Chapter 11
Endogenous Fungal Endophthalmitis
ROY D. BROD, HARRY W. FLYNN, JR., and DARLENE MILLER
Main Menu   Table Of Contents

Search

CLASSIFICATION OF FUNGI
ANTIFUNGAL THERAPY
ENDOGENOUS CANDIDA ENDOPHTHALMITIS
ENDOGENOUS ASPERGILLUS ENDOPHTHALMITIS
ENDOGENOUS CRYPTOCOCCUS ENDOPHTHALMITIS
ENDOGENOUS COCCIDIOIDES ENDOPHTHALMITIS
ENDOGENOUS SPOROTHRIX SCHENCKII ENDOPHTHALMITIS
ENDOGENOUS BLASTOMYCES DERMATITIDIS ENDOPHTHALMITIS
OCULAR HISTOPLASMOSIS
RARE CAUSES OF ENDOGENOUS FUNGAL ENDOPHTHALMITIS
CONCLUSION
REFERENCES

Blood-borne fungal infections of the inner eye remain an uncommon but serious cause of ocular inflammatory disease. The incidence may have increased in recent years because of several factors, including the increasing number of immunosuppressed patients receiving aggressive antineoplastic and other therapeutic regimens, organ transplant patients, as well as the use of newer, potent broad-spectrum antibiotics that reduce normal flora.1,2 Patients with AIDS and patients with a history of intravenous substance abuse also contribute to the increased risk for endogenous fungal endophthalmitis.3–11

The metastatic infection may involve only the choroid and retina (as in localized fungal chorioretinitis), may progress into the vitreous or anterior chamber fluids (fungal endophthalmitis), or may involve all ocular tissues (fungal panophthalmitis). A variety of fungi have been reported to cause endogenous intraocular infection, but by far the most common is Candida species.1 Candida albicans is the most common yeast isolate and Aspergillus is the second most common in reported series. Other fungi reported to cause intraocular infection are shown in Table 1.1–12

 

TABLE 1. Classification of Fungi Recovered in Endogenous Fungal Endophthalmitis

  Yeasts and yeastlike isolates
  Candida species

  C. albicans
  C. parapsilosis
  C. tropicalis
  C. glabrata


  Cryptococcus neoformans
  Trichosporon beigelii
  Hyaline molds--septate (colorless hyphae)
  Aspergillus species

  A. fumigatus
  A. niger
  A. glaucus
  A. flavus


  Pseudoallercheria boydii
  Fusarium species

  F. solani
  F. oxysporum


  Bipolaris hawaiiensis
  Paecilomyces species
  Penicillium
  Hyaline molds--aseptate (colorless hyphae)
  Mucor species
  Absidia species
  Rhizopus species
  Dermatiaceous molds (colored hyphae)
  Scedosporium apiospermum
  Cladophialophora bantiana
  Dimorphic molds
  Blastomyces dermatitidis
  Histoplasma capsulatum
  Sporothrix schenckii
  Coccidioides immitis

 

A myriad of ocular inflammatory and neoplastic conditions can resemble endogenous fungal endophthalmitis (Table 2). In some cases, differentiation is relatively straightforward; in others, it can be difficult. In most of these conditions a detailed history, careful examination, and appropriate laboratory studies will lead to the correct diagnosis.13,14

 

TABLE 2. Differential Diagnosis for Endogenous Fungal Endophthalmitis


Endogenous bacterial endophthalmitisHarada disease
ToxoplasmosisPosterior scleritis
SarcoidosisOcclusive diseases of the choriocapillaris associated with systemic disease (e.g., DIC, TTP)
TuberculosisSteroid-induced severe central serous retinopathy
Pars planitisUveal effusion syndrome
Ocular lymphomaSympathetic ophthalmia
RetinoblastomaPolyarteritis
Toxocara canisAPMPPE
SyphilisSerpiginous choroiditis
Lyme disease 
Beh<acc>et disease 
Lens-induced uveitis 
Amyloidosis 

 

This chapter will review the classification of fungi, predisposing factors, signs and symptoms of the systemic and ocular fungal infections, diagnosis, treatment, and complications. Because Candida species is by far the most common cause of endogenous fungal endophthalmitis, most of the chapter will focus on these organisms.

Back to Top
CLASSIFICATION OF FUNGI
There exist more than 50,000 species of fungi, but only 100 to 150 of them have been implicated in clinical disease (see Table 1). This diverse group may be classified or differentiated by morphology (unicellular [yeast/yeastlike] or multicellular [molds]), pigmentation (moniliaceous or dermatiaceous), virulence (pathogenic or opportunistic), or clinical presentation (cutaneous, subcutaneous, or systemic).

Yeasts or yeastlike fungi (e.g., C. albicans) are single-cell organisms, typically round or oval, that form discrete, smooth, creamy and moist colonies (Fig. 1) within 24 to 72 hours on routine microbiologic media. They reproduce or multiply by budding. The characteristic appearance is shown in Figures 2 and 3. Molds (e.g., Aspergillus fumigatus) are multicellular fungi that contain tubular structures (hyphae) and grow by branching and longitudinal extension. These are identified by their macroscopic (Fig. 4) appearance (e.g., cottony, woolly, powdery) or microscopic structures (conidia, hyphal segmentation), and color (Fig. 5). Some fungi may grow with both yeastlike and hyphal morphology in tissues or culture.

Fig. 1. Candida albicans on culture plate growing on filter paper from vitrectomy specimen.

Fig. 2. Candida albicans on Giemsa stain showing budding.

Fig. 3. Candida albicans on Calcofluor white showing budding.

Fig. 4. Aspergillus fumigatus on culture plate.

Fig. 5. Typical broad hyphae of Aspergillus fumigatus on Giemsa stain.

Back to Top
ANTIFUNGAL THERAPY
The management of endogenous fungal endophthalmitis continues to evolve with the development of newer antifungal agents. The specific management approach remains controversial and depends on the type of fungus, degree of ocular involvement, presence of systemic infection, and the overall health of the patient. There are three major modes of treatment: systemic administration of an antifungal agent, intravitreal administration of an antifungal agent, and pars plana vitrectomy.

AMPHOTERICIN B

Amphotericin B has traditionally been the drug of choice for treating most fungal infections.15 Although it still remains an important drug for treating fungal infections, some of the newer azole compounds are gaining increased acceptance for treatment in many situations.

Amphotericin B is one of several polyene antibiotics (Table 3). It was discovered in 195616 and first used for the treatment of disseminated Candida infection in 1960.15 It acts by binding to cell membrane sterols, destroying the cell membrane and leading to cell death. It is effective against most fungi (Table 4). Amphotericin B is not absorbed by the gastrointestinal tract and must be given intravenously for systemic effect. The usual daily dose is 0.5 to 1 mg/kg; the average daily dose is 40 to 50 mg. It must be administered over a period of 4 to 6 hours. For most infections, treatment needs to be continued for at least several weeks.17–19

 

TABLE 3. Systemic Antifungal Agents


DrugRoute of AdministrationDoseMechanism of ActionApplicationAdverse ReactionsComment
AmphotericinBIntravenous0.5-1.0 mg/kg (recommended total dose varies)Disrupts fungal cell membranes by sterol bindingDrug of choice for severe systemic candidiasis, aspergillosis, blastomycosis, histoplasmosis, cryptococcosis, and coc-cidio-ido-my-co-sis (immunocompromised patients)Renal dysfunction, chills, fever, vomiting, nausea, diarrhea, malaise, anemia, arrhythmia, hypokalemia, hearing loss (concurrent administration of 25 mg hydrocortisone may reduce hyperthermia)Poor intraocular penetration in both animal model and human studies. Severe side effects limit usefulness
FlucytosineOral50–150 mg/kg at 6-hr intervalsInhibits fungal RNA and DNA synthesisUseful in combination with amphotericin B for candidiasis and cryptococcosisNausea, vomiting, diarrhea, rash, headache, anemia, leukopenia, hepatic dysfunction (rare), bowel perforation (rare)Up to 47% of Candida species resistant Fair intraocular penetration
MiconazoleIntravenous600–3600 mg/daydepending on organismInhibits synthesis of fungal membrane sterolsThird- or fourth-line drug for serious systemic infection with Candida, Cryptococcus, and Aspergillus First-line drug for Petriellidium boydii infectionsThrombophlebitis at intravenous access site, nausea, vomiting, diarrhea, pruritus, thrombocytopenia, hyperlipidemia, hyponatremia, arrhythmia, anaphylaxis, central nervous system toxicityIntravitreal penetration variable Taken off market in 1997
KetoconazoleOral200–800 mg/day in one or two doses (prolonged therapy may be required)Inhibits ergosterol synthesis in fungal cell membranesExperimental and clinical studies suggest efficacy in treating systemic candidiasis, aspergillosis, cryptococcosis, blastomycosis, histoplasmosisGenerally mild but may have nausea, vomiting, diarrhea, pruritus, hepatic dysfunction, gynecomastia, decrease in serum testosteroneGood intraocular penetration in an animal model
FluconazoleOral Intravenous200–400 mg/day in one dose (prolonged therapy may be required)Inhibits ergosterol synthesis in fungal cell membranesExcellent activity against Candida albicans, crypto-coccus, coccidioidomycosis, Candida krusei resistant. Not effective against most strains of Aspergilllus. May be used to treat mild cases of endophthalmitis caused by Candida and to supplement local occular therapy in severe casesHepatic dysfunction (usually mild and reversible), nausea, headache, rash, abdominal pain, vomiting, diarrheaExcellent intraocular penetration
ItraconazoleOral200–400 mg/day in two divided doses (prolonged therapy may be required)Inhibits ergosterol synthesis in fungal cell membraneEffective against Aspergillus, Histoplasmosis, Blastomycosis, Pseudallescherio boydiiHepatic dysfunction nausea, vomiting, diarrhea, rash, endocrine effects, hypertension, hyperkalemia, adrenal insufficiencyGood intraocular penetration

 

 

TABLE 4. Susceptibility Patterns for Common Ocular Fungal Pathogens


OrganismAmphotericin BFlucytosineFluconazoleItraconazoleKetoconazole
C. albicans+ ++ ++++ +
C. glabrata+ ++ +++/-+ +
C. parapsilosis+ ++ ++ ++ ++ +
C. tropicalis+ ++ ++/-++ +
C. neoformans+ ++ ++ ++ ++ +
A. flavus+--+ ++ +
A. fumigatus+ +--+ ++ +
A. glaucus+/---++
A. niger+ ++/--++ +
A. terreus- ---+ ++ +

C., Candida; A., Aspergillus; + + , clinically effective or MIC90 > 90% sensitive; + , clinically effective or MIC90 > 60% sensitive; +/, clinical trials lacking or MIC90 30%---60% sensitive; , not clinically effective or MIC90 < 30% sensitive.

 

Besides the need for intravenous administration, amphotericin B has other disadvantages (Table 5). One of the major disadvantages is its significant toxicity.17 Renal dysfunction is the most notable toxic effect and occurs in most patients if the drug is administered for a long enough time. The renal toxicity is usually reversible with cessation of the drug, but permanent renal impairment may result. Another limitation of systemically administered amphotericin B for endogenous fungal endophthalmitis is its poor intraocular penetration.20–23 The nonfenestrated capillaries in the eye do not permit penetration of large and poorly lipid-soluble molecules such as amphotericin B. For this reason, systemic treatment alone is usually not effective for well-established endogenous fungal endophthalmitis with significant vitreous involvement. Although it is usually very effective for many types of fungal chorioretinitis and systemic fungal infections in general, these serious side effects limit the use of amphotericin B for many patients.24

 

TABLE 5. Disadvantages of Systemic Amphotericin B

  Renal toxicity
  Poor intraocular penetration
  Intravenous administration necessary

 

Amphotericin B can be injected directly into the vitreous cavity (Table 6). Axelrod and coworkers25 first demonstrated the effectiveness of this technique in 1973 in a rabbit model experiment. Doses of 5 to 10 μg were shown to be nontoxic to the rabbit eye.26,27 A single amphotericin B injection of up to 20 μg was shown to be nontoxic in the monkey eye.28 Liposomal amphotericin B was tolerated in doses that were at least fourfold higher than that.27,28 When amphotericin B is injected into the vitreous, it readily reaches the choroid and retinal tissues.29 The drug has a long ocular half-life in nonvitrectomized eyes.30,31 Concentrations from 0.4 to 1 μg/ml were found in the vitreous 5 weeks after injections of 5 to 20 μg.28 Lens extraction and vitrectomy considerably reduce the half-life to 1.8 days.30,31 These half-life data are important when considering reinjection of amphotericin B. In nonvitrectomized eyes, reinjection is probably not necessary for at least a week after the initial injection; however, in eyes that have undergone prior vitrectomy, reinjection can be considered at 3 to 4 days if the initial injection fails to achieve a favorable clinical response. The safety of intravitreal amphotericin B in human eyes is well established.32 A near-normal electroretinogram was recorded 1 year after injection of 20 and 30 μg of intravitreal amphotericin B over 48 and 96 hours, respectively, in a patient with postsurgical Candida parapsilosis endophthalmitis.33 In another case report, 11 separate intravitreal injections of 10 μg amphotericin B over 14 days resulted in a normal electroretinogram 5 months and 5 years after treatment.34

 

TABLE 6. Intravitreal Antifungal Agents


DrugDoseApplicationComment
Amphotericin B5–10 μgIntravitreal antifungal agent of choice for most endogenous fungal endophthalmitis with moderate to severe vitreous involvementMay be used in eyes after vitrectomy
MiconazoleUp to 40 μgEndogenous fungal endophthalmitis with moderate to severe vitreous involvement when resistance to amphotericin B and sensitivity to iconazole demonstrated in vitro May be drug of choice for Petriellidium boydii endophthalmitisMay be used in eyes after vitrectomy
Ketoconazole in dimethylsulfoxide540 μg nontoxic in rabbit eyesUnknownExperimental
Fluconazole100 μg nontoxic in rabbit eyes 5–10 μg used without toxicity in humansUnknownExperimental
Itraconazole in dimethylsulfoxide10 μg nontoxic in rabbit eyesUnknownExperimental
Flucytosine100 μg nontoxic in rabbit eyesUnknownExperimental

 

The efficacy of intravitreal amphotericin B in treating endogenous Candida endophthalmitis has been demonstrated in multiple reports.35–43 Favorable outcomes have been reported when used in combination with vitrectomy as well as when used as the sole treatment. Strains of C. albicans resistant to amphotericin B have been reported to cause endogenous endophthalmitis, but it is more common to find resistance in the non-albicans strains of Candida.44–48 Fortunately, they are a much less frequent cause of endogenous fungal endophthalmitis. Pseudallescheria boydii and Aspergillus terreus are also frequently resistant to amphotericin B.

