ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Mycology, pathogenesis, and epidemiology of Fusarium infection

Mycology, pathogenesis, and epidemiology of Fusarium infection
Literature review current through: Jan 2024.
This topic last updated: Jun 27, 2023.

INTRODUCTION — Fusarium species cause a broad spectrum of infections in humans including superficial infections, such as keratitis and onychomycosis, as well as locally invasive and disseminated infections; invasive and disseminated infections occur almost exclusively in severely immunocompromised patients [1]. Fusarium species may also cause allergic diseases, such as sinusitis in immunocompetent individuals [2], and mycotoxicosis following ingestion of food contaminated by toxin-producing Fusarium species [3]. Fusarium species are also important plant pathogens that cause various diseases on cereal grains [3] and occasionally cause infection in animals [4].

The mycology, pathogenesis, and epidemiology of fusariosis will be reviewed here. The clinical manifestations, diagnosis, treatment, and prevention of fusariosis are discussed separately. (See "Clinical manifestations and diagnosis of Fusarium infection" and "Treatment and prevention of Fusarium infection".)

MYCOLOGY — Fusarium species are widely distributed in soil, subterranean and aerial plant parts, plant debris, and other organic matter [3]. They are also present in water worldwide [5].

Growth in vitro — Fusarium species grow rapidly on many media that do not contain cycloheximide, which inhibits its growth. On potato dextrose agar, Fusarium species produce white-, lavender-, pink-, salmon-, or gray-colored colonies with velvety or cottony surfaces [6].

Microscopic appearance — Microscopically, the hyphae of Fusarium in tissue resemble those of Aspergillus species, with septate hyaline hyphae 3 to 8 microns in diameter that typically branch at acute angles (picture 1 and picture 2). Adventitious sporulation, which is the ability to sporulate in tissue and blood, may be present [7]; the identification of hyphal and yeast-like structures in the same specimen is highly suggestive of fusariosis in high-risk patients.

In cultures, the production of both fusoid macroconidia (hyaline, multicellular, banana-like clusters with foot cells at the base) (picture 3 and picture 4) and microconidia (hyaline, unicellular, ovoid to cylindrical) are characteristic of the genus Fusarium.

Species identification — The characteristic appearance of Fusarium macroconidia can be used for identification (picture 3 and picture 4), but species identification is difficult and often requires molecular methods [8-10]. Mass spectrometry using matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) has been shown to correctly identify clinical isolates to the species level [11,12].

Species prevalence — More than 300 species of Fusarium have been identified, but only a few cause infections in humans [13,14]. Fusarium species are grouped into >20 phylogenetic species complexes, 7 of which have been reported to cause significant human disease [15]: Fusarium solani species complex (FSSC), Fusarium oxysporum species complex (FOSC), Fusarium fujikuroi species complex (FFSC), Fusarium incarnatum-equiseti species complex, Fusarium chlamidosporum species complex, Fusarium dimerum species complex, and Fusarium sporotrichoides species complex, with various species within each complex. Approximately 50 percent of cases of invasive fusariosis are caused by FSSC, followed by FOSC and FFSC [1,16]. The distribution of species causing infection varies geographically. For example, in Brazil FSSC is the predominant species complex [17], whereas in various European countries FFSC is the most frequent (especially Fusarium verticillioides and Fusarium proliferatum) [18]. Fusarial keratitis is most commonly caused by FSSC [19], while fusarial onychomycosis is most commonly caused by F. oxysporum [20-22].

PATHOGENESIS — Fusarium spp cause invasive disease by angioinvasion and direct tissue destruction [3,23]. Fusarium species possess several virulence factors including mycotoxins, which suppress humoral and cellular immunity and can also cause allergic reactions [3]. Fusarium spp also have the ability to adhere to prosthetic material [3] and to produce proteases and collagenases [24]. F. solani is thought to be the most virulent Fusarium species [25]. Some Fusarium species produce mycotoxins that are destructive to crops and can cause human disease when ingested with heavily contaminated grains [21].

