, F.; Participants of the YEASTS. No impact of fluconazole to echinocandins replacement as first-line therapy on the epidemiology of yeast fungemia (Hospital-Driven Active Surveillance, 2004–2017, Paris, France). Front. Med. 2021, 8, 641965.
  88. Bienvenu, A.L.; Pradat, P.; Guerin, C.; Aubrun, F.; Fellahi, J.L.; Friggeri, A.; Guichon, C.; Hernu, R.; Menotti, J.; Monard, C.; et al. Evaluation of first-line therapies for the treatment of candidemia in ICU patients: A propensity score analysis. Int. J. Infect. Dis. 2020, 93, 15–21.
  89. Reboli, A.C.; Shorr, A.F.; Rotstein, C.; Pappas, P.G.; Kett, D.H.; Schlamm, H.T.; Reisman, A.L.; Biswas, P.; Walsh, T.J. Anidulafungin compared with fluconazole for treatment of candidemia and other forms of invasive candidiasis caused by Candida albicans: A multivariate analysis of factors associated with improved outcome. BMC Infect. Dis. 2011, 11, 261.
  90. Kullberg, B.J.; Sobel, J.D.; Ruhnke, M.; Pappas, P.G.; Viscoli, C.; Rex, J.H.; Cleary, J.D.; Rubinstein, E.; Church, L.W.; Brown, J.M.; et al. Voriconazole versus a regimen of amphotericin B followed by fluconazole for candidaemia in non-neutropenic patients: A randomised non-inferiority trial. Lancet 2005, 366, 1435–1442.
  91. Vazquez, J.; Reboli, A.C.; Pappas, P.G.; Patterson, T.F.; Reinhardt, J.; Chin-Hong, P.; Tobin, E.; Kett, D.H.; Biswas, P.; Swanson, R. Evaluation of an early step-down strategy from intravenous anidulafungin to oral azole therapy for the treatment of candidemia and other forms of invasive candidiasis: Results from an open-label trial. BMC Infect. Dis. 2014, 14, 97.
  92. Pappas, P.G.; Rotstein, C.M.; Betts, R.F.; Nucci, M.; Talwar, D.; De Waele, J.J.; Vazquez, J.A.; Dupont, B.F.; Horn, D.L.; Ostrosky-Zeichner, L.; et al. Micafungin versus caspofungin for treatment of candidemia and other forms of invasive candidiasis. Clin. Infect. Dis. 2007, 45, 883–893.
  93. Kullberg, B.J.; Viscoli, C.; Pappas, P.G.; Vazquez, J.; Ostrosky-Zeichner, L.; Rotstein, C.; Sobel, J.D.; Herbrecht, R.; Rahav, G.; Jaruratanasirikul, S.; et al. Isavuconazole versus caspofungin in the treatment of candidemia and other invasive candida infections: The ACTIVE trial. Clin. Infect. Dis. 2019, 68, 1981–1989.
  94. Fernández-Ruiz, M.; Aguado, J.M.; Almirante, B.; Lora-Pablos, D.; Padilla, B.; Puig-Asensio, M.; Montejo, M.; García-Rodríguez, J.; Pemán, J.; Ruiz Pérez de Pipaón, M.; et al. Initial use of echinocandins does not negatively influence outcome in Candida parapsilosis bloodstream infection: A propensity score analysis. Clin. Infect. Dis. 2014, 58, 1413–1421.
  95. Kontoyiannis, D.P.; Bassetti, M.; Nucci, M.; Capparella, M.R.; Yan, J.L.; Aram, J.; Hogan, P.A. Anidulafungin for the treatment of candidaemia caused by Candida parapsilosis: Analysis of pooled data from six prospective clinical studies. Mycoses 2017, 60, 663–667.
  96. Chiotos, K.; Vendetti, N.; Zaoutis, T.E.; Baddley, J.; Ostrosky-Zeichner, L.; Pappas, P.; Fisher, B.T. Comparative effectiveness of echinocandins versus fluconazole therapy for the treatment of adult candidaemia due to Candida parapsilosis: A retrospective observational cohort study of the Mycoses Study Group (MSG-12). J. Antimicrob. Chemother. 2016, 71, 3536–3539.
  97. Kullberg, B.J.; Vasquez, J.; Mootsikapun, P.; Nucci, M.; Paiva, J.A.; Garbino, J.; Yan, J.L.; Aram, J.; Capparella, M.R.; Conte, U.; et al. Efficacy of anidulafungin in 539 patients with invasive candidiasis: A patient-level pooled analysis of six clinical trials. J. Antimicrob. Chemother. 2017, 72, 2368–2377.
  98. Pfaller, M.A.; Diekema, D.J.; Gibbs, D.L.; Newell, V.A.; Nagy, E.; Dobiasova, S.; Rinaldi, M.; Barton, R.; Veselov, A.; Global Antifungal Surveillance Group. Candida krusei, a multidrug-resistant opportunistic fungal pathogen: Geographic and temporal trends from the ARTEMIS DISK Antifungal Surveillance Program, 2001 to 2005. J. Clin. Microbiol. 2008, 46, 515–521.
  99. Ademe, M.; Girma, F. Candida auris: From multidrug resistance to pan-resistant strains. Infect. Drug Resist. 2020, 13, 1287–1294.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 439
Revisions: 2 times (View History)
Update Date: 21 Jun 2022
1000/1000
Video Production Service