FLUCYTOSINE

Flucytosine was discovered in 1957 and first used to treat Candida in 1968.49,50 Flucytosine has excellent gastrointestinal absorption. The recommended daily dose is 50 to 150 mg/kg in four divided doses. Peak concentrations occur 2 to 4 hours after a 2-g oral dose. A reduced dose is necessary in patients with impaired kidney function. Side effects are much less common than with amphotericin B (see Table 3).

Flucytosine has reasonably good vitreous penetration after oral administration. Vitreous flucytosine levels as high as 22.2 μg/ml have been documented in humans 18 hours after oral administration in a patient with endophthalmitis.51

A major drawback to the use of flucytosine in treating Candida infections is the high incidence of organism resistance. Resistance occurs in 7% to 47% of Candida isolates, and additional resistance develops commonly during treatment.52,53 For this reason, flucytosine is rarely used alone to treat Candida infections. The combination of flucytosine with amphotericin B may improve its in vivo synergism against C. albicans.47,54–56 The use of intravitreal flucytosine in clinical disease has not been reported, but a 100-μg injection has been found to be nontoxic in the rabbit eye57 (Table 6). Flucytosine has no activity against most of the molds recovered from endogenous endophthalmitis.

MICONAZOLE

Miconazole is one of four azole drugs used as a systemic agent. It must be given intravenously because of poor gastrointestinal absorption.58,59 The recommended daily dose for systemic fungal infection is 600 to 3600 mg in three divided doses. It has a relatively broad spectrum of activity for fungi. Much of the experience with this drug has been in treating coccidioidomycosis.60,61 It is not an ideal drug for Candida infections because of its relatively high minimum inhibitory concentration (MIC) for some Candida species.61 It may be more effective than amphotericin B for infections due to P. boydii and some Paecilomyces species. The intravitreal penetration of systematically administered miconazole may be subtherapeutic. Because of its fairly significant list of side effects, systemic miconazole is not currently available for the treatment of fungal endophthalmitis (see Table 3).62–66 An intravitreal (see Table 6) dose of 40 μg has been found to be nontoxic in animal studies.66

KETOCONAZOLE

Ketoconazole was approved for systemic use in 1981.67 It acts by inhibiting fungal cell membrane sterols.68 Because it is well absorbed through the gastrointestinal tract, ketoconazole can be given orally. Gastrointestinal absorption may be variable, however, and depends on gastric acidity. Serum levels peak at about 3 to 6 μg/ml 2 hours after a 200-mg oral dose.69 It is a relatively nontoxic drug (see Table 3): its major adverse effect is hepatic dysfunction, ranging from asymptomatic elevation of liver enzymes to fatal hepatic necrosis. Asymptomatic elevations in liver function studies occur in 5% to 10% of patients; serious hepatic injury occurs in about 1 in 15,000 persons treated with the drug. Because of this, it is important to monitor liver function in patients taking ketoconazole.70–72 It is active against a variety of fungi; however, resistant strains have been reported.73–76

Conflicting data have been reported regarding the intravitreal penetration and efficacy of orally administered ketoconazole for treating endogenous Candida endophthalmitis.77–79 In some studies, undetectable vitreous ketoconazole levels were found in healthy rabbit eyes despite adequate serum levels.77 The penetration appears to be better in inflamed eyes, however. A vitreous cavity ketoconazole level of 0.92 μg/ml was detected 8 hours after a 600-mg dose in a patient with postoperative Candida endophthalmitis. This eye had undergone a previous vitrectomy.79 The safety of intravitreal ketoconazole (see Table 6) has been shown in animal studies, but its use in treating clinical infection in humans has not been reported.80

Although when first developed ketoconazole was a significant addition to the available antifungal therapeutic agents, newer and more effective azole compounds, including fluconazole and itraconazole, have now surpassed it.

FLUCONAZOLE

Fluconazole is one of the azole group of antifungal agents.81 It is highly aqueous-soluble and has excellent absorption properties to the gastrointestinal tract.82,83 It is the first antifungal agent that can be given both orally and intravenously.81 Its mode of action is inhibition of ergosterol found in the cell membranes of yeasts and other fungi. It is highly selective for fungal cells. Fluconazole is generally well tolerated, with a low incidence of adverse effects (see Table 3). Because of its long half-life, once-daily dosing is usually adequate. The recommended dose varies depending on the type and severity of infection and ranges from 100 to 400 mg/day. Because it is weakly protein-bound and highly water-soluble, it penetrates well into most body tissues, including the eye and cerebrospinal fluid.78,84–86 It is excreted primarily by the kidney, and the dose must be adjusted accordingly in patients with impaired creatinine clearance.82,83

It is active against a large variety of fungi, including most species of Candida. Candida krusei is well known to be resistant to fluconazole. Candida glabrata may develop resistance during therapy, and reports of acquired resistance to azoles by C. albicans have surfaced. This is usually seen in HIV-infected patients with oropharyngeal or esophageal candidiasis.87,88 Most species of Aspergillus are resistant to fluconazole.

A major advantage of fluconazole in treating endogenous endophthalmitis is its excellent intravitreal penetration. Several studies have confirmed this, both in noninflamed and inflamed animal and human eyes.78,84,85,89 Two studies looked at the efficacy of treating experimentally induced Candida endophthalmitis in rabbits.90,91 In both studies, fluconazole was effective in preventing the establishment of ocular infection when administered shortly after inoculation of Candida, but it was not effective when administered 3 to 6 days after inoculation in one study and 7 days after inoculation in the other study. In both studies, the eyes were treated for only a relatively short period, and this may be a major factor in the lack of established efficacy in these studies.

Several clinical case reports have demonstrated the efficacy of oral fluconazole for treating endogenous Candida endophthalmitis.92–100 These cases include eyes also treated with vitrectomy but without intravitreal amphotericin B, as well as eyes in which no vitrectomy was performed. In most patients, the dosage required to eliminate the intraocular infection was 400 mg/day. These successfully treated eyes include those with only chorioretinitis and minimal to no vitritis, as well as a few eyes with significant vitritis. Postmortem evaluation of eyes from a patient with endogenous Candida endophthalmitis treated for 2 weeks with 200 mg/day fluconazole demonstrated no evidence of organisms in the retina or choroids, but organisms were found in vitreous opacities. This case underscores the need for prolonged treatment with fluconazole when infection involves the vitreous.100 Although these successfully treated patients are encouraging, there are a few reports of progressive intraocular Candida infection despite ongoing oral fluconazole therapy.101,102

Intravitreal fluconazole (5 to 10 μg) (see Table 6) has been shown to be safe and effective for treating fungal endophthalmitis in animal and human studies.103,104 Because of the limited clinical experience and lack of a distinct benefit over intraocular amphotericin B, it is not currently recommended.

ITRACONAZOLE

Itraconazole, like fluconazole, is in the class of triazole compounds. Unlike fluconazole, however, itraconazole is very insoluble in aqueous fluids and is high protein-bound. It does not pass into the cerebrospinal fluid and penetrates into the eye less well than fluconazole. Itraconazole has the edge over fluconazole with respect to direct antifungal activity in vitro. It is well absorbed orally, and its toxicity is relatively low and similar to that of fluconazole105,106 (see Table 3).

Despite relatively low drug concentrations in the vitreous compared with fluconazole, it appeared to have similar efficacy in a rabbit model of Candida endophthalmitis.78 In this model it was also similar to fluconazole in its effectiveness in preventing endophthalmitis, but it was less effective in established cases. In general, itraconazole does not appear to have any significant advantage over fluconazole in treating endogenous Candida endophthalmitis, but it is much more effective than fluconazole for Aspergillus endophthalmitis. It is also more effective than amphotericin B for treating infections caused by P. boydii.

NEWER ANTIFUNGAL THERAPIES

New commercially available antifungal agents include the lipid formulations of amphotericin B.26 These include amphotericin B lipid complex, amphotericin B colloidal dispersion, and the small unilamellar vesicle formulation of amphotericin B.2,107 Studies have demonstrated that these compounds are similar or superior to conventional amphotericin B in antifungal efficacy and are less nephrotoxic at therapeutically equivalent doses.2 Data regarding their use in ocular fungal infections are not available, and the poor intraocular penetration will likely limit their potential.108

Newer antifungal compounds that attack novel targets are being developed. Two agents being investigated are the third-generation broad-spectrum triazoles, voriconazole and SCH-56592. Other drugs being investigated include the echinocandins and pneumocandins. These drugs target cell wall biosynthesis by classic pathogenic fungi. In addition, several measures are being studied to augment the host response in the hopes of treating or preventing invasive fungal infections.109

Back to Top
ENDOGENOUS CANDIDA ENDOPHTHALMITIS

CHARACTERISTICS OF CANDIDA SPECIES

Candida species exists in both a yeast and hyphal phase, depending on the environment. Candida grows rapidly on most common culture media at room temperature. Creamy, white colonies begin to appear at 24 to 48 hours after inoculation. C. albicans is a normal inhabitant of the gastrointestinal tract and mucous membranes, including the mouth and vagina.2 Immunosuppression, debilitation, or other predisposing factors may allow Candida organisms to become pathogenic. C. albicans is the most frequent species of Candida causing endogenous Candida endophthalmitis, but several other species of Candida have been isolated (see Table 7).

 

TABLE 7. Candida Species Reported to Cause Endogenous Fungal Endophthalmitis

  Candida albicans
  Candida tropicalis
  Candida parapsilosis
  Candida glabrata
  Candida guillermondi
  Candida krusei

 

PREDISPOSING FACTORS

The same factors that predispose a person to develop candidemia predispose to endogenous Candida endophthalmitis, because by definition this represents a blood-borne infection. In the 1980s, intravenous drug abuse was the most common predisposing factor,12 but in the 1990s, reported cases of endogenous Candida endophthalmitis related to intravenous drug abuse declined.1 The ocular infection usually results from a transient candidemia. Intravenous drug abusers can also develop a distinctive candidiasis syndrome that includes skin and scalp nodules, osteomyelitis, and costochondritis. A published 10-year review (1984 to 1994) of endogenous fungal endophthalmitis demonstrated that 22% of the cases (4 of 18 patients) had a history of intravenous drug abuse.1,4–8,110–116

A more common predisposing factor in cases of endogenous Candida endophthalmitis is use of a long-term intravenous line.1,117,118 In these cases, it is likely that the Candida organisms enter the blood stream directly from contaminated catheter tips. In a 10-year endogenous fungal endophthalmitis study reported by Essman and coworkers,1 67% of the patients has long-term intravenous line placement. Other important predisposing factors are listed in Table 8.119–135 Persons with diabetes are also predisposed to develop systemic Candida infection, possibly because of increased glucose available for sustaining the Candida organism as well as the patient's relative immune compromise. Persons with diabetes are also predisposed to develop cutaneous ulcers that may be contaminated with Candida. Corticosteroid use may also predispose to Candida infection by increasing available glucose.136

 

TABLE 8. Conditions Predisposing to Candidemia and Endogenous Candida Endophthalmitis

  Long-term intravenous line placement
  Parenteral hyperalimentation
  Prolonged antibiotic therapy
  Systemic corticosteroids
  Immunosuppressive therapy
  Abdominal surgery
  Hemodialysis
  Intravenous drug abuse
  Acquired immunodeficiency syndrome
  Malignancy
  Diabetes mellitus
  Pregnancy
  Massive trauma
  Alcoholism
  Hepatic insufficiency
  Postpartum
  Prematurity
  Genitourinary manipulation

 

Multiple predisposing factors are often present in patients with systemic Candida infection or endogenous Candida endophthalmitis.137–149 In rare cases, endogenous Candida endophthalmitis has been reported in otherwise healthy persons with no predisposing factors or events.126,150,151 The ocular infection can occur weeks or months after the predisposing factor has been eliminated, and a careful history looking for preceding risk factors should be elicited in patients suspected of having an intraocular Candida infection.1,135,138,152–156

RELATION BETWEEN CANDIDEMIA AND ENDOGENOUS CANDIDA ENDOPHTHALMITIS

Several studies have evaluated the relation between candidemia, candidiasis, and ocular Candida infection (Table 9).137,157–165 This relation is important in determining which patients are at highest risk for the development of intraocular Candida infection and generating guidelines for screening these high-risk patients. The rate of development of intraocular Candida infection in patients with candidemia or candidiasis has been reported to range from 2.8% to 45%.158–164 Two prospective studies using more rigid clinical criteria for ocular involvement have been reported.158,164 The first study examined 118 patients within 72 hours of positive blood cultures for Candida. Only 11 patients (9%) had fungal chorioretinitis, and none had vitreous involvement. All patients in the study were treated with amphotericin B intravenously, and none subsequently developed vitreous involvement.161 In the second study, 214 eyes in 107 patients with a diagnosis of systemic fungal infection were studied. Of the patients examined, 93.4% already were receiving systemic antifungal therapy at the time of ophthalmologic consultation. Only 3 (2.8%) of the 107 patients examined had findings consistent with fungal chorioretinitis. No patient had vitreous involvement, and the ocular findings did not progress during the course of therapy.164 In these two papers reported in the 1990s, the incidence of Candida endophthalmitis was markedly less than in previously published series in the 1970s and 1980s, in which the incidence of endophthalmitis in patients with candidemia was reported to be in the range of 30% to 40%.137,157,159,161,163 The lower incidence in recently reported studies may be related to earlier diagnosis and more aggressive initial treatment of candidemia and candidiasis by primary and intensive care physicians. Systemic antifungal drugs with better ocular penetration may also have contributed to this. Despite the lower incidence, screening ophthalmologic evaluation of hospitalized patients with documented candidemia or disseminated candidiasis is generally recommended, because early detection and treatment are important in reducing the degree of ocular damage.