Immune response — Innate immunity plays a major role in the defense against Fusarium species [26]. Macrophages and neutrophils damage fusarial hyphae, and their effect is primed by interferon-gamma, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor [27], and interleukin (IL)-15 [28]. The effect of IL-15 is mediated by the release of IL-8 and by direct stimulation of hyphal damage. The role of toll-like receptors in the innate immune recognition of fungi is increasingly being recognized [29], and, although little is known about fusariosis and toll-like receptors, this system is likely to be important in invasive fusariosis.

EPIDEMIOLOGY — Fusarium species cause a broad spectrum of infections in humans, including superficial, locally invasive, and disseminated infections. The clinical form of infection depends upon the immune status of the host and the portal of entry of the pathogen [1].

Immunocompetent hosts — Keratitis and onychomycosis are the most common infections in immunocompetent patients. Other sites of infection have also been reported (table 1) [30-42].

Keratitis — Fusarium spp are a common cause of fungal keratitis, which may develop following the traumatic introduction of Fusarium-contaminated soil or plant material or due to poor hygiene practices of soft contact lens wearers, particularly in patients using glucocorticoid eye drops. A large international outbreak of fusarial keratitis between 2004 and 2006 was linked to the use of a specific contact lens solution (ReNu with MoistureLoc) [43-47]. The solution was not intrinsically contaminated. One study has suggested that in the setting of storage at high temperatures, the antimicrobial agent in the solution, alexidine dihydrochloride, permeates the walls of the plastic bottle, resulting in decreased concentrations in the solution [48]. The fungicidal action of the solution was diminished, allowing Fusarium that had been introduced by the wearer while caring for his or her lenses to grow and contaminate the contact lens and the lens case. The solution was recalled in the United States in 2006, and rates of contact lens-associated Fusarium keratitis have declined since then [49].

The incidence of fungal keratitis caused by Fusarium species is highly variable and depends largely upon the geographic location. For example, in several studies performed at different times over a span of 10 years, the incidence has been noted to range from 8 percent in India to 25 percent in Pennsylvania, 63 percent in Florida, and 75 percent in Tanzania [19,50,51]. These variable rates are probably related to climate characteristics, with tropical and subtropical areas exhibiting the highest rates [19].

Onychomycosis — Fusarium species have been reported as agents of onychomycosis with increasing frequency among immunocompetent individuals [52,53] with variable rates reported from different regions. As an example, in a study from central Italy, fewer than 2 percent of cases of onychomycosis were caused by Fusarium spp [54], compared with more than 8 percent in a study of onychomycosis cases from the northeast region of Brazil [20].

Meningitis — Although rare, Fusarium meningitis has been reported from health care-associated sources. Two separate outbreaks have occurred, one in late 2022 and another between January 1 and May 13, 2023, among individuals who underwent epidural anesthesia for plastic surgery procedures performed at specific clinics in Durango and Matamoros, Mexico [55,56]. (See "Aseptic meningitis in adults", section on 'Fusarium outbreaks'.)

Immunocompromised patients

Portals of entry — Invasive fusariosis has been reported worldwide and may be acquired both in the community and in the hospital setting. The principal modes of infection are airborne or via direct inoculation at various sites (skin and others) of Fusarium-contaminated material, including water (table 2). Airborne infection typically results from the inhalation of fusarial conidia from the air [57], but in some cases have been associated with aerosolization of conidia from a water source [58]. In addition, pre-existing skin breakdown or onychomycosis may facilitate direct introduction of Fusarium spp. An increase in cases of invasive fusariosis with a cutaneous portal of entry has been reported in Brazil [59]. Onychomycosis and intertrigo were the predominant primary lesions. A subsequent study from the same group showed that in hematologic patients at high risk of developing invasive fusariosis, the presence of such lesions growing Fusarium spp at the time of admission increased the risk for the subsequent occurrence of invasive disease [60].

Central venous catheter-related Fusarium infection has also been reported; in one report, 7 cases of genetically related F. oxysporum bloodstream infection occurred in children with cancer who had central venous catheters [61]. Although an environmental source was not identified, the outbreak was presumed to be due to central line manipulation in a specific room as the outbreak ended after implementation of a multidisciplinary central line insertion care bundle.

Risk factors — Patients with compromised immune function are at increased risk for invasive fusariosis, particularly in the setting of prolonged and profound neutropenia and/or severe T cell immunodeficiency [62]. Unlike infection in normal hosts, fusariosis in the immunocompromised population is typically invasive and disseminated [30].

