Next Article in Journal / Special Issue
Antifungal Susceptibility Testing of Fusarium: A Practical Approach
Previous Article in Journal / Special Issue
Molecular Identification and Antifungal Susceptibility Patterns of Clinical Dermatophytes Following CLSI and EUCAST Guidelines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reduced Multidrug Susceptibility Profile Is a Common Feature of Opportunistic Fusarium Species: Fusarium Multi-Drug Resistant Pattern

1
Mycology Unit, Microbiology Division, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar
2
Weill Cornell Medicine-Qatar, P.O. Box 3050 Doha, Qatar
J. Fungi 2017, 3(2), 18; https://doi.org/10.3390/jof3020018
Submission received: 26 February 2017 / Revised: 27 March 2017 / Accepted: 7 April 2017 / Published: 10 April 2017
(This article belongs to the Special Issue Antifungal Susceptibility Testing)

Abstract

:
The resistance among various opportunistic Fusarium species to different antifungal agents has emerged as a cause of public health problems worldwide. Considering the significance of multi-drug resistant (MDR), this paper emphasizes the problems associated with MDR and the need to understand its clinical significance to combat microbial infections. The search platform PubMed/MEDLINE and a review of 32 cases revealed a common multidrug-resistant profile exists, and clinically relevant members of Fusarium are intrinsically resistant to most currently used antifungals. Dissemination occurs in patients with prolonged neutropenia, immune deficiency, and especially hematological malignancies. Amphotericin B displayed the lowest minimum inhibitory concentrarions (MICs) followed by voriconazole, and posaconazole. Itraconazole and fluconazole showed high MIC values, displaying in vitro resistance. Echinocandins showed the highest MIC values. Seven out of ten (70%) patients with neutropenia died, including those with fungemia that progressed to skin lesions. Clinical Fusarium isolates displayed a common MDR profile and high MIC values for the most available antifungal agents with species- and strain-specific differences in antifungal susceptibility. Species identification of Fusarium infections is important. While the use of natamycin resulted in a favorable outcome in keratitis, AmB and VRC are the most used agents for the treatment of fusariosis in clinical settings.

1. Introduction

Fusariuma is a serious opportunistic human pathogen affecting immunocompromised patients [1,2] and represents the second most common cause of filamentous fungi infections after aspergillosis [3,4]. Fusarium species cause a broad spectrum of infections in humans; the invasive and disseminated infections occur predominantly in severely immunocompromised individuals [1,5]. These infections may manifest as a fever that does not respond to antimicrobial therapy [6]. Underlying diseases for development of invasive fusariosis are hematologic malignancies, hematopoietic cell transplantation, neutropenia, and impaired cellular immunity [5,6,7,8,9]. Infections may also occur in immunocompetent individuals [10]. Localized infections, such as onychomycosis, keratitis or endopthalmitis and other skin infections, are frequently manifested in immunocompetent individuals and are often associated with previous trauma [11,12,13,14,15]. Fusarium species are genetically diverse and are commonly environmental organisms including phytopathogens, saprophytes and those isolated from water systems [1,16,17]. Although Fusarium species are plant pathogens, they may occasionally cause infections in animals [18,19], Fusarium from veterinary sources was previously known to infect humans [20]. This hyaline hyphomycete fungus poses a challenge for human disease management because conidia can disperse in the atmosphere over a long distance and infect a new susceptible host [21].
Fusarium species are among the most resistant fungi; infections are commonly refractory against treatment with the most known systemic and conventional antifungal agents [22]. Estimated mortality rates of 50–75% in disseminated infections may arise, especially in immunosuppressed patients [1,5,8,23]. Fusarium pathogens typically show broad in vitro resistance to antifungal agents with a high variability being present within each species [24,25,26,27,28,29]. In vitro susceptibility testing may represent a tool for the selection of an appropriate therapy. In general, members of the Fusarium solani species complex (FSSC) are most commonly observed in all clinical infections and show the highest minimum inhibitory concentrations (MICs) against various antifungal drugs. Amphotericin B (AmB) is the most active drug followed by voriconazole (VRC) [27,29,30]. Members of the Fusarium fujikuroi species complex (FFSC), which are responsible for approximately one-third of the disseminated infections, display a similar susceptibility profile [25]. Based on such in vitro susceptibility data for various Fusarium species, the optimal treatment options remain limited and controversial. ESCMID and ECMM joint guidelines suggested that early therapy with VRC or lipid-based (LAmB) in conjunction with surgical intervention is of utmost importance to prevent dissemination [31]. Most of the available MIC data on AmB and triazoles have been reported for a variety of Fusarium species, using the CLSI-based methods [24,28,32,33]. This paper reviews the most recent cases and discusses the susceptibility and multidrug resistant patterns of clinically important Fusarium species in response to the most available systemic antifungal agents.

2. Definitions

Invasive Fusarium infection (fusariosis) is infection with at least one positive blood culture or the isolation of the same strain from two or more body sites [29]. Localized diseases, as defined previously [29], are infections of the skin, nail, cornea and wound without deep tissue involvement. Proven Fusarium infection requires the visualization of fungal elements by direct microscopy of the clinical specimens, the isolation of the fungal etiology in culture, and compatibility with infectious disease processes [34]. Multidrug resistance (MDR) can be defined as the broad-spectrum tolerance to antifungal agents; more precisely, the phenotype of non-susceptibility to at least one agent from two antifungal classes (e.g., azoles and echinocandins). Antifungal resistance is associated with a high mortality rate and increases the length of hospital stays, resulting in a high cost (Figure 1). MDR resistance decreases the efficacy of treatment, and hence, results in a prolonged time of infection. MDR resistance in Fusarium can be classified into:
(i)
Intrinsic resistance: the innate ability of a Fusarium species to resist activity of an antifungal agent through its inherent structural or functional characteristics without prior exposure to the drug, which allows tolerance of a drug or antifungal class. It occurs naturally in Fusarium species that have never been susceptible to that agent [25,35,36].
(ii)
Acquired resistance: used to describe the resistance that arises in Fusarium after exposure to the antifungal agent. The development of resistance and infection recurrence after drug discontinuation [37] and fungal breakthrough infections associated with posaconazole prophylaxis [38] suggest the emergence of resistant strains.
(iii)
Clinical resistance: a situation in which the infecting Fusarium species is inhibited by the concentration of an antifungal agent that is associated with therapeutic failure or reappearance of infections. Such failure can be attributed to a combination of factors related to impaired host immune function, Fusarium species, or the antifungal agent [39,40].