A 1992 study evaluating the usefulness of screening children at risk for fungal endophthalmitis demonstrated that only patients with deep-tissue fungal infection were at significant risk for developing endogenous fungal endophthalmitis.166 The authors concluded that ophthalmoscopy with pupillary dilation is necessary for patients with invasive fungal disease and may be useful for at-risk patients with superficial fungal colonization, but this type of examination is not of documented value in immunosuppressed children with negative fungal cultures.

 

TABLE 9. Reported Rates of Candida Intraocular Infection in Patients With Systemic Candida Infection


AuthorYearNumber of Patients StudiedNumber with Ocular InfectionComments
Griffin et al15919768225 (30%)Postmortem and clinical cases
Klein and Watanakunakorn13519797711 (14%) 
Parke et al13719822710 (37%)Only patients with C. albicans
Eilard162198714218 (13%)Only patients with C. albicans
Bross et al16119894821 (44%)Diagnostic criteria not specified
Brooks1631989329 (28%)Roth spots included
Donahue et al158199411811 (9%)Chorioretinitis only; prospective study
Scherer and Lee16419971073 (2.8%)Chorioretinitis only

 

The relation between the presence of intraocular Candida infection and systemic Candida infection is important. This relation seems to vary depending on whether the patient with the Candida ocular infection is an inpatient or an outpatient. This relation has also been studied in autopsy series. In hospitalized patients, it appears that the presence of intraocular Candida infection often signifies diffuse systemic involvement. In one study,160 4 of 11 patients with Candida chorioretinitis died within 14 days of their ocular examination, and 2 of the 3 patients described by Scherer and Lee164 died before discharge from the hospital. Previous autopsy series also indicated a very high incidence of coexistent disseminated candidiasis in patients with endogenous Candida endophthalmitis. In a study by Edwards and associates,167 77% of 39 autopsy cases of endogenous Candida endophthalmitis had evidence of candidal infection in major organs. These findings are in sharp contrast to outpatients with intraocular Candida infection. In the study by Essman and associates,1 only 1 of the 16 patients with endogenous Candida intraocular infection had evidence of systemic infection. This patient had a nonhealing foot ulcer that grew Candida. All the patients in this study came to the outpatient eye clinic at an eye institute. Another outpatient series of patients with endogenous Candida endophthalmitis also demonstrated a low incidence of systemic Candida involvement.138 These outpatient cases probably result from a transient candidemia with seeding to the eye. Although the immune system often can eliminate the systemic infection when the offending nidus is removed (an indwelling venous catheter), the vitreous cavity is more isolated, and Candida organisms may continue to replicate in the eye, resulting in endophthalmitis. It is therefore important to consider Candida infection in patients with visual complaints or intraocular inflammation who are not acutely ill but may have had previous systemic risk factors or been recently discharged from a hospital.

CLINICAL FINDINGS

The most common presenting symptoms (Table 10) of Candida endophthalmitis are blurred vision, pain, photophobia, and ocular redness. Patients may also complain of floaters, cobwebs, or veils across their vision. Severely ill or debilitated patients may be unaware of or unable to express visual symptoms. The two most characteristic clinical signs (Table 11) are creamy white, well-circumscribed chorioretinal lesions (Fig. 6), most common in the posterior pole, and yellow or white fluffy vitreous opacities. They may be connected by strands of inflammatory material, producing what has been called a string-of-pearls appearance (Fig. 7). These focal vitreous opacities have been shown to evolve from the chorioretinal inflammatory lesions after they have broken through into the vitreous. These vitreous opacities are composed of inflammatory cells and often contain Candida organisms. Less specific posterior findings include vitritis, vasculitis, papillitis, and retinal hemorrhages. These retinal hemorrhages sometimes surround the white chorioretinal lesions, mimicking a Roth spot.1,3,12,152,153,167,168

 

TABLE 10. Symptoms of Endogenous Candida Endophthalmitis

  Blurred vision
  Floaters
  Pain
  Photophobia
  Ocular redness

 

 

TABLE 11. Signs of Endogenous Candida Endophthalmitis

  Anterior Segment
  Conjunction injection
  Corneal edema
  Anterior chamber cell and flare
  Hypopyon
  Fibrin
  Posterior Segment
  White chorioretinal lesions
  White fluffy vitreous opacities (“string of pearls”)
  Vitritis
  Vasculitis
  Papillitis
  Retinal hemorrhages

 

Fig. 6. A.Candida albicans endogenous fungal endophthalmitis manifesting as multiple chorioretinal lesions in a patient with candidiasis. B. Regressing chorioretinal lesions after systemic treatment with amphotericin B.

Fig. 7. Candida albicans endogenous fungal endophthalmitis with string-of-pearls appearance. (Flynn HW Jr, Essman TF, Brod RD: Endogenous fungal endophthalmitis. In Saer JB: Vitreoretinal and uveitis update. Proceedings of the 47th Annual Symposium of the New Orleans Academy of Ophthalmology, New Orleans, April 3–5, 1998, pp 297–305. The Hague: Kugler Publications, 1998.)

Anterior segment findings are relatively nonspecific and include conjunctival discharge and injection, keratic precipitates, anterior chamber cell, and flare, usually without a hypopyon. When a hypopyon is present it is usually small. Episcleritis and scleritis are uncommon. After treatment, depigmented chorioretinal scars develop in areas of previously active chorioretinitis (Fig. 8). Choroidal neovascular membranes can develop in these scars, particularly when they are in the macular area.169,170

Fig. 8. A. Candida albicans endogenous fungal endophthalmitis with white chorioretinal lesion elevated into the vitreous cavity. Because of progression to vitreous seeding, treatment with vitrectomy and intravitreal amphotericin B was necessary. B. Resolved lesion showing chorioretinal white scar after treatment. (Flynn HW Jr, Essman TF, Brod RD: Endogenous fungal endophthalmitis. In Saer JB: Vitreoretinal and uveitis update. Proceedings of the 47th Annual Symposium of the New Orleans Academy of Ophthalmology, New Orleans, April 3–5, 1998, pp 297–305. The Hague: Kugler Publications, 1998.)

Late findings and complications of the infection include epiretinal membrane formation causing macular distortion as well as tractional and rhegmatogenous retinal detachment.171–174 Cyclitic membranes can lead to hypotony and finally phthisis bulbi. The latter finding is usually seen in chronic cases where there was a delay in initiation of specific treatment. Parenteral drug abusers with Candida endophthalmitis often have a more severe inflammatory response than immunosuppressed or debilitated patients with Candida endophthalmitis. The potential reasons for this difference include a delay in presentation or diagnosis, as well as the lack of an associated immunosuppressive condition contributing to a reduced inflammatory response.

DIAGNOSIS

An important aspect in the diagnosis is maintaining a high index of suspicion for endogenous Candida ocular infection. In patients with known systemic Candida infection and ocular inflammatory findings consistent with intraocular Candida infection, a clinical diagnosis can often be made without the need for intraocular specimens. When systemic Candida infection is not clinically apparent or is undiagnosed in a patient suspected of having intraocular Candida infection, a definitive diagnosis often requires analysis of intraocular specimens (aqueous, vitreous, or both). The fresh specimens should be processed for specific stains and culture (Table 12). The presence of the characteristic mixture of yeast and mycelial phase organisms on stained smears is useful for diagnosis. Candida grows rapidly on most common culture media, but Sabouraud's agar without cyclohexamide at room temperature is recommended when Candida is clinically suspected (Fig. 9). More rapid and specific diagnostic tests are being evaluated by polymerase chain reaction and by detection of fragment length polymorphism.12,175

 

TABLE 12. Laboratory Techniques for Isolation or Identification of Fungi

  Direct Detection
  Stains

  Calcofluor white stain
  Giemsa stain
  Gram stain
  Methenamine silver staining
  Potassium hydroxide
  Periodic acid-Schiff stain
  India Ink


  Molecular detection

  DNA probes
  Polymerase chain reaction
  DNA subtyping (RAPD, REA)


  Cultivation
  Solid media

  Sabouraud's dextrose agar
  Blood agar
  Brain-heart infusion agar


  Liquid media

  Brain-heart infusion agar broth/blood
  Culture media


  Identification
  Growth characteristic and morphology (molds)
  Biochemical kits (yeasts)
  DNA probes (yeasts and molds)
  Polymerase chain reaction (yeasts and molds)
  Immunodiagnosis


RAPD, random amplification of polymorphic DNA; REA, restriction endonuclease analysis.

 

Fig. 9. Candida albicans on Sabouraud's agar.

MANAGEMENT

The management of patients with endogenous Candida endophthalmitis must be individualized and depends on the degree of ocular involvement and presence of systemic infection (Table 13). Patients without evidence of systemic Candida infection who have ocular infection localized to the choroid and retina can usually be successfully treated with oral fluconazole at a dose of 100 to 200 mg/day. Treatment usually needs to be continued for 2 months or more, depending on the clinical response. Oral therapy with fluconazole can usually be administered on an outpatient basis and is generally successful. When mild to moderate vitritis complicates the chorioretinitis, oral fluconazole treatment alone can be considered, but the success rate is not as high and frequent follow-up visits are important to detect progressive vitreous infection.

 

TABLE 13. Management of Endogenous Candida Endophthalmitis

  Document characteristic clinical findings.
  Make presumed diagnosis in presence of known systemic Candida infection.
  Initiate systemic workup when systemic Candida infection not already known.
  When vitreous is involved, obtain specimen for stains and culture.
  Consider newer rapid and specific diagnostic tests (polymerase chain reaction) if available.
  Consider pars plana vitrectomy and injection of intravitreal amphotericin B.

 

When there is a more severe vitritis manifested by vitreous infiltrates, systemic therapy with fluconazole is usually not adequate and vitrectomy combined with intravitreal amphotericin B (5 to 10 μg) is often recommended. Oral fluconazole can be used to supplement the local ocular treatment. One series reported that early vitrectomy might be superior to a delay in these more severely involved eyes.1

The use of intraocular corticosteroids (dexamethasone 400 μg) can be considered as an adjunctive treatment at the time of vitrectomy and intravitreal amphotericin B. This has been shown to reduce more rapidly the intraocular inflammatory response without promoting fungal replication.176,177 It is important to use steroids only when specific antifungal therapy has been administered.

Every patient suspected of having endogenous Candida endophthalmitis should have a thorough workup for evidence of systemic infection. The presence of active systemic Candida infection can sometimes be treated with oral or intravenous fluconazole; however, amphotericin B may be recommended in more severe cases and in immunosuppressed patients. Specific recommendations are usually made in conjunction with an internist or infectious disease specialist.

Back to Top
ENDOGENOUS ASPERGILLUS ENDOPHTHALMITIS
Aspergillus species is second only to Candida species as a cause of endogenous fungal endophthalmitis.1 There are more than 900 species of Aspergillus, but only 8 have been implicated in human disease,178 and 5 have been documented to cause intraocular infection (Table 14).179 A. fumigatus is the most common cause of ocular infection. The name Aspergillus (asper meaning rough and gillus meaning head) is derived from the microscopic appearance of the spore-forming structure.

 

TABLE 14. Aspergillus Species Reported to Cause Endogenous Aspergillus Endophthalmitis

  Aspergillus fumigatus
  Aspergillus flavus
  Aspergillus niger
  Aspergillus terreus
  Aspergillus candidus

 

Aspergillus species are ubiquitous in nature and occur worldwide. Aspergillus conidia are frequently airborne both indoors and outdoors; thus, inhalation is a common route of contamination and infection. The organism is also found in soil, water, plants, and dust and is particularly common in agricultural environments.180

PREDISPOSING CONDITIONS

In a 1998 summary of 10 patients (12 eyes) with culture-proven endogenous Aspergillus endophthalmitis, the most common predisposing factors (Table 15) were chronic obstructive pulmonary disease and intravenous drug abuse.179 The ocular infection can be associated with disseminated aspergillosis, but it is also seen in patients without definitive clinical or laboratory evidence of systemic infection. Most patients are immunocompromised either by their underlying disease or iatrogenically. Endogenous Aspergillus endophthalmitis has been reported in association with cardiac transplantation, endocarditis, lung, liver, and kidney transplants, leukemia, and periodontitis.181–202 Patients with no apparent predisposing conditions may also develop aspergillosis and endogenous Aspergillus endophthalmitis. In 15% to 20% of patients with invasive pulmonary aspergillosis, the chest radiographs are normal. Drug abuse appears to be a more common risk factor than recognized in the earlier literature. Another more recently recognized high-risk group are patients undergoing orthotopic liver transplants.203,204

 

TABLE 15. Predisposing Conditions for Endogenous Aspergillus Endophthalmitis

  Chronic obstructive pulmonary disease
  Intravenous drug abuse
  Organ transplantation
  Endocarditis
  Leukemia
  Periodontitis

 

OCULAR MANIFESTATIONS

Most patients with endogenous Aspergillus endophthalmitis have acute or subacute ocular symptoms.179 The most common presenting symptom is blurred vision, sometimes associated with ocular pain, redness, and lid swelling. Most patients have anterior segment inflammatory signs, including anterior chamber cells and keratic precipitates. A hypopyon may be present. In most cases the organism initially affects the posterior segment, forming a yellowish macular infiltrate (Fig. 10). This begins in the choroid and progresses through the subretinal space into the retina. It frequently involves the macula, and this probably accounts for the rapid onset of visual loss in many cases. A frequent clinical feature is the gravitational layering of inflammatory exudates (pseudohypopyon) in either the preretinal (subhyaloid) or subretinal space (Fig. 11). Although this finding is not pathognomonic of Aspergillus infection, when seen it should prompt a high suspicion for Aspergillus. This is usually associated with a vitritis. Other features include occlusive vasculitis and retinal hemorrhages. Orbital involvement can also occur, but this usually results from contiguous spread from an adjacent sinus infected with Aspergillus. After treatment the area of chorioretinitis progresses to formation of a subretinal scar. Because of the predilection for macular involvement, the visual outcome is generally poor.