The overall incidence of fusariosis among hematopoietic cell transplant (HCT) recipients and patients with acute myeloid leukemia (AML) is low, usually less than 1 percent, as reported in studies conducted in the United States and Italy [63-65]. By contrast, in a multicenter study conducted between 2007 and 2009 in Brazil, invasive fusariosis was the second most common cause of invasive fungal disease among patients with AML (3.8 percent) and the most common cause among allogeneic HCT recipients (5.2 percent) [66]. In a more recent study from Brazil, invasive fusariosis was the third most-frequent invasive fungal disease among patients with hematologic cancers (3 cases among 192 patients [1.6 percent]) [67]. These geographic differences in the incidence of invasive fusariosis must be taken into account in the management of high-risk patients.

Among patients with hematologic malignancy, the infection predominates during periods of neutropenia, typically among patients with leukemia receiving induction chemotherapy [68]. Smoking also appears to be a risk factor in patients with AML or myelodysplastic syndrome [69]. Ibrutinib, a Bruton tyrosine kinase inhibitor, has been associated with invasive fungal infections, including invasive fusariosis [70]. (See "Risk of infections in patients with chronic lymphocytic leukemia", section on 'Bruton tyrosine kinase inhibitors'.)

Invasive fusariosis also occurs with an increased frequency among allogeneic HCT recipients in whom a trimodal distribution of infection has been identified [71]. The first peak occurs during the first 30 days following transplantation and is associated with neutropenia. Severe T cell immunodeficiency (but not neutropenia) is the major risk factor for fusariosis that develops after engraftment and which occurs at a median of 70 days after HCT, commonly in the setting of acute graft-versus-host disease (GVHD) and therapy with glucocorticoids. The third peak is observed >1 year after HCT during treatment for extensive chronic GVHD. In one study, risk factors for invasive fusariosis during the early phase following allogeneic HCT included receipt of antithymocyte globulin, hyperglycemia, and AML [69]. During the late phase following allogeneic HCT, risk factors included nonmyeloablative conditioning regimen, grade III/IV GVHD, and previous invasive mold disease.

The prognosis of fusariosis is directly related to the patient's immune status, with high death rates in patients with persistent immunodeficiencies. In a series of 84 patients with fusariosis and an underlying hematologic malignancy, persistent neutropenia and recent glucocorticoid therapy were the only independent factors associated with poor outcome [68]. (See "Treatment and prevention of Fusarium infection", section on 'Prognosis'.)

Locally invasive Fusarium infections occur occasionally among solid organ transplant recipients, typically during the late post-transplant period; disseminated infections have been reported rarely in such patients [72]. Cases of chronic cutaneous fusariosis have also been reported in patients with STAT3 Hyper-IgE Syndrome [73].

Invasive fungal disease has also been reported in previously immunocompetent individuals who developed severe forms of COVID-19. While most infections were caused by aspergillosis, cases of invasive fusariosis were also reported [74]. The use of glucocorticosteroids for the treatment of severe COVID-19 pneumonia is thought to contribute to the development of these fungal infections [74].

The clinical manifestations of fusariosis are discussed separately. (See "Clinical manifestations and diagnosis of Fusarium infection".)

SUMMARY

Spectrum of diseaseFusarium species are important plant pathogens that cause various diseases on cereal grains and occasionally cause infection in animals. In humans, Fusarium species cause a broad spectrum of infections including superficial infections, such as keratitis and onychomycosis, as well as locally invasive and disseminated infections; invasive and disseminated infections occur almost exclusively in severely immunocompromised patients. (See 'Introduction' above and "Clinical manifestations and diagnosis of Fusarium infection".)

ReservoirFusarium species are widely distributed in soil, subterranean and aerial plant parts, plant debris, and other organic matter. They are also present in water. (See 'Mycology' above.)

Microbiology – Microscopically, the hyphae of Fusarium in tissue resemble those of Aspergillus species, with septate hyaline hyphae 3 to 8 microns in diameter that typically branch at acute angles (picture 1). The production of both fusoid macroconidia (hyaline, multicellular, banana-like clusters with foot cells at the base) (picture 3 and picture 4), and microconidia (hyaline, unicellular, ovoid to cylindrical) are characteristic of the genus Fusarium. (See 'Microscopic appearance' above.)