3. Clinical Significance

Fusarium species cause a broad spectrum of infections in humans. The focus will be on two devastating infections caused by Fusarium species: disseminated fusariosis, which may lead to an unfavorable outcome, and keratitis, which causes injury that may progress to the loss of sight. Fusarium has been found in environmental samples and is widely distributed in soil, plants, plant debris and other organic substrates, as well as in water systems, including the plumbing systems of hospitals [41]. However, it was demonstrated that the infection is a community rather than hospital acquisition, based on a study that investigated the environmental sources of Fusarium infections in a tertiary-care center [42]. The first case of disseminated fusariosis was described in 1973 [43]. Since then, there was a significant increase in the occurrence of disseminated disease, probably reflecting the increase in the number of immunocompromised hosts. Reports have focused on two primary groups: those with hematologic malignancies, susceptible secondarily to neutropenia, and bone marrow transplant recipients. Disseminated infection is characterized by persistent fever not responding to antibiotic treatment and by diffuse metastatic skin lesions with a dark purple central necrosis surrounded by an erythematous ring (Figure 2). The cutaneous lesions are observed in approximately 85% of patients with disseminated Fusarium infections and often occur at an early stage of the disease [44], which is the most frequent presentation of disseminated fusariosis [1,3,5,36]. Patients with disseminated fusariosis have an unfavorable prognosis, and the survival rate may reach 33% [45]. Dissemination may proceed to cause intracranial infection; many proven Fusarium brain abscess infections mostly in patients with cancer or hematological disorders were reported in the literature [10,46,47,48,49]. Fusarium may cause serious osteomyelitis infections in immunosuppressed patients [50]. A review of bone and joint infections by filamentous fungi revealed (n = 14; 9.6%) cases caused by Fusarium species [51].
Although not completely elucidated, the role of the innate immunity and particularly the Toll-like receptors and T-cell defenses seems to be crucial in the progression of fusariosis [1]. The portal of entry of Fusarium species is often not clear. Possibilities include the respiratory tract, gastrointestinal tract, or catheter tip [52]. A toenail infection, or paronychia, in some predisposed individuals, may be the source of disseminated infection [53,54,55,56]. The presence of infections involving the skin or nails should be carefully investigated before initiating immunosuppressive therapy since it has been shown that such lesions can be a focal point for Fusarium dissemination.
Keratitis is still the most common infection caused by Fusarium species; the incidence is increasing in many areas of the world, especially in tropical areas [57,58,59] and in the USA [12,60]. Farm workers at are greater risk of corneal injury and exposure to airborne conidia [61]. Another risk factor for Fusarium keratitis is contact lens wearers and contact lens care solution, as in the Multistate outbreak of Fusarium keratitis, which was investigated by the CDC in 2006 [12]. The emergence of novel opportunists within the genus Fusarium has frequently been reported [62].

4. Susceptibility to Amphotericin B and Voriconazole and Clinical Response

Finding the optimal treatment strategy is a challenge because Fusarium species show reduced susceptibility to the most available antifungal agents. The reverse of immunosuppression [41] by the administration of granulocyte-macrophage colony stimulating factor (GM-CF), which is sufficient to render blood culture negative with relief of neutropenia [29], and simultaneous treatment with VRC or L-AmB, is recommended [31]. An indication of the potential correlation between MICs for Fusarium species and response to treatment was only found in a recent report investigating CLSI-based MICs for seven Fusarium isolates, and the clinical response was documented for patients with invasive fusariosis [42]. Fusarium susceptibility to VRC is variable [25,29,43]; several out of 16 F. proliferatum strains investigated displayed high MICs against VRC (n = 6) with values >16 µL/mL [30], and breakthrough infection has been reported in 16 out of 44 patients with invasive fusariosis—69% were receiving prophylaxis with VRC (8/16; 50%) or posaconazole (POS) (3/16; 19%) [23].
AmB was most active agent against clinical and reference strains in vitro with MICs ranging from 0.062–2 µg/mL [29], whereas an MIC range from 1 to 8 µg/mL was reported for most species and may not be related to clinical outcomes [25,36]. This suggests that the role of any in vitro data for AmB is controversial. Treatment failed in one of the two patients with disseminated fusariosis who received AmB therapy [29]. Important parameters may influence the outcome of an infection, e.g., drug doses, treatment duration, and drug serum levels.
Due to the poor prognosis obtained with monotherapy, combination therapy may be considered in severe Fusarium species infections. In vitro susceptibility of AmB plus VRC has shown favorable results [44], in addition to immunocompromised patients [21,45]. In vitro combination of antifungal activity, of natamycin and VRC for fungal keratitis displayed 70% synergistic effects against a significant number of isolates [46]. Combination therapy for disseminated fusariosis in immunocompromised patients was previously reviewed [47]; the patients in 14/20 (70%) cases had favorable treatment responses. All patients in these 14 case reports had underlying hematologic diseases, among which four (29%) underwent hematopoietic stem cell transplantation prior to the disseminated infection. The most common combination regimens used in these 14 cases with successful responses were L-AmB plus VRC (5/14, 36%), followed by AmB deoxycholate plus VRC (2/14, 15%) and L-AmB plus terbinafine (2/14, 15%) [47].
In vitro antifungal susceptibility of clinical Fusarium species revealed that AmB displayed a lower MIC compared with VRC, and reference Fusarium species exhibited lower MIC values than the clinical isolates (Figure 3). As concluded earlier [29], this is probably due to previous exposure to antifungal therapy in clinical settings, as most of the Westerdijk Fungal Biodiversity Center (previously CBS) reference strains were isolated from the environment and collected in the era of pre-antifungal use. Interestingly, the two strains from the Westerdijk Fungal Biodiversity Center of human origin displayed high MIC values to the antifungal agents tested [29].
The outcome is improved in disseminated fusariosis; there has been a 21% increase in the survival rate in the last decade due to changes in the treatment practice by shifting therapy to L-AmB and VRC [48].

5. Multi-Drug Resistant Cases

The prognosis may be not always favorable when Fusarium species exhibit a reduced susceptibility profile. To identify infections caused by Fusarium species, the PubMed/MEDLINE database was searched using the keywords, Fusarium infections AND fungemia, disseminated fusariosis, Fusarium susceptibility and Fusarium keratitis for the years “Jan. 2011—Feb. 2017”. Reports included cases in the final analysis with data on the site of infection, underlying disease, etiologic agent, antifungal susceptibility and medical and surgical therapy. Exclusion criteria were cases of non-English literature, incomplete identification of the etiologic agent of the disease, cases missing full text, and cases without Fusarium susceptibility data. Descriptive statistics were used to summarize all demographic and clinical characteristics of the patients, and p values <0.05 were considered as statistically significant. Analysis was performed using the statistical packages SPSS 19.0 (SPSS Inc. Chicago, IL, USA). The study was approved by the local ethics committee, Medical Research Center (MRC) at Hamad Medical Corporation, Project #16149/16.
Thirty-two of the most recent proven cases of invasive and localized fusariosis were reviewed from the literature. Information pertaining to the source of Fusarium species isolation, demographic and clinical data of the patients yielding these isolates is provided in Table 1. Twenty (62.5%) of the cases were proven invasive fusariosis, including fungemia and disseminated infections, and 12 (37.5%) were localized Fusarium infections, including mycetoma, keratitis, skin, and onychomycosis. Etiologic agents were F. solani species complex (FSSC) n = 19, F. fujikuroi species complex (FFSC) n = 12, and F. oxysporum species complex (FOSC) n = 1. The patients were treated with AmB, VRC or a combination treatment with a variable degree of responses, and the mortality was 60% (n = 12/20) of the total invasive disease cases irrespective of antifungal therapy. Underlying conditions for invasive infections were hematological malignancy, transplant patients, and autoimmune diseases, and ten patients were receiving prophylaxis with antifungal drugs at the time of onset of the invasive fusariosis (Table 1). Seven out of ten (70%) patients with neutropenia died, including those with fungemia that progressed to skin lesions. The reported mortality might be 100% for persistent neutropenic patients with disseminated lesions [1] and 50%–75% for patients with disseminated fusariosis [63].
The in vitro susceptibility pattern of Fusarium isolates obtained from these cases showed a multidrug resistant profile (Table 2). Although the new echinocandins drugs such as anidulafungin, micafungin and caspofungin are very important for treating common Aspergillus and Candida infections, they are inactive for Fusarium species [25,64,65,66,67], except for F. temperatum case # 21 (Table 2). Hence, it is not recommended in the clinical settings to waste efforts testing Fusarium species against echinocandins agents. Although we lack clinical break points for Fusarium species and antifungal agents, CLSI epidemiological cutoff values (ECVs) were established for members of the more common Fusarium species complexes [68]. The lowest MIC of 0.5 µg/mL AmB was recorded for 7 cases (Table 2), and all FSSC isolates showed MICs within the wild type range ≤ECV value (8 µg/mL) for AmB [68]. The two isolates of F. verticillioides (cases 22 & 31) showed high MICs for AmB, above the ECV (4 µg/mL), but case 22 exhibited a low MIC for VRC (1 µg/mL) which is within the wild type range, lower than the reported ECV value for this species (4 µg/mL) [68]. All 19 isolates of the reported FSSC fell within the wild type range for VRC (ECV value = 32 µg/mL), whereas, 11/13 isolates of FSSC tested for POS showed MIC values within the wild type range (≤ the ECV value 32 µg/mL). Overall, FLC and ITC showed poor activity with high MIC values for all the reported cases.
The new triazole antifungal, isavuconazole (ISV), was recently granted approval by the US Food and Drug Administration and the European Medicines Agency for the treatment of invasive aspergillosis and mucormycosis. A randomized, double-blind comparison trial for the treatment of invasive aspergillosis found ISV noninferior to voriconazole. Evaluating the efficacy of ISV in the treatment of mucormycosis revealed comparable response rates to AmB and POS [69,70]. ISV MICs were reported for 7 cases (Table 2) with a range of 4– > 16 µg/mL; there is no controlled trial on the use of ISV in invasive fusariosis, but the MIC values reported for these cases are not encouraging.