Fig. 10. A. Aspergillus chorioretinal infiltrate in macula of patient with a history of intravenous drug abuse. B. After treatment with vitrectomy, intravitreal amphotericin B injection, and systemic amphotericin B, the infection resolved and a macular scar remains. (Weishaar PD, Flynn HW Jr, Murray TG et al: Endogenous Aspergillus endophthalmitis: Clinical features and treatment outcomes. Ophthalmology 105:57, 1998.)

Fig. 11. A. Aspergillus chorioretinal infiltrate with pseudohypopyon in preretinal and subretinal location. B. After treatment with pars plana vitrectomy and intravitreal amphotericin B, the lesion resolved into a chorioretinal scar. (Weishaar PD, Flynn HW Jr, Murray TG et al: Endogenous Aspergillus endophthalmitis: Clinical features and treatment outcomes. Ophthalmology 105:57, 1998.)

TREATMENT

Visual outcomes after treatment for endogenous Aspergillus endophthalmitis are generally worse than Candida cases but have improved in recent years. This is most likely due to earlier detection of the infection, leading to more rapid initiation of specific treatment. Previously reported cases were often diagnosed at the time of enucleation or after death.

Although there are significant phenotypic differences in susceptibility between different Aspergillus isolates, the antifungal agent of choice for Aspergillus infection is usually amphotericin B. In vitro sensitivities to amphotericin B are not always reliable and do not necessarily correspond to in vivo response to treatment. To help reduce the toxicity and enhance the effectiveness of intravenous amphotericin B, combinations with 5-fluorouracil and azole compounds have been used. Ketoconazole, clotrimazole, and miconazole have inconsistent efficacy for Aspergillus and are not recommended. Despite fluconazole's excellent vitreous penetration, it shows a relatively high MIC against Aspergillus and is not usually recommended. In one study using in vitro testing, fluconazole did not inhibit any of 45 pathogenic strains of A. fumigatus.205

Itraconazole exhibits a favorably low MIC for many species of Aspergillus, and in selected reports it has shown a favorable response for the treatment of invasive aspergillosis. The vitreous penetration of itraconazole is better than amphotericin but remains only a fraction of the serum level.144,206

Most patients with endogenous Aspergillus endophthalmitis have significant vitreous involvement at the time of diagnosis. For this reason, vitrectomy is recommended in most cases. This is combined with intravitreal injection of amphotericin B, 5 to 10 μg. The goal should be to perform a fairly complete vitrectomy in the hopes of removing as many organisms as possible. The use of intravitreal corticosteroids can be considered to reduce the potentially destructive associated inflammatory response associated with endogenous Aspergillus endophthalmitis. Its role is still unproven, but successfully treated cases using a combination of vitrectomy, amphotericin B, and intravitreal dexamethasone have been reported.179 Intravenous amphotericin B may be used to supplement local ocular treatment, although the poor intraocular penetration may not warrant the potential systemic toxicity of amphotericin B in cases where there is no evidence of disseminated or invasive aspergillosis, such as in intravenous drug abusers. A better choice to supplement the local ocular treatment is itraconazole. At least two reports of successfully treated endogenous Aspergillus endophthalmitis without the use of systemic antifungal therapy have been reported.183,207

When there is evidence of active systemic Aspergillus infection, intravenous amphotericin B is usually indicated, either alone or in combination with other antifungal agents.205,208 The role of itraconazole in this situation is controversial. The presence of a localized Aspergillus chorioretinitis without significant vitreous involvement is rare, and one can only speculate on the optimal treatment in this situation. Systemic treatment with amphotericin B or itraconazole may be effective in these relatively rare cases. There are previously reported cases in which one eye had significant vitreous involvement and was treated with vitrectomy and intravitreal amphotericin B, and the fellow eye had only a chorioretinitis and responded to systemic amphotericin B alone.179 All patients suspected of having endogenous Aspergillus endophthalmitis should be thoroughly evaluated for evidence of systemic aspergillosis in cooperation with an internist or infectious disease subspecialist.

Back to Top
ENDOGENOUS CRYPTOCOCCUS ENDOPHTHALMITIS
Cryptococcosis is caused by Cryptococcus neoformans, a round to oval yeastlike fungus. The genus Cryptococcus contains several species, but only C. neoformans is reported with human disease. It is not endemic to any particular area and has been found throughout the world. Pigeons are the chief vector for the organism, and it can survive for 2 or more years in pigeon excretions.

C. neoformans can be identified by direct examination using a wet-mount preparation mixed with a drop of India ink. This technique clearly outlines the characteristic mucoid capsule surrounding the organism. The organism can also be identified in stained histopathologic specimens. The most reliable test for identifying the organism is culture. The organism grows well on Sabouraud's glucose agar within 24 to 48 hours of inoculation.209,210 Rapid tests for detection from sterile fluid include latex agglutination and various serologic tests.

C. neoformans has been isolated from milk, fruit, and the atmosphere. The primary mode of human infection is through the respiratory tract. This can result in an asymptomatic pneumonitis or can lead to a severe disseminated infection involving any part of the body, with a predilection for the brain and meninges. Intraocular infection may occur by direct extension from the central nervous system (CNS) along the meningeal sheath of the optic nerve or by hematogenous spread from either a localized or disseminated cryptococcal infection.

Associated systemic conditions include lymphoma, systemic lupus erythematosus in addition to other collagen vascular diseases, sarcoidosis, tuberculosis, diabetes mellitus, drug abuse, Cushing syndrome, and pregnancy. Organ transplant recipients have a 2% to 3% infection rate, and approximately 10% of patients with AIDS are reported to have some form of cryptococcal infection.211–216 Many patients with ocular cryptococcal infection have evidence of cryptococcal infection elsewhere in the body, although cases without extraocular involvement have been reported.217,218 In many patients, no known predisposing systemic risk factors can be identified.219–225

OCULAR MANIFESTATIONS

Up to 40% of patients with CNS involvement have ophthalmic complications, including papilledema, optic atrophy, extraocular muscle paresis, chorioretinitis and endophthalmitis.

The symptoms of intraocular cryptococcal infection are nonspecific and most commonly include blurred vision, pain, photophobia, floaters, and redness. The ocular findings may also be nonspecific and vary with the severity and location of the intraocular infection. They are frequently confined to the fundus. There may be a mild anterior segment inflammatory reaction, including conjunctival and episcleral injection and anterior chamber cells with large “mutton fat” keratic precipitates. A patient who had an isolated iris mass has been reported.226 There is often a vitritis that may range from a few cells to marked inflammation sufficient to obscure fundus detail. Fundus findings include intraretinal hemorrhage, vascular sheathing, nerve fiber layer infarcts, optic nerve edema, optic atrophy, glistening retinal and preretinal lesions, and a yellow-white chorioretinal mass or scarlike lesion. Retinal detachment may complicate the infection. The symptoms and signs may be present for only a few weeks or for several years. The infection has been misdiagnosed as sterile uveitis.216

DIAGNOSIS

In addition to observing the organism directly on stained smears and culturing it, various serologic tests, including complement fixation, tube agglutination, immunodiffusion, and an indirect immunofluorescent test, can be used. The most commonly used test for detecting the disease in body fluids is the latex agglutination for antigen detection of polysaccharides on cryptococcal capsules. A titer of greater than 1:8 is significant.227

The diagnosis of endogenous cryptococcal endophthalmitis can be difficult to make in the presence of nonspecific ocular inflammatory signs. Any patient with known predisposing conditions with ocular inflammatory disease of unknown etiology not responding to local anti-inflammatory therapy should be considered suspect for cryptococcal infection. Suspicion of infection should be greater in the presence of a yellow or white chorioretinal abscess. When cryptococcal infection is known to be present elsewhere in the body, a presumptive diagnosis of ocular involvement may cautiously be made. In the absence of known systemic infection or if the ocular infection does not respond to ongoing therapy for systemic cryptococcal infection, more specific ocular diagnostic techniques should be considered, including diagnostic vitrectomy and in some cases biopsy of the chorioretinal abscess using needle-aspiration techniques previously described.228

TREATMENT

Because of the relative rarity of ocular cryptococcal infection, there are no well-established guidelines for treatment. The treatment of systemic infection has evolved during the past several years with the addition of new antifungal agents. A previously described treatment for systemic cryptococcal infection includes amphotericin B with or without flucytosine.229–232 Results from clinical trials indicate that both fluconazole and itraconazole may be as effective as amphotericin B.217,233 They both have the advantage of reduced systemic toxicity and better intraocular penetration. Fluconazole has the overall best intraocular penetration and for this reason may be the ideal systemic drug when there is intraocular infection. The cure rate for cryptococcal meningitis using itraconazole is 65%. There is a 25% partial cure rate (persistent positive cultures but symptoms improve) and a 10% failure rate. The systemic treatment is required for several months. The median time required for cerebrospinal fluid cultures to become negative in cryptococcal meningitis in AIDS patients was 42 days for patients treated with amphotericin B and 64 days for patients treated with fluconazole.233

There is limited experience using intravitreal amphotericin for the treatment of endogenous cryptococcal endophthalmitis.222–224,228,229 In one well-documented case, a 5-μg intravitreal amphotericin B injection resulted in clearing of the vitreous. The patient developed an epiretinal membrane and cataract. Persistent organisms were identified in the epiretinal membrane at the time of removal of the membrane. A second intravitreal amphotericin B injection was administered, and 4 months later the eye was free of inflammation and intraocular cultures were negative.229

Back to Top
ENDOGENOUS COCCIDIOIDES ENDOPHTHALMITIS
Coccidioides immitis is a relatively uncommon cause of intraocular infection. The organism responsible for coccidioidomycosis is a dimorphic fungus. It exists in a saprophytic phase (natural habitat) and a parasitic phase (in the host). It grows in soil and routine culture media as a saprophytic mold. Reproduction is through release of arthroconidia. In tissues and under special culture conditions, a parasitic form exists as spherules that reproduce by endospores. The hyphae consist of chains of thin-walled structures known as arthroconidia, which are easily detached and released into the atmosphere. In the host, it forms large spherules containing clear endospores, which are released when the spherule wall ruptures. The endospores can then give rise to future generations of the organism.234

The saprophytic mold fungus grows well on most routine culture media at room temperature. Definitive identification requires documenting the production of spherules at 40°C in liquid culture or in experimentally infected animals.234

The infection results from inhalation of arthroconidia. The arthroconidia are easily detached from the hyphae and released into the atmosphere. The organism is commonly found in dust. For this reason, agricultural and construction workers are particularly prone to infection. C. immitis is one of the most virulent of all fungi, and inhalation of only a few conidia in an immunocompetent host will produce infection. After exposure of the organism to the respiratory tract, the infection may result in a subclinical pneumonitis or mild to severe respiratory disease. In the immunocompetent person, it is usually self-limited and resolves in 2 to 3 weeks without significant sequelae. In unusual cases it can lead to chronic pulmonary disease and pulmonary scarring. Disseminated infection may occur in immunosuppressed patients, including AIDS patients and patients receiving steroids. The disseminated infection may result in a chronic cutaneous disease, a widespread lymphatic disease, bone involvement, visceral organ involvement, or meningitis.235–237

Coccidioidomycosis is highly endemic in the southwestern United States, northern Mexico, Central America, and several South American countries. The San Joaquin Valley is the most endemic area in the world. Since 1990, there has been a dramatic increase in cases of coccidioidomycosis documented in California. This recent increase in reported cases was related to cycles of severe aridity followed by intense rainfall. In addition, increases in construction with associated soil turnover in the area may have played a factor.238,239

DIAGNOSIS

Culture of the organism from body tissues or direct identification histopathologically is the only way to make a definitive diagnosis. In addition, a skin test using extract from the saprophytic phase of the fungus may be helpful. Serologic tests, including the tube precipitin test, latex agglutination test, immunodiffusion test, and complement fixation test, may be used.234,237,240–242

OCULAR MANIFESTATIONS

The first histologically proven case of endophthalmitis secondary to C. immitis was reported in 1948.243 Since then, several culture-positive and histopathologically proven cases have been reported.244–260 A prospective study found that 50% of patients with chronic pulmonary or disseminated coccidioidomycosis had ocular involvement.254 Because of this high incidence of ocular involvement, any patient with chronic pulmonary or systemic coccidioidomycosis should have a screening ocular examination.