The characteristic appearance of Fusarium macroconidia is used for identification (picture 3 and picture 4), but species identification is difficult and often requires molecular methods or matrix-assisted laser desorption/ionization time of flight (MALDI-TOF). (See 'Species identification' above.)

Taxonomy – More than 300 species of Fusarium have been identified, but only a few cause infections in humans. Fusarium species are grouped into various phylogenetic species complexes (more than 20), 7 of which have been reported to cause significant disease in humans: Fusarium solani species complex (50 percent of infections), Fusarium oxysporum species complex, Fusarium fujikuroi species complex, Fusarium incarnatum-equiseti species complex, Fusarium chlamidosporum species complex, Fusarium dimerum species complex, and Fusarium sporotrichoides species complex. (See 'Species prevalence' above.)

PathophysiologyFusarium species cause invasive disease by angioinvasion and direct tissue destruction. Innate immunity plays a major role in the defense against various molds, including Fusarium species. Macrophages and neutrophils damage fusarial hyphae, and their effect is primed by interferon-gamma, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, and interleukin-15. (See 'Pathogenesis' above.)

Risk factorsFusarium species cause a broad spectrum of infections in humans, including superficial, locally invasive, and disseminated infections. The clinical form of infection depends upon the immune status of the host and the portal of entry of the pathogen (table 1). (See 'Epidemiology' above.)

Patients with compromised immune function are at high risk for invasive fusariosis, particularly in the setting of prolonged and profound neutropenia and/or severe T cell immunodeficiency, such as in patients with hematologic malignancies receiving induction chemotherapy or allogeneic hematopoietic cell transplantation. (See 'Risk factors' above.)