6. Conclusions

Successful treatment of invasive disease was achieved with the use of AmB and its liposomal form, VRC or combination therapy in addition to GM-CF. In some patients, treatment failed, but others improved (Table 1). Although they are the recommended agents in the ESCMID and ECMM joint guidelines [31], the efficacy of AmB and VRC for treating invasive fusariosis is still controversial as the percentage of patients cured in the cases of Table 1 and in different clinical trials is low [22]. It is difficult to draw a clear correlation of in vitro and in vivo obtained data since many factors influence the outcome of an infection, such as, treatment duration, drug doses, and drug serum level, which are all important parameters. Members of FSSC were the main agents of invasive infections, and they are less susceptible to VRC; these observations agree with the data of a previous report [29]. AmB is the most active agent against Fusarium species; clinical Fusarium isolates displayed a common MDR profile and high MIC values for the most available antifungal agents with species- and strain-specific differences in antifungal susceptibility. Species identification of Fusarium infections is important and may be erroneous or missed in many diagnostic laboratories, which can greatly affect the choice of an appropriate antifungal therapy. These observations emphasize the need to further understand the mechanism of Fusarium resistance to combat invasive infections.

Acknowledgments

The Qatar National Research Fund (a member of Qatar Foundation) supported this research (grant number NPRP9-094-3-017).