The ocular involvement can range from lid lesions to panophthalmitis. The anterior segment involvement may be extraocular and include conjunctivitis, episcleritis, scleritis, or keratoconjunctivitis. Intraocular involvement may range from a chronic granulomatous iridocyclitis to a fulminant acute anterior segment inflammatory response. Posterior segment involvement includes vitritis, choroiditis, and retinitis. There is a predilection for the peripapillary choroid to be involved, and patients may have peripheral punched-out scars similar to those seen in ocular histoplasmosis. Active choroidal lesions are often white and elevated and may involve the overlying retina. Subretinal fluid and exudate may be present. Scars may be seen in asymptomatic patients with a prior history of systemic coccidioidomycosis. In one study, 5 of 54 patients with documented systemic coccidioidomycosis had inactive peripheral chorioretinal scars.257

TREATMENT

There is limited information regarding the treatment of ocular C. immitis infection.258–260 The organism usually responds to amphotericin B, although failures have occurred, both in the treatment of systemic infection and ocular infection.234,237,258–260 Intravenous miconazole has been used in cases of systemic coccidioidomycosis with ocular involvement, but the recurrence rate is high, and in one case the chorioretinitis developed while the patient was receiving the drug; in this case the infection subsequently responded to intravenous amphotericin B.255 Ketoconazole has been used in the treatment of systemic infection, but recurrence rates as high as 25% have been reported.261,262 In one study of 112 patients with nonmeningeal coccidioidomycosis, a success rate of 23% was obtained using 400 mg ketoconazole daily and 32% using 800 mg daily.262 Fluconazole and itraconazole appear to be effective for systemic coccidioidomycosis; however, their efficacy for ocular involvement is unproven.263,264

It is important to maintain a high index of suspicion for C. immitis infection in any patient with ocular inflammation of unknown etiology living in or traveling to an endemic area. This is particularly true for patients who have previous exposure to the organism. Ocular involvement has been reported as late as 22 years after primary systemic infection.258

Back to Top
ENDOGENOUS SPOROTHRIX SCHENCKII ENDOPHTHALMITIS
Sporothrix schenckii is a relatively rare but well-documented cause of endogenous endophthalmitis. The organism is a dimorphic fungus existing in both a yeast and mold phase. In the environment and culture at 25°C, it exists as a conidia-producing hyphal mold. The hyphae produce oval conidia. When the organism is introduced into animals or cultured at 37°C, the organism grows as a cigar-shaped yeast.2

Demonstration of S. schenckii is necessary for definitive diagnosis of this infection. Culture recovery is the best way of identifying the organism, but it can also be found using periodic acid-Schiff and Gomori stains of biopsy tissue. Immunochemical staining of biopsy specimens may be useful when other stains and cultures are negative and a definitive diagnosis is necessary for treatment. Identification of the antibodies to the organism is helpful when there is systemic involvement.265

Sporotrichosis can be either a subacute or chronic fungal disease. It occurs in four major forms: cutaneous or lymphocutaneous (most common), pulmonary disease, osteoarticular disease, and disseminated infection. Disseminated infection usually occurs in immunocompromised persons, including AIDS patients. Infection usually results from a skin puncture by material containing the organism. The organism is most commonly found in soil and decaying vegetation introduced into the body by pricks of thorns or barbs and can be initially manifested as a skin nodule. A less common means of acquiring the organism is by inhalation.266–273 It is an occupational hazard for farmers, gardeners, and horticulturists.

OCULAR MANIFESTATIONS

The first case of intraocular S. schenckii was reported in 1909.274 There are fewer than 20 reported cases of endogenous S. schenckii endophthalmitis in the literature.274–280 The ocular infection has been reported in immunocompromised patients with disseminated sporotrichosis as well as in otherwise healthy persons with no evidence of extraocular involvement. Any patient with known extraocular S. schenckii infection and ocular inflammatory disease should be suspected of having endogenous S. schenckii infection. Ocular involvement in sporotrichosis includes orbital, eyelid or lacrimal apparatus lesions, keratitis, scleritis, and endophthalmitis. The ocular findings vary depending on whether the anterior segment or posterior segment is primarily involved. Most of the reported patients had primarily anterior segment inflammation initially, which often progressed to endophthalmitis or panophthalmitis. The presenting ocular symptoms are nonspecific and include decreased vision, redness, and pain. The anterior segment inflammation may vary in severity and occasionally is present as a hypopyon. The uveitis may appear granulomatous or nongranulomatous. White iris nodules have been observed clinically. Posterior synechiae, glaucoma, and cataracts may develop as a result of chronic uveitis. Posterior segment involvement includes vitritis as well as chorioretinitis. The posterior segment lesion has been reported to be a fluffy white necrotic retinal lesion with overlying vitreous haze. This lesion can rapidly progress to panophthalmitis. It is important to recognize the ocular inflammatory disease as a potential infectious process and not treat solely with corticosteroid therapy, because this can cause a rapid exacerbation.

TREATMENT

In previously reported cases of endogenous S. schenckii endophthalmitis, treatment results have been disappointing and salvage of useful vision has not been reported. An AIDS patient with disseminated S. schenckii infection developed a rapidly progressive endophthalmitis. This progressed despite intravenous amphotericin B therapy, and the patient was subsequently treated with intravitreal amphotericin B injections (total of 25 μg). Despite treatment, the ocular inflammatory response persisted and the eye was enucleated. The vitreous culture obtained before enucleation was negative, and electron microscope examination of the anterior chamber demonstrated disorganization of the organisms, suggesting the treatment was effective in killing the organism.279

Antifungal agents used to treat sporotrichosis include saturated solution of potassium iodide (SSKI), 5-fluorocytosine, amphotericin B, ketoconazole, fluconazole, and itraconazole. There are no comparative studies of treatment with various agents available to determine the antifungal agent of choice. SSKI is effective, but unfortunately it is poorly accepted by most patients because of side effects, including increased lacrimation, increased salivation, salivary gland swelling, gastrointestinal upset, and rash. Ketoconazole (200 to 800 mg/day) has been used for lymphocutaneous and systemic sporotrichosis but has largely been replaced by itraconazole. This remains the drug of choice at a recommended dose of 200 mg orally twice a day. In patients whose disease is unresponsive to itraconazole, or in the presence of meningitis, amphotericin B is recommended at a total dose of 2 g. There is no reported clinical experience using itraconazole for intraocular sporothrix infection. After systemic administration, itraconazole has an advantage over amphotericin B of better intraocular penetration.281–284

The treatment of choice for endogenous S. schenckii endophthalmitis, particularly when there is significant vitreous involvement, is a combination of vitrectomy and intravitreal amphotericin B injection.

Back to Top
ENDOGENOUS BLASTOMYCES DERMATITIDIS ENDOPHTHALMITIS
Blastomyces dermatitidis is an infection caused by a dimorphic fungus considered a saprophyte in nature. Gilchrist first identified the organism in 1894.285 At room temperature it grows as a spore-producing mold. Inhalation of the spores is the most common route of human clinical infection. When the organism gains access to the body or when it is incubated at 37°C, it converts to the yeast form, which reproduces by budding. The bud has a characteristic broad base of attachment to the parent cell. This characteristic feature is helpful in positive identification of the organism. The organism can be identified using 10% KOH as a wet-prep specimen or using a fixed specimen stained with hematoxylin and eosin, periodic acid-Schiff, or Gomori's methenamine silver stains. The organism grows well on blood and Sabouraud's agar at 25°C. Because the growth rate is slow, cultures should be kept at least 2 months in suspected cases. Positive identification from culture is obtained by examining a subculture of the specimen grown at 37°C, demonstrating the characteristic yeast form.2

The organism is endemic to parts of the midwestern and south-central United States and Canada, but blastomycosis has been reported worldwide. In the United States the disease is concentrated in an area along the Mississippi and Ohio River basins and around the Great Lakes.2,286

Because the organism usually obtains access to the body through inhalation, the most common form of clinical infection is pulmonary. This can range from an asymptomatic or self-limited flulike illness to a chronic pneumonia characterized by cough, chest pain, hemoptysis, weight loss, and low-grade fever. Hematogenous dissemination from the lung can occur, resulting in extrapulmonary infection, including skin infection in 40% to 80% of cases and multiple organ involvement, including the eye in approximately 50% to 60% of cases. After the lung and skin, other sites of involvement include bones and joints as well as the male genitourinary system. CNS involvement is uncommon. It is an uncommon opportunistic pathogen but may cause severe disseminated infection in the immunocompromised host.286–289

OCULAR MANIFESTATIONS

The spectrum of ocular disease includes eyelid lesions, keratitis, choroiditis, endophthalmitis, and panophthalmitis.290–299 Orbital blastomycoses has also been reported.299,300 In most cases, the ocular involvement is associated with obvious systemic disease, but cases of ocular involvement with no evidence of systemic disease have been reported.

There are at least 11 reported cases of intraocular blastomycosis in the literature.290–300 Six of these cases were verified by histologic examination of the enucleated globe and three were presumed diagnoses based on the presence of systemic blastomycosis.

Ocular symptoms are nonspecific and depend on the site of ocular involvement. They include pain, redness, photophobia, and blurred vision. The onset may be subacute or acute. A pale-yellow elevated choroidal lesion appears to be characteristic of B. dermatitidis choroiditis. The choroidal lesions may be multiple or solitary and associated with hemorrhage and periphlebitis. Lesions can be unilateral or bilateral.

TREATMENT

The three drugs approved for the treatment of blastomycoses are amphotericin B, itraconazole, and ketoconazole.288,289,301,302 Previously reported treatment for systemic disease has been amphotericin B, and this continues to be the preferred treatment when there is CNS involvement. Itraconazole has been gaining increased acceptance for non-CNS disease.302 Its efficacy approximates that of amphotericin B. Ketoconazole is less efficacious but was shown to be effective in approximately 80% of cases in one series.301 Ketoconazole has the advantage of being less expensive, and because treatment needs to be continued with either oral agent for at least 6 months, this may be an important consideration in infections that are responsive to ketoconazole. Amphotericin B is generally reserved for patients with disseminated life-threatening disease, particularly in immunocompromised patients, patients with CNS disease, or patients in whom oral therapy has failed. The total recommended dose of amphotericin B is 1.5 to 2.5 g. Itraconazole can be started at 200 mg/day and advanced by 100-mg increments at monthly intervals to a maximum of 400 mg/day. Ketoconazole can be initiated at 400 mg/day and advanced by 200-mg increments monthly to 800 mg/day as needed.

Patients with systemic disease associated with an isolated choroiditis can probably be successfully treated with an oral agent, preferably itraconazole. When there is significant vitreous involvement, pars plana vitrectomy combined with intravitreal amphotericin B should be considered. This should be supplemented with oral itraconazole for either documented or presumed systemic infection.

Back to Top
OCULAR HISTOPLASMOSIS
Histoplasma capsulatum, the organism responsible for histoplasmosis, is a dimorphic fungus. In the environment it grows as a mold, producing small infective spores ranging from 2 to 5 microns. The organism is found in soil rich in nitrogen; this explains the frequent occurrence of histoplasmosis in association with chicken coops, bird roosts, and bat caves. Although the organism can be found worldwide, the most endemic area of the United States is the Ohio/Mississippi Valley. This includes Missouri, Kentucky, Tennessee, southern Illinois, Indiana, and Ohio.2,234,303

When the spores are disturbed, they become airborne. Human infection results from inhalation of these infectious particles. When they reach the alveoli, they convert to small yeast, which represents the tissue-invasive form. Once the organism gains access to the pulmonary alveoli, it travels to organs rich in cells of the reticuloendothelial system before immunity to the organism develops. After development of adequate cell-mediated immunity, macrophages kill the fungi in most immunocompetent persons. Greater than 90% of infected persons are asymptomatic, and the only evidence of previous infection is conversion of the histoplasmin skin test to positive. A few patients who inhale the infecting particles become acutely ill. In immunocompetent persons, this is usually an influenza-like illness with fever, chills, and nonproductive cough. It may be associated with erythema multiforme or erythema nodosum. If the infective dose is extremely high, patients may experience a fulminant pneumonitis with secondary respiratory compromise. Most immunocompetent patients with pulmonary histoplasmosis recover completely, but they are often left with pulmonary scars. The characteristic scar is a round fibrotic lesion with associated calcification (coin lesions). These pulmonary scars can often be found in patients living in areas endemic for histoplasmosis.303

A progressive, disseminated form of histoplasmosis can develop in patients with deficient cell-mediated immunity. This usually leads to a rapidly progressive wasting systemic illness. HIV-infected persons living in endemic areas are at greatest risk for this form of histoplasmosis.2,234,304,305

OCULAR MANIFESTATIONS

Three main forms of ocular involvement by H. capsulatum have been identified in humans306–315: disseminated histoplasmosis with uveal, retinal, or optic nerve involvement; solitary histoplasmic chorioretinal granuloma; and the presumed ocular histoplasmosis syndrome. The latter syndrome is presumably due to histoplasmosis, although no organisms have been convincingly identified in the eye in most of these patients.314 The classic features of the presumed ocular histoplasmosis syndrome include peripapillary scars in combination with macular and peripheral pigmented or atrophic punctate scars. The major significance of these lesions is the later development of choroidal neovascularization resulting from macular scars. The findings are often picked up on routine fundus examination, and these patients are usually asymptomatic until they develop choroidal neovascularization.314 Occasionally these patients also develop what appears to be an active focal choroiditis that in some cases can be difficult to distinguish from a choroidal neovascular membrane clinically or on fluorescein angiography. The histoplasmin skin test is often positive in these patients. The scars in these patients are presumed to be due to an acute choroidal infection with the organism, but this is rarely identified.314,315 One report of an acute histoplasmosis choroiditis in two immunocompetent brothers enabled a rare opportunity to observe the acute form of what is believed to result in the typical presumed ocular histoplasmosis syndrome lesions.315

Endogenous histoplasmic endophthalmitis has been reported in several patients with disseminated systemic histoplasmosis.306–309 Most of the previously reported cases were in patients with AIDS.306,307 The ocular infection may result in a granulomatous anterior uveitis. Vitritis may be mild to severe. The classic fundus lesion is a cream-colored or gray-white chorioretinal infiltrate, often surrounded by hemorrhage. There may be associated retinal pigment epithelial alteration, indicating partial spontaneous healing. The lesions may be solitary or multiple and can range from less than a disc diameter to several disc diameters. Histoplasmic optic neuritis has also been reported in association with the chorioretinitis. The ocular findings are not specific for histoplasmic endophthalmitis, and the diagnosis should be suspected in any immunosuppressed patient living in an area endemic for histoplasmosis. Most patients have clinical and laboratory evidence of disseminated infection, in which case the ocular infection can be presumed to be due to histoplasmosis based on the known systemic infection. When the ocular infection is uncertain and there is significant vitreous involvement, a diagnostic vitrectomy should be considered. When there is no significant vitritis in a patient with choroidal infiltrates of unknown cause, chorioretinal biopsy can be considered, although this can be associated with significant risk. This is probably not indicated when the diagnosis is suspected based on known systemic histoplasmosis.