  1. Nucci M, Anaissie E. Fusarium infections in immunocompromised patients. Clin Microbiol Rev 2007; 20:695.
  2. Wickern GM. Fusarium allergic fungal sinusitis. J Allergy Clin Immunol 1993; 92:624.
  3. Nelson PE, Dignani MC, Anaissie EJ. Taxonomy, biology, and clinical aspects of Fusarium species. Clin Microbiol Rev 1994; 7:479.
  4. Evans J, Levesque D, de Lahunta A, Jensen HE. Intracranial fusariosis: a novel cause of fungal meningoencephalitis in a dog. Vet Pathol 2004; 41:510.
  5. Elvers KT, Leeming K, Moore CP, Lappin-Scott HM. Bacterial-fungal biofilms in flowing water photo-processing tanks. J Appl Microbiol 1998; 84:607.
  6. Silveira FP, Queiroz-Telles F, Nucci M. Invasive mold infections. In: Diagnosis of fungal infections, Maertens JA, Marr KA (Eds), Informa Healthcare, 2007. p.171.
  7. Liu K, Howell DN, Perfect JR, Schell WA. Morphologic criteria for the preliminary identification of Fusarium, Paecilomyces, and Acremonium species by histopathology. Am J Clin Pathol 1998; 109:45.
  8. Healy M, Reece K, Walton D, et al. Use of the Diversi Lab System for species and strain differentiation of Fusarium species isolates. J Clin Microbiol 2005; 43:5278.
  9. Fleming RV, Anaissie EJ. Hyalohyphomycosis - Infection due to hyaline moulds. In: Diagnosis and Treatment of Human Mycoses, Hospenthal DR, Rinaldi MG (Eds), Humana Press, Totowa 2008. p.201.
  10. O'Donnell K, Sutton DA, Rinaldi MG, et al. Internet-accessible DNA sequence database for identifying fusaria from human and animal infections. J Clin Microbiol 2010; 48:3708.
  11. de Almeida JN Júnior, Sztajnbok J, da Silva AR Junior, et al. Rapid identification of moulds and arthroconidial yeasts from positive blood cultures by MALDI-TOF mass spectrometry. Med Mycol 2016; 54:885.
  12. Triest D, Stubbe D, De Cremer K, et al. Use of matrix-assisted laser desorption ionization-time of flight mass spectrometry for identification of molds of the Fusarium genus. J Clin Microbiol 2015; 53:465.
  13. Alastruey-Izquierdo A, Cuenca-Estrella M, Monzón A, et al. Antifungal susceptibility profile of clinical Fusarium spp. isolates identified by molecular methods. J Antimicrob Chemother 2008; 61:805.
  14. Al-Hatmi AM, Hagen F, Menken SB, et al. Global molecular epidemiology and genetic diversity of Fusarium, a significant emerging group of human opportunists from 1958 to 2015. Emerg Microbes Infect 2016; 5:e124.
  15. van Diepeningen AD, Al-Hatmi AMS, Brankovics B, de Hoog GS. Taxonomy and clinical spectra of Fusarium species: Where do we stand in 2014? Curr Clin Microbiol Rep 2014; 1:10.
  16. Balajee SA, Borman AM, Brandt ME, et al. Sequence-based identification of Aspergillus, fusarium, and mucorales species in the clinical mycology laboratory: where are we and where should we go from here? J Clin Microbiol 2009; 47:877.
  17. Herkert PF, Al-Hatmi AMS, de Oliveira Salvador GL, et al. Molecular Characterization and Antifungal Susceptibility of Clinical Fusarium Species From Brazil. Front Microbiol 2019; 10:737.
  18. Tortorano AM, Prigitano A, Esposto MC, 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.
  19. Dóczi I, Gyetvai T, Kredics L, Nagy E. Involvement of Fusarium spp. in fungal keratitis. Clin Microbiol Infect 2004; 10:773.
  20. Brilhante RS, Cordeiro RA, Medrano DJ, et al. Onychomycosis in Ceará (Northeast Brazil): epidemiological and laboratory aspects. Mem Inst Oswaldo Cruz 2005; 100:131.
  21. Godoy P, Nunes E, Silva V, et al. Onychomycosis caused by Fusarium solani and Fusarium oxysporum in São Paulo, Brazil. Mycopathologia 2004; 157:287.
  22. Ninet B, Jan I, Bontems O, et al. Molecular identification of Fusarium species in onychomycoses. Dermatology 2005; 210:21.
  23. Torres HA, Kontoyiannis DP. Hyalohyphomycoses (hyaline moulds). In: Essentials of Clinical Mycology, 2nd ed, Kauffman C, Pappas PG, Sobel JD (Eds), Springer, New York 2011. p.281.
  24. Krátká J, Kováciková E. The effect of temperature and age of strains of Fusarium oxysporum on its enzymatic activity. Zentralbl Bakteriol Naturwiss 1979; 134:154.
  25. Mayayo E, Pujol I, Guarro J. Experimental pathogenicity of four opportunist Fusarium species in a murine model. J Med Microbiol 1999; 48:363.