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Nucci, M.; Anaissie, E. Fusarium infections in immunocompromised patients. Clin. Microbiol. Rev. 2007, 20, 695–704. [Google Scholar] [CrossRef] [PubMed]
  2. Kredics, L.; Narendran, V.; Shobana, C.S.; Vagvolgyi, C.; Manikandan, P.; Indo-Hungarian Fungal Keratitis Working Group. Filamentous fungal infections of the cornea: A global overview of epidemiology and drug sensitivity. Mycoses 2015, 58, 243–260. [Google Scholar] [CrossRef] [PubMed]
  3. Bodey, G.P.; Boktour, M.; Mays, S.; Duvic, M.; Kontoyiannis, D.; Hachem, R.; Raad, I. Skin lesions associated with Fusarium infection. J. Am. Acad. Dermatol. 2002, 47, 659–666. [Google Scholar] [CrossRef] [PubMed]
  4. Cooke, N.S.; Feighery, C.; Armstrong, D.K.; Walsh, M.; Dempsey, S. Cutaneous Fusarium solani infection in childhood acute lymphoblastic leukaemia. Clin. Exp. Dermatol. 2009, 34, e117–e119. [Google Scholar] [CrossRef] [PubMed]
  5. Muhammed, M.; Anagnostou, T.; Desalermos, A.; Kourkoumpetis, T.K.; Carneiro, H.A.; Glavis-Bloom, J.; Coleman, J.J.; Mylonakis, E. Fusarium infection: Report of 26 cases and review of 97 cases from the literature. Medicine 2013, 92, 305–316. [Google Scholar] [CrossRef] [PubMed]
  6. Nucci, M.; Anaissie, E.J.; Queiroz-Telles, F.; Martins, C.A.; Trabasso, P.; Solza, C.; Mangini, C.; Simoes, B.P.; Colombo, A.L.; Vaz, J.; et al. Outcome predictors of 84 patients with hematologic malignancies and Fusarium infection. Cancer 2003, 98, 315–319. [Google Scholar] [CrossRef] [PubMed]
  7. Avelino-Silva, V.I.; Ramos, J.F.; Leal, F.E.; Testagrossa, L.; Novis, Y.S. Disseminated Fusarium infection in autologous stem cell transplant recipient. Braz. J. Infect. Dis. 2015, 19, 90–93. [Google Scholar] [CrossRef] [PubMed]
  8. Nucci, M.; Marr, K.A.; Queiroz-Telles, F.; Martins, C.A.; Trabasso, P.; Costa, S.; Voltarelli, J.C.; Colombo, A.L.; Imhof, A.; Pasquini, R.; et al. Fusarium infection in hematopoietic stem cell transplant recipients. Clin. Infect. Dis. 2004, 38, 1237–1242. [Google Scholar] [CrossRef] [PubMed]
  9. Delia, M.; Monno, R.; Giannelli, G.; Ianora, A.A.; Dalfino, L.; Pastore, D.; Capolongo, C.; Calia, C.; Tortorano, A.; Specchia, G. Fusariosis in a patient with acute myeloid leukemia: A case report and review of the literature. Mycopathologia 2016, 181, 457–463. [Google Scholar] [CrossRef] [PubMed]
  10. Peterson, A.; Pham, M.H.; Lee, B.; Commins, D.; Cadden, J.; Giannotta, S.L.; Zada, G. Intracranial fusarium fungal abscess in an immunocompetent patient: Case report and review of the literature. J. Neurol. Surg. Rep. 2014, 75, e241–e245. [Google Scholar] [PubMed]
  11. Takenaka, M. A case of cutaneous fusariosis of the scrotum as a complication of acute myeloid leukemia. Med. Mycol. J. 2016, 57, J65–J70. [Google Scholar] [CrossRef] [PubMed]
  12. Chang, D.C.; Grant, G.B.; O’Donnell, K.; Wannemuehler, K.A.; Noble-Wang, J.; Rao, C.Y.; Jacobson, L.M.; Crowell, C.S.; Sneed, R.S.; Lewis, F.M.; et al. Multistate outbreak of Fusarium keratitis associated with use of a contact lens solution. JAMA 2006, 296, 953–963. [Google Scholar] [CrossRef] [PubMed]
  13. Godoy, P.; Cano, J.; Gene, J.; Guarro, J.; Hofling-Lima, A.L.; Lopes Colombo, A. Genotyping of 44 isolates of Fusarium solani, the main agent of fungal keratitis in Brazil. J. Clin. Microbiol. 2004, 42, 4494–4497. [Google Scholar] [CrossRef] [PubMed]
  14. Zaias, N.; Escovar, S.X.; Rebell, G. Opportunistic toenail onychomycosis. The fungal colonization of an available nail unit space by non-dermatophytes is produced by the trauma of the closed shoe by an asymmetric gait or other trauma. A plausible theory. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 1002–1006. [Google Scholar] [CrossRef] [PubMed]
  15. Morales-Cardona, C.A.; Valbuena-Mesa, M.C.; Alvarado, Z.; Solorzano-Amador, A. Non-dermatophyte mould onychomycosis: A clinical and epidemiological study at a dermatology referral centre in Bogota, Colombia. Mycoses 2014, 57, 284–293. [Google Scholar] [CrossRef] [PubMed]
  16. Anaissie, E.J.; Kuchar, R.T.; Rex, J.H.; Francesconi, A.; Kasai, M.; Muller, F.M.; Lozano-Chiu, M.; Summerbell, R.C.; Dignani, M.C.; Chanock, S.J.; et al. Fusariosis associated with pathogenic fusarium species colonization of a hospital water system: A new paradigm for the epidemiology of opportunistic mold infections. Clin. Infect. Dis. 2001, 33, 1871–1878. [Google Scholar] [CrossRef] [PubMed]
  17. Enoch, D.A.; Ludlam, H.A.; Brown, N.M. Invasive fungal infections: A review of epidemiology and management options. J. Med. Microbiol. 2006, 55, 809–818. [Google Scholar] [CrossRef] [PubMed]
  18. Cutuli, M.T.; Gibello, A.; Rodriguez-Bertos, A.; Blanco, M.M.; Villarroel, M.; Giraldo, A.; Guarro, J. Skin and subcutaneous mycoses in tilapia (Oreochromis niloticus) caused by Fusarium oxysporum in coinfection with aeromonas hydrophila. Med. Mycol. Case Rep. 2015, 9, 7–11. [Google Scholar] [CrossRef] [PubMed]
  19. Salter, C.E.; O’Donnell, K.; Sutton, D.A.; Marancik, D.P.; Knowles, S.; Clauss, T.M.; Berliner, A.L.; Camus, A.C. Dermatitis and systemic mycosis in lined seahorses hippocampus erectus associated with a marine-adapted fusarium solani species complex pathogen. Dis. Aquat. Organ. 2012, 101, 23–31. [Google Scholar] [CrossRef] [PubMed]
  20. O’Donnell, K.; Sutton, D.A.; Wiederhold, N.; Robert, V.A.; Crous, P.W.; Geiser, D.M. Veterinary fusarioses within the united states. J. Clin. Microbiol. 2016, 54, 2813–2819. [Google Scholar] [CrossRef] [PubMed]
  21. Brown, G.D.; Denning, D.W.; Gow, N.A.; Levitz, S.M.; Netea, M.G.; White, T.C. Hidden killers: Human fungal infections. Sci. Transl. Med. 2012, 4, 165rv113. [Google Scholar] [CrossRef] [PubMed]
  22. Guarro, J. Fusariosis, a complex infection caused by a high diversity of fungal species refractory to treatment. Eur. J. Clin. Microbiol. Infect. Dis. 2013, 32, 1491–1500. [Google Scholar] [CrossRef] [PubMed]
  23. Campo, M.; Lewis, R.E.; Kontoyiannis, D.P. Invasive fusariosis in patients with hematologic malignancies at a cancer center: 1998–2009. J. Infect. 2010, 60, 331–337. [Google Scholar] [CrossRef] [PubMed]
  24. Alastruey-Izquierdo, A.; Cuenca-Estrella, M.; Monzon, A.; Mellado, E.; Rodriguez-Tudela, J.L. Antifungal susceptibility profile of clinical Fusarium spp. Isolates identified by molecular methods. J. Antimicrob. Chemother. 2008, 61, 805–809. [Google Scholar] [CrossRef] [PubMed]
  25. Al-Hatmi, A.M.; van Diepeningen, A.D.; Curfs-Breuker, I.; de Hoog, G.S.; Meis, J.F. Specific antifungal susceptibility profiles of opportunists in the Fusarium fujikuroi complex. J. Antimicrob. Chemother. 2015, 70, 1068–1071. [Google Scholar] [CrossRef] [PubMed]