Because culturing the organism can be difficult, the diagnosis of histoplasmosis is often made by pseudodiagnosis. A complement fixation test becomes positive 3 weeks after the onset of infection, and by 6 weeks a diagnostic titer may be obtained in up to 75% of patients. A Histoplasma polysaccharide antigen test is useful in the diagnosis of histoplasmosis in HIV-infected patients where the complement of organisms is high. The test is not sensitive for patients with low organism counts. The histoplasmin skin test is useful for determining whether immunocompetent patients have been exposed to the organism.316

TREATMENT

As with other intraocular fungal infections, the systemic antifungal therapy is determined by the presence and extent of systemic involvement. Most acutely infected immunocompetent patients with histoplasmosis require no treatment. In the unusual situation where there is severe pulmonary infection, intravenous amphotericin B is initiated at a dose of 500 mg in 50-mg daily doses. Once the patient is stable, this can be switched to itraconazole 200 mg twice daily.317 Chronic pulmonary histoplasmosis in immunocompetent patients can be treated with either ketoconazole 400 to 800 mg/day318 or itraconazole 200 to 400 mg twice daily. In the case of failure to respond to this oral therapy, intravenous amphotericin B can be considered. For progressive disseminated infection, amphotericin B is the initial treatment of choice.319 Once stable, the patient can be switched to itraconazole 200 to 400 mg twice daily.

There is limited clinical experience in the treatment of endogenous ocular histoplasmic infection. When the infection is limited to the choroid or retina, systemic therapy alone with amphotericin B, ketoconazole, or itraconazole may be curative. If there is significant vitreous involvement, local ocular therapy, including vitrectomy and intravitreal amphotericin B, can be considered. This local ocular therapy should probably be combined with systemic antifungal therapy, depending on the presence and extent of systemic infection.

Back to Top
RARE CAUSES OF ENDOGENOUS FUNGAL ENDOPHTHALMITIS
Other fungi reported to cause endogenous fungal endophthalmitis include P. boydii,320–322 Fusarium species,323–328 Mucormycosis,329 Paecilomyces species,330–332 Pencillium species,333 Bipolaris hawaiiensis,334 and Trichosporon beigelii.335 Because of the rarity of these causes of endogenous fungal endophthalmitis, they will not be reviewed in detail in this chapter, and the reader is referred to individual case reports.

Because Nocardia asteroides can clinically and histopathologically resemble endogenous fungal eye infection, it should be considered in the differential diagnosis of endogenous fungal endophthalmitis.336–340 The organisms resemble fungi microscopically and appear as delicate intertwining, branched hyphae. Nocardia are slow-growing soil-borne filamentous bacteria. They usually enter the human body through the respiratory tract. Clinical infection is usually seen in immunosuppressed patients, particularly when cell-mediated immunity is impaired. Hematogenous dissemination occurs after the initial pneumonitis in about half of patients. The infection can spread to virtually every organ in the body, but the CNS is the most common site. Ocular infection is uncommon, but at least 14 cases have been reported.336–341 The characteristic fundus lesion is a discrete white subretinal mass with overlying subretinal fluid and hemorrhage. A subretinal pseudohypopyon similar to that seen in patients with Aspergillus endophthalmitis has been reported.336

Nocardia responds well to sulfonamides, and sulfadiazine 6 to 8 g/day is typically used in adults. Trimethoprim-sulfamethoxazole may be substituted as an alternative first-line agent. Minocycline, amikacin, and ampicillin have also been successful in treating patients without brain abscess or patients who fail to respond to the sulfonamides. Treatment usually needs to be continued for several months.340

Back to Top
CONCLUSION
Endogenous fungal endophthalmitis should be considered in the differential diagnosis of progressive intraocular inflammation of unknown cause in persons predisposed to systemic fungal infection. Persons at higher risk include immunocompromised and debilitated patients as well as intravenous drug abusers. Candida infection should also be suspected in patients who have undergone recent gastrointestinal surgery, or in the presence of indwelling venous catheters and prolonged antibiotic therapy. Because certain fungi, such as C. immitis and H. capsulatum, are more common in specific areas of the country, a higher index of suspicion for these infections should be maintained when patients live in or have recently visited endemic areas.

In some cases of endogenous fungal endophthalmitis, the diagnosis is made based on the presence of known systemic infection; in other cases, the diagnosis can be determined only by evaluating intraocular fluids. The eye may be the organ most accessible to obtaining infected material, which may be useful in determining the cause of an unknown systemic infection.

In many cases, the treatment of endogenous fungal endophthalmitis depends on the extent of ocular involvement. When the infection is isolated to the choroid and retina, systemic treatment alone may be curative. When there is vitreous involvement, a vitrectomy and injection of amphotericin B can be considered. In these cases, the systemic treatment is guided by the extent of systemic disease, and treatment should be initiated in conjunction with an infectious disease specialist. Newer, less toxic antifungal medications may improve the outcomes of treatment and reduce potential treatment complications.

Back to Top
REFERENCES

1. Essman TF, Flynn HW Jr, Smiddy WE et al: Treatment outcomes in a 10-year study of endogenous fungal endophthalmitis. Ophthalmic Surg Lasers 28:185, 1997

2. Walsh TJ: Emerging fungal pathogens: Evolving challenges to immunocompromised patients. In Scheld WM, Armstrong D, Hughes JM (eds): Emerging Infections 1, chap. 15. Washington DC: ASM Press, 1998

3. Kresloff MS, Castellarin AA, Zarbin MA: Endophthalmitis. Surv Ophthalmol 43:193, 1998

4. Martinez-Vazquez C, Fernandez-Ulloa J, Bordon J et al: Candida albicans endophthalmitis in brown heroin addicts: Response to early vitrectomy preceded and followed by antifungal therapy. Clin Infect Dis 27:1130, 1998

5. Getnick RA, Rodrigues MM: Endogenous fungal endophthalmitis in a drug addict. Am J Ophthalmol 77:680, 1974

6. Shankland GS, Richardson MD: Source of infection in Candida endophthalmitis in drug addicts. Br Med J (Clin Res) 292:1106, 1986

7. Servant JB, Dutton GN, Ong-Tone L et al: Candidal endophthalmitis in Glaswegian heroin addicts: Report of an epidemic. Trans Ophthalmol Soc UK 104:297, 1985

8. Hutchins GP: Fusarium endophthalmitis in an intravenous drug abuser. Am J Ophthalmol 122:119, 1996

9. Heinemann MH, Bloom AF, Horowitz J: Candida albicans endophthalmitis in a patient with AIDS: Case report. Arch Ophthalmol 105:1172, 1987

10. Denning DW, Armstrong RW, Fishman M, Stevens DA: Endophthalmitis in a patient with cryptococcosis and AIDS who was treated with itraconazole. Rev Infect Dis 13: 126, 1991

11. Kirkpatrick CH: Host factors in defense against fungal infections. Am J Med 77(Suppl 4D):1, 1984

12. Behlau I, Baker AS: Fungal infections of the eye. In Albert DA, Jakobiec F (eds): Principles and Practice of Ophthalmology, pp 4884–4917. Philadelphia: WB Saunders, 2000 13. Gass JDM: Stereoscopic Atlas of Macular Diseases: Diagnosis and Treatment, pp 601–736. St Louis: Mosby, 1997

13. Gass JDM:Stereoscopic Atlas of Mascular Diseases: Diagnosis and Treatment, pp 601–736. St Louis: Mosby. 1997

14. Regillo CD, Brown GC, Flynn HW Jr (eds): Vitreoretinal Disease: The Essentials, pp 393–438. New York: Thieme, 1999

15. Louria DB, Dincen P: Amphotericin B in the treatment of disseminated moniliasis. JAMA 174:273, 1960

16. Gold W, Stout HA, Pagano JF, Danovick R: Amphotericins A and B: antifungal antibiotics produced by a streptomycete: I. In vitro studies. In Welch H, Marti-Ibanez F (eds): Antibiotics Annual, 1955–1956, p 579. New York: Medical Encyclopedia, 1956

17. Butler WT: Pharmacology, toxicity and therapeutic usefulness of amphotericin B. JAMA 195:371, 1966

18. Kotler-Brajtburg J, Price HD, Medoff G et al: Molecular basis for the selective toxicity of amphotericin B for yeast and filipin for animal cells. Antimicrob Agents Chemother 5:377, 1974

19. Bennett JE: Chemotherapy of systemic mycoses. N Engl J Med 290, 1974

20. Green WR, Bennett JE, Goos RD: Ocular penetration of amphotericin B: A report of laboratory studies and a case report of post surgical cephalosporium endophthalmitis. Arch Ophthalmol 73:769, 1965

21. Stern WH, Tamura E, Jacobs RA et al: Epidemic post surgical Candida parapsilosis endophthalmitis: Clinical findings and management of 15 consecutive cases. Ophthalmology 92:1701, 1985

22. Fisher JF, Taylor AT, Clark J et al: Penetration of amphotericin B into the human eye. J Infect Dis 147:164, 1983

23. O'Day DM, Head WS, Robinson RD et al: Intraocular penetration of systemically administered antifungal agents. Curr Eye Res 4:131, 1985 (published erratum appears in Curr Eye Res 5:547, 1986)

24. Barza M: Editorial response: Treatment options for candidal endophthalmitis. CID 27:1134, 1998

25. Axelrod AJ, Peyman GA, Apple DJ: Toxicity of intravitreal injection of amphotericin B. Am J Ophthalmol 76:578, 1973

26. Tremblay C, Barza M, Szoka F et al: Reduced toxicity of liposome-associated amphotericin B injected intravitreally in rabbits. Invest Ophthalmol Vis Sci 26:711, 1985

27. McGetrick JJ, Peyman GA, Nyberg MA: Amphotericin B methyl ester: Evaluation for intravitreous use in experimental fungal endophthalmitis. Ophthalmic Surg 10:25, 1979

28. Barza M Baum J, Tremblay C et al: Ocular toxicity of intravitreally injected liposomal amphotericin B in rhesus monkey. Am J Ophthalmol 100:259, 1985

29. Axelrod AJ, Peyman GA: Intravitreal amphotericin B treatment of experimental fungal endophthalmitis. Am J Ophthalmol 76:584, 1973

30. Doft BH, Weiskopf J, Nilsson-Ehle I, Wingard LB Jr: Amphotericin clearance in vitrectomized versus nonvitrectomized eyes. Ophthalmology 92:1601, 1985

31. Wingard LB Jr, Zurlaveff J, Doft B et al: Intraocular distribution of intravitreally administered amphotericin B in normal and vitrectomized eyes. Invest Ophthalmol Vis Sci 30:2184, 1989

32. Perraut LE Jr, Perraut LE, Bleiman B, Lyons J: Successful treatment of Candida albicans endophthalmitis with intravitreal amphotericin. Br Arch Ophthalmol 99:1565, 1981

33. Stern WH, Tamura E, Jacobs RA et al: Epidemic post surgical Candida parapsilosis endophthalmitis: Clinical findings and management of 15 consecutive cases. Ophthalmology 92:1701, 1985

34. Minogue MJ, Playfair TJ, Gregory-Roberts JC, Robinson LP: Cure of Paecilomyces endophthalmitis with multiple intravitreal injections of amphotericin B: Case report. Arch Ophthalmol 107:1281, 1989

35. Huang K, Peyman GA, McGetrick J: Vitrectomy in experimental endophthalmitis: I. Fungal infections. Ophthalmic Surg 10:84, 1979

36. Stern GA, Fetkenhour CL, O'Grady RB: Intravitreal amphotericin B treatment of Candida endophthalmitis. Arch Ophthalmol 95:89, 1977

37. Snip RC, Michels RG: Pars plana vitrectomy in the management of endogenous Candida endophthalmitis. Am J Ophthalmol 82:699, 1976

38. Salmon JF, Patridge BM, Spalton DJ: Candida endophthalmitis in a heroin addict: A case report. Br J Ophthalmol 67:306, 1983

39. Kroll P, Emmerich KH, Fegeler W: Candida albicans endophthalmitis: Ergebnisse de Pars Plana Vitrektomie ohne intraokulare antimymkotische Therapie. Klin Monatsbl Augenheilkd 184:104, 1984

40. Furia M, Parent de Curzon H, Campinchi R: Evolution favorable d'une endophthalmie bilaterale a Candida albicans: Interet de la vitrectomie precoce. J Fr Ophthalmol 7:689, 1984

41. Mersch-Ausset MH, Salvanet-Bouchara A, Forestier F et al: Etude angiographique et vitrectomie diagnostique et therapeutique au cors d'une endoptalmie a Candida albicans chez une heroinomane. Bull Soc Ophtalmol Fr 86:19, 1986

42. Barrie T: The place of elective vitrectomy in the management of patients with Candida endophthalmitis. Graefes Arch Clin Exp Ophthalmol 225:107, 1987

43. Dunn ET, Mansour AM: Retinal striae as a sign of resolving Candida chorioretinitis. Graefes Arch Clin Exp Ophthalmol 226:591, 1988

44. Rennie CJ, Johnson J, Farah S, Hellman L: Disseminated candidiasis due to amphotericin B-resistant Candida albicans. J Infect Dis 165:761, 1992

45. Hamory JN: Endophthalmitis caused by non-albicans Candida. Rev Infect Dis 13:281, 1991

46. Edwards JE Jr, Montgomerie JZ, Ishida K et al: Experimental hematogenous endophthalmitis due to Candida: Species variation in ocular pathogenicity. J Infect Dis 155:294, 1977

47. McQuillen DP, Zingman BS, Meunier F, Levitz SM: Invasive fungal infections due to Candida krusei: Report of 10 cases of fungemia that include three cases of endophthalmitis. Clin Infect Dis 14:472, 1992

48. Edwards JE Jr, Montgomerie JZ, Ishida K et al: Experimental hematogenous endophthalmitis due to Candida: Species variation in ocular pathogenicity. J Infect Dis 135:294, 1977

49. Tassel D, Madoff MA: Treatment of Candida sepsis and Cryptococcus meningitis with 5-fluorocytosine: A new antifungal agent. JAMA 206:830, 1968

50. Diasio RB. Bennett JE, Myers CE: Mode of action of 5-fluorocytosine. Biochem Pharmacol 27:703, 1978

51. Walsh A, Haft DA, Miller MH et al: Ocular penetration of 5-fluorocytosine. Invest Ophthalmol Vis Sci 17:691, 1978

52. Drouhet E, Mercier-Soucy L, Montplaisir S: Sensibilite et resistance des levures pathogenes aux 5-fluoropyrimidines: I. Relation entre les phenotypes de resistance a la 5-fluorocytosine, le serotype de Candida albicans, et l'ecologie de differentes especes de Candida d'origine humaine. Ann Microbiol (Paris) 126B:25, 1975