  26. Shoham S, Levitz SM. The immune response to fungal infections. Br J Haematol 2005; 129:569.
  27. Gaviria JM, van Burik JA, Dale DC, et al. Comparison of interferon-gamma, granulocyte colony-stimulating factor, and granulocyte-macrophage colony-stimulating factor for priming leukocyte-mediated hyphal damage of opportunistic fungal pathogens. J Infect Dis 1999; 179:1038.
  28. Winn RM, Gil-Lamaignere C, Roilides E, et al. Effects of interleukin-15 on antifungal responses of human polymorphonuclear leukocytes against Fusarium spp. and Scedosporium spp. Cytokine 2005; 31:1.
  29. Romani L. Immunity to fungal infections. Nat Rev Immunol 2004; 4:1.
  30. Nucci M, Anaissie E. Cutaneous infection by Fusarium species in healthy and immunocompromised hosts: implications for diagnosis and management. Clin Infect Dis 2002; 35:909.
  31. Rippon JW, Larson RA, Rosenthal DM, Clayman J. Disseminated cutaneous and peritoneal hyalohyphomycosis caused by Fusarium species: three cases and review of the literature. Mycopathologia 1988; 101:105.
  32. Kerr CM, Perfect JR, Craven PC, et al. Fungal peritonitis in patients on continuous ambulatory peritoneal dialysis. Ann Intern Med 1983; 99:334.
  33. Flynn JT, Meislich D, Kaiser BA, et al. Fusarium peritonitis in a child on peritoneal dialysis: case report and review of the literature. Perit Dial Int 1996; 16:52.
  34. Kurien M, Anandi V, Raman R, Brahmadathan KN. Maxillary sinus fusariosis in immunocompetent hosts. J Laryngol Otol 1992; 106:733.
  35. Madhavan M, Ratnakar C, Veliath AJ, et al. Primary disseminated fusarial infection. Postgrad Med J 1992; 68:143.
  36. Sander A, Beyer U, Amberg R. Systemic Fusarium oxysporum infection in an immunocompetent patient with an adult respiratory distress syndrome (ARDS) and extracorporal membrane oxygenation (ECMO). Mycoses 1998; 41:109.
  37. Murray CK, Beckius ML, McAllister K. Fusarium proliferatum superficial suppurative thrombophlebitis. Mil Med 2003; 168:426.
  38. Sturm AW, Grave W, Kwee WS. Disseminated Fusarium oxysporum infection in patient with heatstroke. Lancet 1989; 1:968.
  39. Pflugfelder SC, Flynn HW Jr, Zwickey TA, et al. Exogenous fungal endophthalmitis. Ophthalmology 1988; 95:19.
  40. Gabriele P, Hutchins RK. Fusarium endophthalmitis in an intravenous drug abuser. Am J Ophthalmol 1996; 122:119.
  41. Jakle C, Leek JC, Olson DA, Robbins DL. Septic arthritis due to Fusarium solani. J Rheumatol 1983; 10:151.
  42. Bourguignon RL, Walsh AF, Flynn JC, et al. Fusarium species osteomyelitis. Case report. J Bone Joint Surg Am 1976; 58:722.
  43. Chang DC, Grant GB, O'Donnell K, et al. Multistate outbreak of Fusarium keratitis associated with use of a contact lens solution. JAMA 2006; 296:953.
  44. Khor WB, Aung T, Saw SM, et al. An outbreak of Fusarium keratitis associated with contact lens wear in Singapore. JAMA 2006; 295:2867.
  45. Patel A, Hammersmith K. Contact lens-related microbial keratitis: recent outbreaks. Curr Opin Ophthalmol 2008; 19:302.
  46. Saw SM, Ooi PL, Tan DT, et al. Risk factors for contact lens-related fusarium keratitis: a case-control study in Singapore. Arch Ophthalmol 2007; 125:611.
  47. Bullock JD, Elder BL, Khamis HJ, Warwar RE. Effects of time, temperature, and storage container on the growth of Fusarium species: implications for the worldwide Fusarium keratitis epidemic of 2004-2006. Arch Ophthalmol 2011; 129:133.
  48. Bullock JD, Elder BL, Warwar RE, et al. Mechanism of drug failure in fusarium keratitis, 2004-2006. N Engl J Med 2014; 370:88.
  49. Yildiz EH, Abdalla YF, Elsahn AF, et al. Update on fungal keratitis from 1999 to 2008. Cornea 2010; 29:1406.
  50. Tanure MA, Cohen EJ, Sudesh S, et al. Spectrum of fungal keratitis at Wills Eye Hospital, Philadelphia, Pennsylvania. Cornea 2000; 19:307.
  51. Rosa RH Jr, Miller D, Alfonso EC. The changing spectrum of fungal keratitis in south Florida. Ophthalmology 1994; 101:1005.
  52. Guilhermetti E, Takahachi G, Shinobu CS, Svidzinski TI. Fusarium spp. as agents of onychomycosis in immunocompetent hosts. Int J Dermatol 2007; 46:822.