  26. Araujo, R.; Oliveira, M.; Amorim, A.; Sampaio-Maia, B. Unpredictable susceptibility of emerging clinical moulds to tri-azoles: Review of the literature and upcoming challenges for mould identification. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 1289–1301. [Google Scholar] [CrossRef] [PubMed]
  27. Azor, M.; Gene, J.; Cano, J.; Guarro, J. Universal in vitro antifungal resistance of genetic clades of the Fusarium solani species complex. Antimicrob. Agents Chemother. 2007, 51, 1500–1503. [Google Scholar] [CrossRef] [PubMed]
  28. Tortorano, A.M.; Prigitano, A.; Dho, G.; Esposto, M.C.; Gianni, C.; Grancini, A.; Ossi, C.; Viviani, M.A. Species distribution and in vitro antifungal susceptibility patterns of 75 clinical isolates of Fusarium spp. from Northern Italy. Antimicrob. Agents Chemother. 2008, 52, 2683–2685. [Google Scholar] [CrossRef] [PubMed]
  29. Taj-Aldeen, S.J.; Salah, H.; Al-Hatmi, A.M.; Hamed, M.; Theelen, B.; van Diepeningen, A.D.; Boekhout, T.; Lass-Florl, C. In vitro resistance of clinical fusarium species to Amphotericin B and voriconazole using the eucast antifungal susceptibility method. Diagn. Microbiol. Infect. Dis. 2016, 85, 438–443. [Google Scholar] [CrossRef] [PubMed]
  30. Tortorano, A.M.; Prigitano, A.; Esposto, M.C.; Arsic Arsenijevic, V.; Kolarovic, J.; Ivanovic, D.; Paripovic, L.; Klingspor, L.; Nordoy, I.; Hamal, P.; et al. European Confederation of Medical Mycology (ECMM) epidemiological survey on invasive infections due to Fusarium species in Europe. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1623–1630. [Google Scholar] [CrossRef] [PubMed]
  31. Tortorano, A.M.; Richardson, M.; Roilides, E.; van Diepeningen, A.; Caira, M.; Munoz, P.; Johnson, E.; Meletiadis, J.; Pana, Z.D.; Lackner, M.; et al. ESCMID and ECMM joint guidelines on diagnosis and management of hyalohyphomycosis: Fusarium spp., Scedosporium spp. and others. Clin. Microbiol. Infect. 2014, 20, 27–46. [Google Scholar] [CrossRef] [PubMed]
  32. Espinel-Ingroff, A.; Johnson, E.; Hockey, H.; Troke, P. Activities of voriconazole, itraconazole and Amphotericin B in vitro against 590 moulds from 323 patients in the voriconazole phase III clinical studies. J. Antimicrob. Chemother. 2008, 61, 616–620. [Google Scholar] [CrossRef] [PubMed]
  33. O’Donnell, K.; Sutton, D.A.; Fothergill, A.; McCarthy, D.; Rinaldi, M.G.; Brandt, M.E.; Zhang, N.; Geiser, D.M. Molecular phylogenetic diversity, multilocus haplotype nomenclature, and in vitro antifungal resistance within the Fusarium solani species complex. J. Clin. Microbiol. 2008, 46, 2477–2490. [Google Scholar] [CrossRef] [PubMed]
  34. De Pauw, B.; Walsh, T.J.; Donnelly, J.P.; Stevens, D.A.; Edwards, J.E.; Calandra, T.; Pappas, P.G.; Maertens, J.; Lortholary, O.; Kauffman, C.A.; et al. Revised definitions of invasive fungal disease from the European organization for research and treatment of cancer/invasive fungal infections cooperative group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) consensus group. Clin. Infect. Dis. 2008, 46, 1813–1821. [Google Scholar] [PubMed]
  35. Kebabci, N.; van Diepeningen, A.D.; Ener, B.; Ersal, T.; Meijer, M.; Al-Hatmi, A.M.; Ozkocaman, V.; Ursavas, A.; Cetinoglu, E.D.; Akalin, H. Fatal breakthrough infection with Fusarium andiyazi: New multi-resistant aetiological agent cross-reacting with aspergillus galactomannan enzyme immunoassay. Mycoses 2014, 57, 249–255. [Google Scholar] [CrossRef] [PubMed]
  36. Pereira, G.H.; de Angelis, D.A.; Brasil, R.A.; dos Anjos Martins, M.; de Matos Castro e Silva, D.; Szeszs, M.W.; de Souza Carvalho Melhem, M. Disseminated amphotericin-resistant fusariosis in acute leukemia patients: Report of two cases. Mycopathologia 2013, 175, 107–114. [Google Scholar] [CrossRef] [PubMed]
  37. Kandeel, A.; Abu-Elmagd, K.; Spinner, M.; Khanna, A.; Hashimoto, K.; Fujiki, M.; Parsi, M.; Bennett, A.; El-Gazzaz, G.; Abd-Elaal, A. Atypical clinical presentation of a newer generation anti-fungal drug-resistant Fusarium infection after a modified multi-visceral transplant. Ann. Transpl. 2015, 20, 512–518. [Google Scholar] [CrossRef] [PubMed]
  38. Bose, P.; Parekh, H.D.; Holter, J.L.; Greenfield, R.A. Disseminated fusariosis occurring in two patients despite posaconazole prophylaxis. J. Clin. Microbiol. 2011, 49, 1674–1675. [Google Scholar] [CrossRef] [PubMed]
  39. Loeffler, J.; Stevens, D.A. Antifungal drug resistance. Clin. Infect. Dis. 2003, 36, S31–S41. [Google Scholar] [CrossRef] [PubMed]
  40. Ersal, T.; Al-Hatmi, A.S.; Cilo, B.D.; Curfs-Breuker, I.; Meis, J.F.; Ozkalemkas, F.; Ener, B.; van Diepeningen, A.D. Fatal disseminated infection with Fusarium petroliphilum. Mycopathologia 2015, 179, 119–124. [Google Scholar] [CrossRef] [PubMed]
  41. Short, D.P.; O’Donnell, K.; Zhang, N.; Juba, J.H.; Geiser, D.M. Widespread occurrence of diverse human pathogenic types of the fungus Fusarium detected in plumbing drains. J. Clin. Microbiol. 2011, 49, 4264–4272. [Google Scholar] [CrossRef] [PubMed]
  42. Raad, I.; Tarrand, J.; Hanna, H.; Albitar, M.; Janssen, E.; Boktour, M.; Bodey, G.; Mardani, M.; Hachem, R.; Kontoyiannis, D.; et al. Epidemiology, molecular mycology, and environmental sources of Fusarium infection in patients with cancer. Infect. Control Hosp. Epidemiol. 2002, 23, 532–537. [Google Scholar] [CrossRef] [PubMed]
  43. Cho, C.T.; Vats, T.S.; Lowman, J.T.; Brandsberg, J.W.; Tosh, F.E. Fusarium solani infection during treatment for acute leukemia. J. Pediatr. 1973, 83, 1028–1031. [Google Scholar] [CrossRef]
  44. Jossi, M.; Ambrosioni, J.; Macedo-Vinas, M.; Garbino, J. Invasive fusariosis with prolonged fungemia in a patient with acute lymphoblastic leukemia: Case report and review of the literature. Int. J. Infect. Dis. 2010, 14, e354–e356. [Google Scholar] [CrossRef] [PubMed]
  45. Horn, D.L.; Freifeld, A.G.; Schuster, M.G.; Azie, N.E.; Franks, B.; Kauffman, C.A. Treatment and outcomes of invasive fusariosis: Review of 65 cases from the path Alliance® registry. Mycoses 2014, 57, 652–658. [Google Scholar] [CrossRef] [PubMed]
  46. Al-Hatmi, A.M.; Meletiadis, J.; Curfs-Breuker, I.; Bonifaz, A.; Meis, J.F.; De Hoog, G.S. In vitro combinations of natamycin with voriconazole, itraconazole and micafungin against clinical Fusarium strains causing keratitis. J. Antimicrob. Chemother. 2016, 71, 953–955. [Google Scholar] [CrossRef] [PubMed]
  47. Antunes, N.L.; Hariharan, S.; DeAngelis, L.M. Brain abscesses in children with cancer. Med. Pediatr. Oncol. 1998, 31, 19–21. [Google Scholar] [CrossRef]
  48. Kleinschmidt-Demasters, B.K. Disseminated Fusarium infection with brain abscesses in a lung transplant recipient. Clin. Neuropathol. 2009, 28, 417–421. [Google Scholar] [CrossRef] [PubMed]