53. Shadomy S: Further in vitro studies with 5-fluorocytosine, Infect Immun 2:484, 1970

54. Medoff G, Comfort M, Kobayashi GS: Synergistic action of amphotericin B and 5-fluorocytosine against yeast-like organisms. Proc Soc Exp Biol Med 138:571, 1971

55. Montgomerie JZ, Edwards JE Jr, Guze LB: Synergism of amphotericin B and 5-fluorocytosine for Candida species. J Infect Dis 132:82, 1975

56. Cohen M, Montgomerie JZ: Hematogenous endophthalmitis due to Candida tropicalis: Report of two cases and review. Clin Infect Dis 17:270, 1993

57. Yoshizumi MO, Silverman C: Experimental intravitreal 5-fluorocytosine. Ann Ophthalmol 17:58, 1985

58. Dixon D, Shadomy S, Shadomy JH et al: Comparison of the in vitro antifungal activities of miconazole and a new imidazole R 41, 400. J Infect Dis 138:245, 1978

59. Jodan WM, Bodey GP, Ridriguez V et al: Miconazole therapy for treatment of fungal infections in cancer patients. Antimicrob Agents Chemother 16:792, 1979

60. Blumenkranz MD, Stevens DA: Therapy of endogenous fungal endophthalmitis: Miconazole or amphotericin B for coccidioidal and candidal infection. Arch Ophthalmol 98: 1216, 1980

61. Gallo J, Grunstein H, Clifton-Bligh et al: Miconazole in fungal endophthalmitis. Lancet 1:53, 1982

62. Fainstein V, Bodey GP: Cardiorespiratory toxicity due to miconazole. Ann Intern Med 93:432, 1980

63. Niell HB: Miconazole carrier solution, hyperlipidemia and hematologic problems. N Engl J Med 296:1479, 1977

64. Lutwick KI, Galgiani JN, Johnson RH, Stevens DA: Visceral fungal infections due to Petriellidium boydii (Allescheria boydii): In vitro drug sensitivity studies. Am J Med 61:632, 1976

65. Jones DB: Therapy of postsurgical fungal endophthalmitis. Ophthalmology 85:357, 1978

66. Tolentino FI, Foster CS, Lahav M et al: Toxicity of intravitreous miconazole. Arch Ophthalmol 100:1504, 1982

67. Ketoconazole approved for systemic fungal infections. FDA Drug Bull 11:18, 1981

68. Borgers M, Van den Bossche H, DeBrabander M: The mechanism of action of the new antimycotic ketoconazole. Am J Med 74:2, 1983

69. Utz JP, Drouhet E: Treatment of Candida infections. In Bodey GP, Fainstein V (eds): Candidiasis, p 261. New York: Raven Press, 1985

70. Jansen PAJ, Symoens JE: Hepatic reactions during ketoconazole treatment. Am J Med 74:80, 1983

71. Lewis JH, Zimmerman HJ, Benson GD, Ishak KG: Hepatic injury associated with ketoconazole therapy: Analysis of 33 cases. Gastroenterology 86:503, 1984

72. Grosso DS, Boyden TW, Pamenter RW et al: Ketoconazole inhibition of testicular secretion of testosterone and displacement of steroid hormones from serum transport proteins. Antimicrob Agents Chemother 23:207, 1983

73. VanCutsen J: The antifungal activity of ketoconazole. Am J Med 74:9, 1983

74. Drouhet E, Dupont B: Laboratory and clinical assessment of ketoconazole in deep-seated mycoses. Am J Med 74:30, 1983

75. Lechi A, Montesi G, Dean P et al: Ketoconazole: Clinical evaluation in severe fungal infections. Chemioterapia 5:18, 1986

76. Levine HB: Resistance to ketoconazole. Lancet 2:211, 1982

77. Malecaze F, Linas MD, Mathis A et al: Experimental study of intra-ocular penetration of ketoconazole in rabbits. Graefes Arch Clin Exp Ophthalmol 225:163, 1987

78. Savani DV, Perfect JR, Cobo LM, Durack DT: Penetration of new azole compounds into the eye and efficacy in experimental Candida endophthalmitis. Antimicrob Agents Chemother 31:6, 1987

79. Goodman DF, Stern WH: Oral ketoconazole and intraocular amphotericin B for treatment of postoperative Candida parapsilosis endophthalmitis. Arch Ophthalmol 105:172, 1987

80. Yoshizumi MO, Banihashemi AR: Experimental intravitreal ketoconazole in DMSO. Retina 8:210, 1988

81. Investigators' Reference Manual: Fluconazole (UK-49,858) Antifungal, Oral and Intravenous. Groton, CT: Pfizer Central Research, 1986

82. Humphrey MJ, Jevons S, Tarbit MH: Pharmacokinetic evaluation of UK-49,858, a metabolically stable triazole antifungal drug in animals and humans. Antimicrob Agents Chemother 28:648, 1985

83. Debryne D, Ryckelynck J: Pharmacokinetics of fluconazole. Clin Pharmacokinet 24:10, 1993

84. O'Day DM, Foulds G, Williams TE et al: Ocular uptake of fluconazole following oral administration. Arch Ophthalmol 108:1006, 1990

85. Mochizuki K, Yamashita Y, Torisaka T et al: Intraocular penetration and effect on the retina of fluconazole. Lens Eye Tox Res 9:537, 1992

86. Foulds G, Brennan DR, Wajszczuk C et al: Fluconazole penetration into cerebrospinal fluid in humans. J Clin Pharmacol 28:363, 1988

87. Boken DJ, Swindells S, Rinaldi MG: Fluconazole-resistant Candida albicans. Clin Infect Dis 17:1018, 1993

88. Wingard JR, Merz WG, Rinaldi MG et al: Increase in Candida krusei infection among patients with bone marrow transplantation and neutropenia treated prophylactically with fluconazole. N Engl J Med 325:1274, 1991

89. Aust R, Kruse F, Wildfever A et al: Fluconazole level in the aqueous humor after oral drug administration. Ophthalmology 92:829, 1995

90. Filler SG, Crislip MA, Mayer CL, Edwards JE Jr: Comparison of fluconazole and amphotericin B for treatment of disseminated candidiasis and endophthalmitis in rabbits. Antimicrob Agents Chemother 35:288, 1991

91. Isobe Y, Hatano H: Study of suppressive effect of intravenous fluconazole on endogenous Candida endophthalmitis in rabbits. Jpn J Ophthalmol 36:23, 1992

92. Akler ME, Vellend H, McNeely DM et al: Use of fluconazole in the treatment of candidal endophthalmitis. Clin Infect Dis 20:657, 1995

93. Christmas NJ, Smiddy WE: Vitrectomy and systemic fluconazole for treatment of endogenous fungal endophthalmitis. Ophthalmic Surg Lasers 27:1012, 1996

94. Laatikainen L, Tuominen M, von Dickhoff K: Treatment of endogenous fungal endophthalmitis with systemic fluconazole with or without vitrectomy. Am J Ophthalmol 113: 205, 1992

95. Luttrull J, Wan WL, Kubak B et al: Treatment of ocular fungal infections with oral fluconazole. Am J Ophthalmol 119:477, 1995

96. Taniguchi S, Hatano H, Isobe H, Ohno S: Eight cases of endogenous Candida endophthalmitis treated with fluconazole. Curr Aspects Ophthalmol 1254, 1992

97. Urbak SF, Degen T: Fluconazole in the treatment of Candida albicans endophthalmitis. Act Ophthalmol 70:528, 1992

98. Zarbin MA, Becker E, Witcher J et al: Treatment of fungal endophthalmitis with oral fluconazole. Ophthalmic Surg Lasers 27:628, 1996

99. Penk A, Pittrow L: Status of fluconazole in the therapy of Candida endophthalmitis. Mycoses 41(Suppl 2):41, 1998

100. Ohnishi Y, Tawara A, Murata T et al: Postmortem findings 2 weeks after oral treatment for metastatic Candida endophthalmitis with fluconazole. Ophthalmologica 213:341, 1999

101. Nomura J, Ruskin J: Failure of therapy with fluconazole for candidal endophthalmitis. Clin Infect Dis 17:888, 1993

102. Venditti M, Bernardis FD, Micozzi A et al: Fluconazole treatment of catheter-related right-sided endocarditis caused by Candida albicans and associated with endophthalmitis and folliculitis. Clin Infect Dis 14:422, 1992

103. Schulman JA, Peyman G, Fiscella R et al: Toxicity of intravitreal injection of fluconazole in the rabbit. Can J Ophthalmol 22:304, 1987

104. Su CY, Lin CP, Wang HZ et al: Intraocular use of fluconazole in the management of ocular fungal infection. Kao Hsiung I Hsueh Ko Tsa Chih 15:218, 1999

105. Gakiani JN: Fluconazole, a new antifungal agent. Ann Intern Med 113:177–179, 1990

106. van Wout JW, Novakova I, Verhagen CA et al: The efficacy of itraconazole against fungal infections in neutropenic patients: a randomized comparative study with amphotericin B. J Infect 22:45, 1991

107. Neppert B, Guthoff R, Heidemann HT: Endogenous Candida endophthalmitis in a drug-dependent patient: Intravenous therapy with liposome encapsulated amphotericin B. Klin Monatsbl Augenheilkd 201:122, 1992

108. Virata SR, Kylstra JA, Brown JC et al: Worsening of endogenous endophthalmitis during therapy with intravenous lipid complex amphotericin B. Clin Infect Dis 28:1177, 1999

109. Georgopapadakou NH, Walsh TJ: Antifungal therapy: Chemotherapeutic targets and immunologic strategies. Antimicrob Agents Chemother 40:279, 1996

110. Collignon PJ, Sorrell TC: Disseminated candidiasis: Evidence of a distinctive syndrome in heroin abusers. Br Med J (Clin Res) 287:861, 1983

111. Hogeweg M, DeJong PTVM: Candida endophthalmitis in heroin addicts. Doc Ophthalmol 55:63, 1983

112. Tarr KH: Candida endophthalmitis and drug abuse. Aust J Ophthalmol 8:303, 1980

113. Elliott JH, O'Day DM, Gutow GS et al: Mycotic endophthalmitis in drug abusers. Am J Ophthalmol 88:66, 1979

114. Aguilar GL, Blumenkranz MS, Egbert PR, McCulley JP: Candida endophthalmitis after intravenous drug abuse. Arch Ophthalmol 97:96, 1979

115. Edwards JE Jr, Foos RY, Malecaze F et al: Candida endophthalmitis after heroin abuse. Mycopathologia 92:73, 1985

116. Gallo J, Playfair J, Gregory-Roberts J et al: Fungal endophthalmitis in narcotic abusers: Medical and surgical therapy in 10 patients. Med J Aust 142:386, 1985

117. Aabel R Jr, Baster T: Candida albicans endophthalmitis due to catheter tip sepsis. Del Med J 48:121, 1976

118. Rose HD: Venous catheter-associated candidemia. Am J Med Sci 275:265, 1978

119. Graham E, Chignell AH, Eykyn S: Candida endophthalmitis: A complication of prolonged intravenous therapy and antibiotic treatment. J Infect 13:167, 1986

120. Seelig MS: The role of antibiotics in the pathogenesis of Candida infections. Am J Med 40:887, 1966

121. Kennedy MJ, Volz PA: Effect of various antibiotics on gastrointestinal colonization and dissemination by Candida albicans. Sabouraudia 23:265, 1985

122. Nahata MC, Davidorf FH, Caldwell JH et al: Candida endophthalmitis associated with total parenteral nutrition. J Parenteral Enteral Nutr 5:150, 1981

123. Dellon AL, Stark WJ, Chretien PB: Spontaneous resolution of endogenous Candida endophthalmitis complicating intravenous hyperalimentation. Am J Ophthal 79:648, 1975

124. Montgomerie JZ, Edwards JB: Association of infection due to Candida albicans with intravenous hyperalimentation. J Infect Dis 137:197, 1978

125. Henderson DK, Edwards JE Jr, Montgomerie JZ: Hematogenous Candida endophthalmitis in patients receiving parenteral hyperalimentation fluids. J Infect Dis 143:655, 1981

126. Schmid S, Marttenet AC, Oelz O: Candida endophthalmitis: Clinical presentation, treatment and outcome in 23 patients. Infection 19:21, 1991

127. Henderson T, Irfan S: Bilateral endogenous Candida endophthalmitis and chorioretinitis following toxic megacolon. Eye 10:755, 1996

128. Airas KA, Nikoskelainen J: Haematogenous Candida endophthalmitis after abdominal surgery. Acta Ophthalmol 65:450, 1987

129. Weickert CH, Richard G: Defekte der Killer-T-Lymphozyten als Ursache einer endogenous Candida albicans-Endophthalmitis. Fortscher Ophthalmol 84:350, 1987

130. Young RC, Bennett JE, Geelhoed GW, Levine AS: Fungemia with compromised host resistance: A study of 70 cases. Am Intern Med 80:605, 1974

131. Wingard JR, Merz WG, Saral R: Candida tropicalis: A major pathogen in immunocompromised patients. Ann Intern Med 91:539, 1979

132. Hawkins C, Armstrong D: Fungal infections in the immunocompromised host. Clin Haematol 13:599, 1984

133. Bodey GP: Infections in cancer patients. Cancer Treat Rev 2:89, 1975

134. Ezdinli EZ, O'Sullivan DD, Wasser LP et al: Oral amphotericin for candidiasis in patients with hematologic neoplasms. JAMA 20:258, 1979

135. Klein JJ, Watanakunakorn C: Hospital-acquired fungemia: Its natural course and clinical significance. Am J Med 67:51, 1979

136. Wade JC: Epidemiology of Candida infections. In Bodey GP (ed): Candidiasis: Pathogenesis, Diagnosis and Treatment, pp 85–107. New York, Raven Press, 1993

137. Parke DW II, Jones DB, Gentry LO: Endogenous endophthalmitis among patients with candidemia. Ophthalmology 89:789, 1982

138. Brod RD, Flynn HW Jr, Clarkson JG et al: Endogenous Candida endophthalmitis: Management without intravenous amphotericin B. Ophthalmology 96:111, 1989