  53. Hilmioğlu-Polat S, Metin DY, Inci R, et al. Non-dermatophytic molds as agents of onychomycosis in Izmir, Turkey - a prospective study. Mycopathologia 2005; 160:125.
  54. Romano C, Papini M, Ghilardi A, Gianni C. Onychomycosis in children: a survey of 46 cases. Mycoses 2005; 48:430.
  55. Centers for Disease Control and Prevention. Fungal meningitis outbreak associated with procedures performed under epidural anesthesia in Matamoros, Mexico. https://www.cdc.gov/hai/outbreaks/meningitis-epidural-anesthesia.html#:~:text=Early%20testing%20and%20treatment%2C%20especially,mild%20or%20absent%20at%20first. (Accessed on June 14, 2023).
  56. Interim recommendations for diagnosis and management of fungal meningitis associated with epidural anesthesia administered in Matamoros, Mexico. https://funguseducationhub.org/wp-content/uploads/2023/06/interim-recommendations-Matamoros-FM-outbreak-6_09_23.pdf (Accessed on June 14, 2023).
  57. Moretti ML, Busso-Lopes AF, Tararam CA, et al. Airborne transmission of invasive fusariosis in patients with hematologic malignancies. PLoS One 2018; 13:e0196426.
  58. Anaissie EJ, Kuchar RT, Rex JH, 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.
  59. Nucci M, Varon AG, Garnica M, et al. Increased incidence of invasive fusariosis with cutaneous portal of entry, Brazil. Emerg Infect Dis 2013; 19:1567.
  60. Varon AG, Nouer SA, Barreiros G, et al. Superficial skin lesions positive for Fusarium are associated with subsequent development of invasive fusariosis. J Infect 2014; 68:85.
  61. Carlesse F, Amaral AC, Gonçalves SS, et al. Outbreak of Fusarium oxysporum infections in children with cancer: an experience with 7 episodes of catheter-related fungemia. Antimicrob Resist Infect Control 2017; 6:93.
  62. Boutati EI, Anaissie EJ. Fusarium, a significant emerging pathogen in patients with hematologic malignancy: ten years' experience at a cancer center and implications for management. Blood 1997; 90:999.
  63. Kontoyiannis DP, Marr KA, Park BJ, et al. Prospective surveillance for invasive fungal infections in hematopoietic stem cell transplant recipients, 2001-2006: overview of the Transplant-Associated Infection Surveillance Network (TRANSNET) Database. Clin Infect Dis 2010; 50:1091.
  64. Pagano L, Caira M, Nosari A, et al. Fungal infections in recipients of hematopoietic stem cell transplants: results of the SEIFEM B-2004 study--Sorveglianza Epidemiologica Infezioni Fungine Nelle Emopatie Maligne. Clin Infect Dis 2007; 45:1161.
  65. Pagano L, Caira M, Candoni A, et al. The epidemiology of fungal infections in patients with hematologic malignancies: the SEIFEM-2004 study. Haematologica 2006; 91:1068.
  66. Nucci M, Garnica M, Gloria AB, et al. Invasive fungal diseases in haematopoietic cell transplant recipients and in patients with acute myeloid leukaemia or myelodysplasia in Brazil. Clin Microbiol Infect 2013; 19:745.
  67. Souza L, Nouér SA, Morales H, et al. Epidemiology of invasive fungal disease in haematologic patients. Mycoses 2021; 64:252.
  68. Nucci M, Anaissie EJ, Queiroz-Telles F, et al. Outcome predictors of 84 patients with hematologic malignancies and Fusarium infection. Cancer 2003; 98:315.
  69. Garnica M, da Cunha MO, Portugal R, et al. Risk factors for invasive fusariosis in patients with acute myeloid leukemia and in hematopoietic cell transplant recipients. Clin Infect Dis 2015; 60:875.
  70. Chan TS, Au-Yeung R, Chim CS, et al. Disseminated fusarium infection after ibrutinib therapy in chronic lymphocytic leukaemia. Ann Hematol 2017; 96:871.
  71. Nucci M, Marr KA, Queiroz-Telles F, et al. Fusarium infection in hematopoietic stem cell transplant recipients. Clin Infect Dis 2004; 38:1237.
  72. Sampathkumar P, Paya CV. Fusarium infection after solid-organ transplantation. Clin Infect Dis 2001; 32:1237.
  73. Abbara S, Freeman AF, Cohen JF, et al. Primary Invasive Cutaneous Fusariosis in Patients with STAT3 Hyper-IgE Syndrome. J Clin Immunol 2023; 43:647.
  74. Gangneux JP, Dannaoui E, Fekkar A, et al. Fungal infections in mechanically ventilated patients with COVID-19 during the first wave: the French multicentre MYCOVID study. Lancet Respir Med 2022; 10:180.
Topic 2426 Version 26.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