  49. Steinberg, G.K.; Britt, R.H.; Enzmann, D.R.; Finlay, J.L.; Arvin, A.M. Fusarium brain abscess. Case report. J. Neurosurg. 1983, 58, 598–601. [Google Scholar] [CrossRef] [PubMed]
  50. Hiebert, R.M.; Welliver, R.C.; Yu, Z. Fusarium osteomyelitis in a patient with pearson syndrome: Case report and review of the literature. Open Forum Infect. Dis. 2016, 3, ofw183. [Google Scholar] [CrossRef] [PubMed]
  51. Taj-Aldeen, S.J.; Rammaert, B.; Gamaletsou, M.; Sipsas, N.V.; Zeller, V.; Roilides, E.; Kontoyiannis, D.P.; Miller, A.O.; Petraitis, V.; Walsh, T.J.; et al. Osteoarticular infections caused by non-Aspergillus filamentous fungi in adult and pediatric patients: A systematic review. Medicine 2015, 94, e2078. [Google Scholar] [CrossRef] [PubMed]
  52. Gupta, A.K.; Baran, R.; Summerbell, R.C. Fusarium infections of the skin. Curr. Opin. Infect. Dis. 2000, 13, 121–128. [Google Scholar] [CrossRef] [PubMed]
  53. Girmenia, C.; Arcese, W.; Micozzi, A.; Martino, P.; Bianco, P.; Morace, G. Onychomycosis as a possible origin of disseminated Fusarium solani infection in a patient with severe aplastic anemia. Clin. Infect. Dis. 1992, 14, 1167. [Google Scholar] [CrossRef] [PubMed]
  54. Arrese, J.E.; Pierard-Franchimont, C.; Pierard, G.E. Fatal hyalohyphomycosis following Fusarium onychomycosis in an immunocompromised patient. Am. J. Dermatopathol. 1996, 18, 196–198. [Google Scholar] [CrossRef] [PubMed]
  55. Bourgeois, G.P.; Cafardi, J.A.; Sellheyer, K.; Andea, A.A. Disseminated Fusarium infection originating from paronychia in a neutropenic patient: A case report and review of the literature. Cutis 2010, 85, 191–194. [Google Scholar] [PubMed]
  56. King, B.A.; Seropian, S.; Fox, L.P. Disseminated Fusarium infection with muscle involvement. J. Am. Acad. Dermatol. 2011, 65, 235–237. [Google Scholar] [CrossRef] [PubMed]
  57. Oechsler, R.A.; Yamanaka, T.M.; Bispo, P.J.; Sartori, J.; Yu, M.C.; Melo, A.S.; Miller, D.; Hofling-Lima, A.L. Fusarium keratitis in Brazil: Genotyping, in vitro susceptibilities, and clinical outcomes. Clin. Ophthalmol. 2013, 7, 1693–1701. [Google Scholar] [CrossRef] [PubMed]
  58. Wong, T.Y.; Fong, K.S.; Tan, D.T. Clinical and microbial spectrum of fungal keratitis in Singapore: A 5-year retrospective study. Int. Ophthalmol. 1997, 21, 127–130. [Google Scholar] [CrossRef] [PubMed]
  59. Bhartiya, P.; Daniell, M.; Constantinou, M.; Islam, F.M.; Taylor, H.R. Fungal keratitis in Melbourne. Clin. Exp. Ophthalmol. 2007, 35, 124–130. [Google Scholar] [CrossRef] [PubMed]
  60. Thomas, P.A. Current perspectives on ophthalmic mycoses. Clin. Microbiol. Rev. 2003, 16, 730–797. [Google Scholar] [CrossRef] [PubMed]
  61. Bharathi, M.J.; Ramakrishnan, R.; Meenakshi, R.; Padmavathy, S.; Shivakumar, C.; Srinivasan, M. Microbial keratitis in South India: Influence of risk factors, climate, and geographical variation. Ophthalmic Epidemiol. 2007, 14, 61–69. [Google Scholar] [CrossRef] [PubMed]
  62. Salah, H.; Al-Hatmi, A.M.; Theelen, B.; Abukamar, M.; Hashim, S.; van Diepeningen, A.D.; Lass-Florl, C.; Boekhout, T.; Almaslamani, M.; Taj-Aldeen, S.J. Phylogenetic diversity of human pathogenic Fusarium and emergence of uncommon virulent species. J. Infect. 2015, 71, 658–666. [Google Scholar] [CrossRef] [PubMed]
  63. Jensen, T.G.; Gahrn-Hansen, B.; Arendrup, M.; Bruun, B. Fusarium fungaemia in immunocompromised patients. Clin. Microbiol. Infect. 2004, 10, 499–501. [Google Scholar] [CrossRef] [PubMed]
  64. De Souza, M.; Matsuzawa, T.; Lyra, L.; Busso-Lopes, A.F.; Gonoi, T.; Schreiber, A.Z.; Kamei, K.; Moretti, M.L.; Trabasso, P. Fusarium napiforme systemic infection: Case report with molecular characterization and antifungal susceptibility tests. Springerplus 2014, 3, 492. [Google Scholar] [CrossRef] [PubMed]
  65. Kawakami, H.; Inuzuka, H.; Hori, N.; Takahashi, N.; Ishida, K.; Mochizuki, K.; Ohkusu, K.; Muraosa, Y.; Watanabe, A.; Kamei, K. Inhibitory effects of antimicrobial agents against Fusarium species. Med. Mycol. 2015, 53, 603–611. [Google Scholar] [CrossRef] [PubMed]
  66. Gupta, C.; Jongman, M.; Das, S.; Snehaa, K.; Bhattacharya, S.N.; Seyedmousavi, S.; van Diepeningen, A.D. Genotyping and in vitro antifungal susceptibility testing of Fusarium isolates from onychomycosis in India. Mycopathologia 2016, 181, 497–504. [Google Scholar] [CrossRef] [PubMed]
  67. Carneiro, H.A.; Coleman, J.J.; Restrepo, A.; Mylonakis, E. Fusarium infection in lung transplant patients: Report of 6 cases and review of the literature. Medicine 2011, 90, 69–80. [Google Scholar] [CrossRef] [PubMed]
  68. Espinel-Ingroff, A.; Colombo, A.L.; Cordoba, S.; Dufresne, P.J.; Fuller, J.; Ghannoum, M.; Gonzalez, G.M.; Guarro, J.; Kidd, S.E.; Meis, J.F.; et al. International evaluation of mic distributions and epidemiological cutoff value (ECV) definitions for Fusarium species identified by molecular methods for the CLSI broth microdilution method. Antimicrob. Agents Chemother. 2016, 60, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  69. Natesan, S.K.; Chandrasekar, P.H. Isavuconazole for the treatment of invasive aspergillosis and mucormycosis: Current evidence, safety, efficacy, and clinical recommendations. Infect. Drug Resist. 2016, 9, 291–300. [Google Scholar] [CrossRef] [PubMed]
  70. Donnelley, M.A.; Zhu, E.S.; Thompson, G.R., 3rd. Isavuconazole in the treatment of invasive aspergillosis and mucormycosis infections. Infect. Drug Resist. 2016, 9, 79–86. [Google Scholar] [PubMed]
  71. Moroti, R.V.; Gheorghita, V.; Al-Hatmi, A.M.; de Hoog, G.S.; Meis, J.F.; Netea, M.G. Fusarium ramigenum, a novel human opportunist in a patient with common variable immunodeficiency and cellular immune defects: Case report. BMC Infect. Dis. 2016, 16, 79. [Google Scholar] [CrossRef] [PubMed]
  72. Ricna, D.; Lengerova, M.; Palackova, M.; Hadrabova, M.; Kocmanova, I.; Weinbergerova, B.; Pavlovsky, Z.; Volfova, P.; Bouchnerova, J.; Mayer, J.; et al. Disseminated fusariosis by Fusarium proliferatum in a patient with aplastic anaemia receiving primary posaconazole prophylaxis—Case report and review of the literature. Mycoses 2016, 59, 48–55. [Google Scholar] [CrossRef] [PubMed]
  73. Al-Hatmi, A.M.; Bonifaz, A.; Tirado-Sanchez, A.; Meis, J.F.; de Hoog, G.S.; Ahmed, S.A. Fusarium species causing eumycetoma: Report of two cases and comprehensive review of the literature. Mycoses 2017, 60, 204–212. [Google Scholar] [CrossRef] [PubMed]
  74. Jiang, Y.; Al-Hatmi, A.M.; Xiang, Y.; Cao, Y.; van den Ende, A.H.; Curfs-Breuker, I.; Meis, J.F.; Lu, H.; de Hoog, G.S. The concept of ecthyma gangrenosum illustrated by a Fusarium oxysporum infection in an immunocompetent individual. Mycopathologia 2016, 181, 759–763. [Google Scholar] [CrossRef] [PubMed]
  75. Sara, S.; Sharpe, K.; Morris, S. Multidrug-resistant Fusarium keratitis: Diagnosis and treatment considerations. BMJ Case Rep. 2016, 2016, bcr2016215401. [Google Scholar] [CrossRef] [PubMed]
  76. Dabas, Y.; Bakhshi, S.; Xess, I. Fatal cases of bloodstream infection by Fusarium solani and review of published literature. Mycopathologia 2016, 181, 291–296. [Google Scholar] [CrossRef] [PubMed]
  77. Bansal, Y.; Chander, J.; Kaistha, N.; Singla, N.; Sood, S.; van Diepeningen, A.D. Fusarium sacchari, a cause of mycotic keratitis among sugarcane farmers—A series of four cases from North India. Mycoses 2016, 59, 705–709. [Google Scholar] [CrossRef] [PubMed]
  78. Al-Hatmi, A.M.; Bonifaz, A.; Calderon, L.; Curfs-Breuker, I.; Meis, J.F.; van Diepeningen, A.D.; de Hoog, G.S. Proximal subungual onychomycosis caused by Fusarium falciforme successfully cured with posaconazole. Br. J. Dermatol. 2015, 173, 253–255. [Google Scholar] [CrossRef] [PubMed]
  79. Dananche, C.; Cassier, P.; Sautour, M.; Gautheron, N.; Wegrzyn, J.; Perraud, M.; Bienvenu, A.L.; Nicolle, M.C.; Boibieux, A.; Vanhems, P. Fungaemia caused by Fusarium proliferatum in a patient without definite immunodeficiency. Mycopathologia 2015, 179, 135–140. [Google Scholar] [CrossRef] [PubMed]
  80. Da Silva-Rocha, W.P.; Zuza-Alves, D.L.; Melo, A.S.; Chaves, G.M. Fungal peritonitis due to Fusarium solani species complex sequential isolates identified with DNA sequencing in a kidney transplant recipient in Brazil. Mycopathologia 2015, 180, 397–401. [Google Scholar] [CrossRef] [PubMed]
  81. Al-Hatmi, A.M.; Bonifaz, A.; de Hoog, G.S.; Vazquez-Maya, L.; Garcia-Carmona, K.; Meis, J.F.; van Diepeningen, A.D. Keratitis by Fusarium temperatum, a novel opportunist. BMC Infect. Dis 2014, 14, 588. [Google Scholar] [CrossRef] [PubMed]
  82. Georgiadou, S.P.; Velegraki, A.; Arabatzis, M.; Neonakis, I.; Chatzipanagiotou, S.; Dalekos, G.N.; Petinaki, E. Cluster of Fusarium verticillioides bloodstream infections among immunocompetent patients in an internal medicine department after reconstruction works in Larissa, central Greece. J. Hosp. Infect. 2014, 86, 267–271. [Google Scholar] [CrossRef] [PubMed]
  83. Kang, Y.; Li, L.; Zhu, J.; Zhao, Y.; Zhang, Q. Identification of Fusarium from a patient with fungemia after multiple organ injury. Mycopathologia 2013, 176, 151–155. [Google Scholar] [CrossRef] [PubMed]
  84. Inano, S.; Kimura, M.; Iida, J.; Arima, N. Combination therapy of voriconazole and terbinafine for disseminated fusariosis: Case report and literature review. J. Infect. Chemother. 2013, 19, 1173–1180. [Google Scholar] [CrossRef] [PubMed]
  85. Campos-Macias, P.; Arenas-Guzman, R.; Hernandez-Hernandez, F. Fusarium subglutinans: A new eumycetoma agent. Med. Mycol. Case Rep. 2013, 2, 128–131. [Google Scholar] [CrossRef] [PubMed]
  86. Taylan Sekeroglu, H.; Erdem, E.; Yagmur, M.; Gumral, R.; Ersoz, R.; Ilkit, M.; Harbiyeli, I.I. Successful medical management of recalcitrant Fusarium solani keratitis: Molecular identification and susceptibility patterns. Mycopathologia 2012, 174, 233–237. [Google Scholar] [CrossRef] [PubMed]
  87. Edelstein, S.L.; Akduman, L.; Durham, B.H.; Fothergill, A.W.; Hsu, H.Y. Resistant Fusarium keratitis progressing to endophthalmitis. Eye Contact Lens 2012, 38, 331–335. [Google Scholar] [CrossRef] [PubMed]
  88. Cocchi, S.; Codeluppi, M.; Venturelli, C.; Bedini, A.; Grottola, A.; Gennari, W.; Cavrini, F.; Di Benedetto, F.; De Ruvo, N.; Rumpianesi, F.; et al. Fusarium verticillioides fungemia in a liver transplantation patient: Successful treatment with voriconazole. Diagn. Microbiol. Infect. Dis. 2011, 71, 438–441. [Google Scholar] [CrossRef] [PubMed]
  89. Liu, J.Y.; Chen, W.T.; Ko, B.S.; Yao, M.; Hsueh, P.R.; Hsiao, C.H.; Kuo, Y.M.; Chen, Y.C. Combination antifungal therapy for disseminated fusariosis in immunocompromised patients: A case report and literature review. Med. Mycol. 2011, 49, 872–878. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clinical and economic factors associated with MDR Fusarium infections.
Figure 1. Clinical and economic factors associated with MDR Fusarium infections.
Jof 03 00018 g001
Figure 2. Erythematous cutaneous lesions on the thigh of a 24-year-old T-cell lymphoma female with neutropenia (<100 cells/µL) who died from F. solani fungemia irrespective of antifungal treatment (Case #15 Table 1 and Table 2). Photography by Saad J. Taj-Aldeen (Hamad Medical Corporation, Qatar).
Figure 2. Erythematous cutaneous lesions on the thigh of a 24-year-old T-cell lymphoma female with neutropenia (<100 cells/µL) who died from F. solani fungemia irrespective of antifungal treatment (Case #15 Table 1 and Table 2). Photography by Saad J. Taj-Aldeen (Hamad Medical Corporation, Qatar).
Jof 03 00018 g002
Figure 3. Mann-Whitney U and Kruskal-Wallis tests. Box plot distribution of MIC values of amphotericin B (A) and voriconazole (B) for clinical (39 isolates) and CBS (Westerdijk Institute) reference Fusarium strains (12 isolates), showing significantly (p-value < 0.05) lower MIC values of the CBS reference strains. Outliers, values that do not fall in the inner fences; (°) cases with values between 1.5 and 3 times the interquartile (IQ) range, i.e., beyond the whiskers; (*) values more than three times the height of the boxes (extremes are cases with values more than 3 times the IQ range).
Figure 3. Mann-Whitney U and Kruskal-Wallis tests. Box plot distribution of MIC values of amphotericin B (A) and voriconazole (B) for clinical (39 isolates) and CBS (Westerdijk Institute) reference Fusarium strains (12 isolates), showing significantly (p-value < 0.05) lower MIC values of the CBS reference strains. Outliers, values that do not fall in the inner fences; (°) cases with values between 1.5 and 3 times the interquartile (IQ) range, i.e., beyond the whiskers; (*) values more than three times the height of the boxes (extremes are cases with values more than 3 times the IQ range).
Jof 03 00018 g003
Table 1. Demographic and clinical data obtained by review of the most recent (2011–2016) proven case series of fusariosis with MDR-resistant Fusarium species.
Table 1. Demographic and clinical data obtained by review of the most recent (2011–2016) proven case series of fusariosis with MDR-resistant Fusarium species.
Case No.Age/GenderOrganismUnderlying DiseaseNeutropeniaProphylaxis InfectionTreatment OutcomeRef. No.
132/MF. ramigenumCommon variable immunodeficiencyNoNoInvasive lung infection4 mg/kg IV q12h VRC for 6 months and continuous immunoglobulin substitution with 25 g/d, (5 d/month)Survival[71]
221/MF. petroliphilumAplastic anemiaYesPOC (200 mg three times/d)Fungemia/skin lesionsL-AmB lipid complex 4 mg/kg/d, then VRC 200 mg/d + granulocyte transfusion Died[72]
344/MF. solaniAMLYes
(<500/µL)
POC (600 mg/d)Fungemia/skin lesions, Lung infectionsL-AmB (5 mg/kg/day) + VRC (4 mg/kg/d; 6 mg/kg/first day loading dose). Died[9]
464/MF. keratoplasticumNoneNoNoMycetoma, right ankleITC 400 mg/day (14 months), then terbinafine 250 mg/d for 10 monthsSurvival[73]
537/MF. pseudensiformeNone NoNoMycetoma, left footOral ITC (400 mg/d)Improved, lost follow up after six months
617/MF. oxysporumNoneNoNoEcthyma gangrenosumVRC (400 mg/d orally + surgical debridementImproved/lost follow up[74]
746/MF. solaniNoneNoNokeratitisTopical 1% VRC + 5% natamycin, +0.01 mg AmB + systemic VRC [75]
865/MF. solaniT cell large anaplastic lymphoma erythroderma without systemic involvementNoFLC (200 mg IV BD)FungemiaAmB (20 mg IV OD)Died[76]
960/FF. sacchariNone/trauma with sugar cane leafNoNokeratitisOral ITC/topical VRC/KeratoplastyResponded to the treatment[77]
1045/MF. sacchariNone/trauma with sugar cane leafNoNokeratitisOral ITC/topical VRC +AmB/KeratoplastyResponded to the treatment
1140/MF. sacchariNone/trauma with sugar cane leafNoNokeratitisTopical VRC/KeratoplastyResponded to the treatment
1260/FF. sacchariNone/trauma with vegetative matterNoNokeratitisOral ITC/topical VRC + natamycinResponded to the treatment
1380/FF. petroliphilumAutoimmune disease on corticosteroidsYesNoFungemiaFLC (empiric)Died[29]
1437/MF. petroliphilumAMLYesNoFungemiaAmB + GM-CSFRecovered
1524/FF. solani sensu latoT-cell lymphomaYes?Fungemia/skin lesions AmB + VRC + GM-CSFDied
1664/MF. falciformeAIDSNoNoToe nail OnychomycosisITC 200 mg/d, then terbinafine 250 mg/d (for 75 d), Changed to POS 800 mg/d for one week/month (continued for 4 months)Survival [78]
1778/FF. proliferatumNone/right total hip arthroplasty replacementNoFLC (400 mg twice a day at D1 then once a day from D2) FungemiaOral VRC (400 mg twice/d, then (200 mg twice/d) for 72 dRecovered[79]
1838/FF. solaniKidney transplant, DMNDNoInvasive/peritoneal fluidAmB (50 mg/d) Alive[80]
1965/MF. andiyaziAMLYesOral POC (3 × 200 mg/d)Disseminated lung infectionAMB; (3 mg kg/d)Died[35]
2048/FF. petroliphilumALLYesNoFungemia/skin lesionsAMB; (3 mg/kg/day)Died[40]
21NDF. temperatumNone/trauma with maize plantNoNoKeratitisTopical natamycin 5% + ITC 200 mg/d Improved, No follow up[81]
2274/MF. verticillioidesDiabetes mellitusNoNoFungemiaNDDied[82]
2360/FF. napiformeStage III multiple myelomaYesAmB deoxycholateFungemia/skin lesionsAMB deoxycholate/for one monthDied[64]
2467/MF. solaniAcute biphenotyic pneumoniaYes Fungemia/skin lesions/pancytopeniaLAmB (3 mg/kg/day)Died[36]
2521/MF. solaniMultiple organ injury?FLC (200 mg/d)FungemiaNoneDied[83]
2665/MF. solaniAMLYesITCDisseminated/endocarditis/skin lesions 5 mg/kg/d + VRC (4 mg/kg for 25 d, then AmB + Terbinafine (500 mg/d) + GM-CFNo relapse on maintenance therapy [84]
2729/MF. subglutinansNoneNoNoMycetoma, osteomyelitisITC 200 mg twice daily for 4 monthsImproved[85]
2814/MF. solaniOcular traumaNoNokeratitisVRC (10 mg/mL) every hour + topical natamycin (5%) five times daily, + with 500 mg oral ketoconazole twice a day at 12-h intervals (1 g/day). Improved[86]
2952/MF. solaniCorneal injuryNoNoKeratitis and endophthalmitis Topical 5% natamycin + 0.15% AmB + oral FLC 200 mg/d, And vitrectomy, AmB injection, Then topical 1% VRC + 200 mg twice daily + POS 200 mg four times dailyInfection persist[87]
3036/FF. prolifertarumLung transplanNoITCLung infectionL AmB, VRCDied[67]
3130/FF. verticillioidesLiver transplant?FLCFungemia/skin lesionsVRC 6 mg/kg (360 mg) bid, followed by 4 mg/kg (240 mg) bid for 20 days , then oral(200 mg bid) for a further 5 weeks alive[88]
3227/MF. solaniCutaneous T cell lymphoma with leukemia YesFLC 400 mg/d (loading dose 800 mg)Fungemia/skin lesions/Lung infectionAmB deoxycholate + VRC 4.5 mg/kg every 12 h. Discharged on oral VRC + Granulocyte transfusionAlive after 6 months[89]
AmB: Amphotericin B; VRC: voriconazole; POS: posaconazole; FLC: fluconazole; AML: acute myeloid leukemia; ALL: acute lymphoblastic leukemia; ?: Data not available.
Table 2. Reported MIC values of Fusarium species of 9 antifungal agents reported for clinical cases presented in Table 1.
Table 2. Reported MIC values of Fusarium species of 9 antifungal agents reported for clinical cases presented in Table 1.
Case No.Fusarium spp.Fusarium Species ComplexMIC/MEC (µg/mL)Ref No.
AmBFLCITCVRCPOSISVCASMCAANI
1F. ramigenumFFSC1 >16214 >8>8[71]
2F. petroliphilumFSSC4 >324>32 [72]
3F. solaniFSSC4>256>1616>32 >32>32>32[9]
4F. keratoplasticumFSSC1>6444>168 >16>16[73]
5F. pseudensiformeFSSC0.5>64>16848 >16>16
6F. oxysporumFOSC1>64>168>168 >16>16[74]
7F. solaniFSSC4 88 [75]
8F. solaniFSSC2–4>64>80.25–0.52–8 >16>16 [76]
9F. sacchariFFSC0.5>64≥16≥16 >64 ≥16[77]
10F. sacchariFFSC0.5>64≥164 >64 ≥16
11F. sacchariFFSC1>6440.0625 16 ≥16
12F. sacchariFFSC >64>164 >64 ≥16
13F. petroliphilumFSSC2>128>4>16>16 84>16[29]
14F. petroliphilumFSSC1>128>4>16>16 8>16>16
15F. solani sensu latoFSSC0.5>128>4>16>16 84>16
16F. falciformeFSSC0.5>64>1680.5 >8 [78]
17F. petroliphilumFSSC6 120.750.25 >32 [79]
18F. solaniFSSC2≥64≥16 ≥8 [80]
19F. andiyaziFFSC81682148>8 [35]
20F. petroliphilumFSSC1>64>168>16>16 >8>8[40]
21F. temperatumFFSC0.5>64>1610.254 0.0314[81]
22F. verticillioidesFFSC>32 >32132 >16>16[82]
23F. napiformeFFSC2–41–2>84 >16 [64]
24F. solaniFSSC8 >80.12 [36]
25F. solaniFSSC1.5>256>322 >32 [83]
26F. solaniFSSC1>4>8>8 >16 [84]
27F. subglutinansFFSC ≥64≥16 ≥16 ≥16 ≥32[85]
28F. solaniFSSC0.5>64>168>16 >16 >16[86]
29F. solaniFSSC4>64>16>168 [87]
30F. prolifertarumFFSC4>128>128 128 [67]
31F. verticillioidesFFSC8-16>256324 ≥32 [88]
32F. solaniFSSC1 4 [89]
Range (total) 0.5–1616– > 2564– > 1280.0625– > 160.5– > 324– > 168– > 1280.031– > 324– > 32
Abbreviations: FSSC: F. solani species complex; FFSC: F. fujikuroi species complex; FOSC: F. oxysporum species complex. AmB: Amphotericin B; FLC: fluconazole; ITC: itraconazole; VRC: voriconazole; POS: posaconazole; ISV: isavuconazole; CAS: Caspofungin; MCA: megafungin; ANI: anidulafungin.

Share and Cite

MDPI and ACS Style

Taj-Aldeen, S.J. Reduced Multidrug Susceptibility Profile Is a Common Feature of Opportunistic Fusarium Species: Fusarium Multi-Drug Resistant Pattern. J. Fungi 2017, 3, 18. https://doi.org/10.3390/jof3020018

AMA Style

Taj-Aldeen SJ. Reduced Multidrug Susceptibility Profile Is a Common Feature of Opportunistic Fusarium Species: Fusarium Multi-Drug Resistant Pattern. Journal of Fungi. 2017; 3(2):18. https://doi.org/10.3390/jof3020018

Chicago/Turabian Style

Taj-Aldeen, Saad J. 2017. "Reduced Multidrug Susceptibility Profile Is a Common Feature of Opportunistic Fusarium Species: Fusarium Multi-Drug Resistant Pattern" Journal of Fungi 3, no. 2: 18. https://doi.org/10.3390/jof3020018

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

Article Metrics

Back to TopTop