139. Billson FA, Playfair TJ: The place for vitrectomy in posterior segment infection. Aust J Ophthalmol 9:21, 1981

140. Palmer EA: Endogenous Candida endophthalmitis in infants. Am J Ophthalmol 89:388, 1980

141. Speer ME, Hittner HM, Rudolph AJ: Candida endophthalmitis: A manifestation of candidiasis in the neonate. South Med J 73:1407, 1980

142. Baley JE, Annable WL, Kliegman RM: Candida endophthalmitis in the premature infant. J Pediatr 98:458, 1981

143. Clinch TE, Duker JS, Eagle RC Jr et al: Infantile endogenous Candida endophthalmitis presenting as a cataract. Surv Ophthalmol 34:107, 1989

144. Fleming KO: Candida albicans abscess of retina. Can J Ophthalmol 7:132, 1972

145. Greenwald BD, Tunkel AR, Morgan KM et al: Candidal endophthalmitis after lithotripsy of renal calculi. South Med J 85:773, 1992

146. Chen SJ, Chung YM, Liu JH: Endogenous endophthalmitis after induced abortion. Am J Ophthalmol 125:873, 1998

147. Sixbey JW, Caplan ES: Candida parapsilosis endophthalmitis. Ann Intern Med 89:1010, 1978

148. Guex-Crosier Y, Chave JP, Herbort CP: Postpartum Candida endophthalmitis treated with fluconazole. Ophthalmologica 206:214, 1993

149. Wiesinger EC, Mayerhofer S, Wenisch C et al: Fluconazole in Candida albicans sepsis during pregnancy: Case report and review of the literature. Infection 24:263, 1996

150. Stanbury RM et al: Endogenous Candida endophthalmitis with no apparent predisposing factors. Eye 12:321, 1998

151. Kostick TK, Foster RE, Lowder CY et al: Endogenous endophthalmitis caused by Candida albicans in a healthy women. Am J Ophthalmol 15:593, 1992

152. Flynn HW Jr, Essman TF, Brod RD: Endogenous fungal endophthalmitis. In Saer J (ed): Proceedings of the 47th Annual Symposium of the New Orleans Academy of Ophthalmology, pp 297–305. The Hague: Kugler Publications, 1998

153. Elliott AJ: Endophthalmitis in systemic disease. A review of Candida albicans endophthalmitis. Semin Ophthalmol 2:229, 1987

154. He Y et al: An analysis of the cause of misdiagnosis with ocular opportunistic infection. Chung Hua Yen Ko Tsa Chih 33:45, 1997

155. Samiy N et al: Endogenous fungal endophthalmitis. Int Ophthalmol Clin 36:147, 1996

156. Shmuely H, Kremer I, Sagie A, Pitlik S: Candida tropicalis endophthalmitis as the only initial manifestation of pacemaker endophthalmitis. Am J Ophthalmol 123:559, 1997

157. Ellis CA, Spivack ML: The significance of candidemia. Ann Intern Med 67:511, 1967

158. Donahue SP, Greven CM, Zuravleff JJ et al: Intraocular candidiasis in patients with candidemia. Ophthalmology 101:102, 1994

159. Griffin JR, Foos RY, Petit TH: Relationship between Candida endophthalmitis, candidemia, and disseminated candidiasis. In: XXII Concilium Ophthalmologicum, Paris 1974, vol 2, p 661, 1976

160. McDonnell PJ, McDonnell JM, Brown RH, Green WR: Ocular involvement in patients with fungal infections. Ophthalmology 92:706, 1985

161. Bross J, Talbot GH, Maislon G et al: Risk factors for nosocomial candidemia: A case control study in adults without leukemia. Am J Med 87:614, 1989

162. Eilard T: Isolation of fungi in blood cultures: A review of fungal infections in the western part of Sweden 1970-1982. Scand J Infect Dis 19:145, 1987

163. Brooks RG: Prospective study of Candida endophthalmitis in hospitalized patients with candidemia. Arch Intern Med 149:2226, 1989

164. Scherer WJ, Lee K: Implication of early systemic therapy on the incidence of endogenous fungal endophthalmitis. Ophthalmol 104:1593, 1997

165. Nolla-Salas J, Sitges-Serra A, Leon C et al: Candida endophthalmitis in non-neutropenic patients. Eur J Microbiol Infect Dis 15:503, 1996

166. Enzenauer RW, Calderwood S, Levin AV et al: Screening for fungal endophthalmitis in children at risk. Pediatrics 90:451, 1992

167. Edwards JE Jr, Foos RY, Montgomerie JZ, Guze LB: Ocular manifestation of Review of seventy-six cases of hematogenous Candida endophthalmitis. Review of seventy-six cases of hematogenous Candida endophthalmitis. Candida septicemia. Medicine 53:47, 1974

168. Griffin JR, Pettit TH, Fishman LS, Foos RY: Blood-borne Candida endophthalmitis: A clinical and pathologic study of 21 cases. Arch Ophthalmol 89:450, 1973

169. Jampol LM, Sung J, Walker JD et al: Choroidal neovascularization secondary to Candida albicans chorioretinitis. Am J Ophthalmol 121:643, 1996

170. Shah GK, Fischer D, Fineman MS, Maguire J: Subretinal neovascularization secondary to Candida endophthalmitis: Long-term follow-up after submacular surgery. Retina 19:81, 1999

171. Naoi N et al: Effect of vitrectomy on epiretinal membranes after endogenous fungal endophthalmitis. Jpn J Ophthalmol 119:477, 1996

172. McDonald HR, De Bustros S, Sipperly JO: Vitrectomy for epiretinal membrane with Candida chorioretinitis. Ophthalmology 97:466, 1990

173. Pesin SR, Thomas MA, Smith ME: Combined rhegmatogenous-traction retinal detachment following successful treatment of Candida chorioretinitis. Arch Ophthalmol 110:1051, 1992

174. Foster RE, Rubsamen PE, Joondeph BC et al: Concurrent endophthalmitis and retinal detachment. Ophthalmology 101:490, 1994

175. Okhravi N, Adamson P, Mant R et al:Polymerase chain reaction and restriction fragment length polymorphism mediated detection and speciation of Candida sp. causing intraocular infection. Invest Ophthal Vis Sci 39:859, 1998

176. Schulman JA, Peyman GA: Intravitreal corticosteroids as an adjunct in the treatment of bacterial and fungal endophthalmitis. Retina 12:336, 1992

177. Coats ML et al: Intravitreal corticosteroids in the treatment of exogenous fungal endophthalmitis. Retina 12:46, 1992

178. Young RC, Bennett JE, Vogel CL et al: Aspergillosis: The spectrum of disease in 98 patients. Medicine 49:147, 1970

179. Weishaar PD, Flynn HW Jr, Murray TG et al: Endogenous Aspergillus endophthalmitis: Clinical features and treatment outcomes. Ophthalmology 105:57, 1998

180. Bennett JE: Aspergillus species. In Mandell GL, Bennett JE, Dolin R (eds): Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases, pp 1–12. Vol 2, 4th ed, chap 238. New York: Churchill Livingstone, 1995

181. Doft BH, Clarkson JG, Rebell G, Forster RK: Endogenous Aspergillus endophthalmitis in drug abusers. Arch Ophthalmol 98:859, 1980

182. Kotwal MR, Rinchhen CZ: Primary aspergillosis with multisystem dissemination. Lancet 1:562, 1981

183. Boldrey EE: Bilateral endogenous Aspergillus endophthalmitis. Retina 1:171, 1981

184. Michelson JB, Freedman SD, Boyden DG: Aspergillus endophthalmitis in a drug abuser. Ann Ophthalmol 14: 1051, 1982

185. Vishniavsky N, Sagar KB, Markowitz SM: Aspergillus fumigatus endocarditis on a normal heart valve. South Med J 76:506, 1983

186. Wilmarth SS, May DR, Roth AM et al: Aspergillus endophthalmitis in an intravenous drug abuser. Ann Ophthalmol 15:470, 1983

187. Roney P, Barr CC, Chun CH, Raff MJ: Endogenous Aspergillus endophthalmitis. Rev Infect Dis 8:955, 1986

188. Bosley TM, Kaufman KJ, Folberg R et al: Disseminated aspergillosis in a patient with ocular reticulum cell sarcoma. Br J Ophthalmol 71:528, 1987

189. Sihota R, Agarwal HC, Grover AK, Sood NN: Aspergillus endophthalmitis. Br J Ophthalmol 71:611, 1987

190. Naylor CD, Shkrum MJ, Edmonds MW, Cholod EJM: Pulmonary aspergillosis and endophthalmitis: Complications of Cushing's syndrome. Can Med Assoc J 138:719, 1988

191. Lance SE, Fribert TR, Kowalski RP: Aspergillus flavus endophthalmitis and retinitis in an intravenous drug abuser. A therapeutic success. Ophthalmology 95:947, 1988

192. Weiss JN, Hutchins RK, Balogh K: Simultaneous Aspergillus endophthalmitis and cytomegalovirus retinitis after kidney transplantation. Retina 8:193, 1988

193. Demicco DD, Reichman RC, Violette EJ, Winn WC Jr: Disseminated aspergillosis presenting with endophthalmitis: A case report and a review of the literature. Cancer 53:1995, 1984

194. Halperin LS, Roseman RL: Successful treatment of a subretinal abscess in an intravenous drug abuser. Arch Ophthalmol 106:1651, 1988

195. Bodoia RD, Kinyoun JL, Qingli L, Bunt-Milam AH: Aspergillus necrotizing retinitis: A clinico-pathologic study and review. Retina 9:226, 1989

196. Naidoff MA, Green WR: Endogenous Aspergillus endophthalmitis occurring after kidney transplant. Am J Ophthamol 79:502, 1975

197. Kammer RB, Utz JP: Aspergillus species endocarditis: The new face of a not-so-rare disease. Am J Med 56:506, 1974

198. Grahm DA, Kinyoun JL, George DP: Endogenous Aspergillus endophthalmitis after lung transplantation. Am J Ophthalmol 119:107, 1995

199. Kalina PH, Campbell RJ: Aspergillus terreus endophthalmitis in a patient with chronic lymphocyctic leukemia. Arch Ophthamol 109:102, 1991

200. Myles WM, Brownstein S, Deschenes J: Clinically unsuspected Aspergillus endophthalmitis. Can J Ophthalmol 32:182, 1997

201. Valluri S, Moorthy RS, Liggett PE, Rao NA: Endogenous Aspergillus endophthalmitis in an immunocompetent individual. Int Ophthalmol 17:131, 1993

202. Matsuo T, Nakagawa H, Matsuo N: Endogenous Aspergillus endophthalmitis associated with periodontitis. Ophthalmologica 209:109, 1995

203. Hunt KE, Glasgow BJ: Aspergillus endophthalmitis: An unrecognized endemic disease in orthotopic liver transplantation. Ophthalmology 103:757, 1996

204. Papanicolaou GA, Meyers BR, Fuchs WS et al: Infectious ocular complications in orthotopic liver transplant patients. Clin Infect Dis 24:1172, 1997

205. Kitahara M, Seth VK, Medoff G, Kobayashi GS: Antimicrobial susceptibility testing of six clinical isolates of Aspergillus. Antimicrob Agents Chemother 9:908, 1976

206. Denning DW, Lee JY, Hostetler JS et al: NIAID mycoses study group multicenter trial of oral itraconazole therapy for invasive aspergillosis. Am J Med 97:135, 1994

207. Gross JG: Endogenous Aspergillus endophthalmitis-induced endophthalmitis: Successful treatment without systemic antifungal medication. Retina 12:341, 1992

208. Georgiev VS: Treatment and developmental therapeutics in aspergillosis: I. Amphotericin B and its derivatives. Respiration 59:291, 1992

209. Rippon JW: Cryptococcosis. In: Medical Mycology. The Pathogenic Fungi and the Actinomycetes, pp 582–609. 3rd ed. Philadelphia: WB Saunders, 1988

210. Littman ML, Walter JE: Cryptococcosis: Current status. Am J Med 45:922, 1968

211. Chuck SL, Sande MA: Infections with Cryptococcus neoformans in the acquired immunodeficiency syndrome. N Engl J Med 321:794, 1989

212. Henson DJ, Hill AR: Cryptococcal pneumonia: A fulminant presentation. Ann J Med Sci 288:221, 1984

213. Lehmann PF, Morgan RJ, Freimer EH: Infection with Cryptococcus neoformans leading to a pulmonary cryptococcoma and meningitis. J Infect 9:301, 1984

214. Perfect JR, Durack DT, Gallis HA: Cryptococcemia. Medicine 62:98, 1985

215. Okun E, Butler WT: Ophthalmologic complications of cryptococcal meningitis. Arch Ophthalmol 71:52, 1964

216. Custis PH, Haller JA, de Juan E Jr: An unusual case of cryptococcal endophthalmitis. Retina 15:300, 1995

217. Denning DW, Armstrong RW, Fishman M, Stevens DA: Endophthalmitis in a patient with disseminated cryptococcosis and AIDS who was treated with itraconazole. Rev Infect Dis 13:1126, 1991

218. Ehrhorn J, Grosse G, Staib F, Wollensak J: Intraokulare Cryptococcose. Klin Monatsbl Augenheilkd 168:577, 1976

219. Condon PI, Terry SI, Falconer H: Cryptococcal eye disease. Doc Ophthalmol 44:49, 1977

220. Kumar RK, Lykke AWJ: Disseminated cryptococcosis with ocular involvement. Aust NZ J Med 9:444, 1979

221. Shields JA, Wright DM, Augsburger JJ, Wolkowicz MI: Cryptococcal chorioretinitis. Am J Ophthalmol 89:210, 1980

222. O'Dowd GJ, Frable WJ: Cryptococcal endophthalmitis: Diagnostic vitreous aspiration cytology. Am J Clin Pathol 79:382, 1983

223. Henderly DE, Liggett PE, Rao NA: Cryptococcal chorioretinitis and endophthalmitis. Retina 7:75, 1987

224. Malton ML, Rinkoff JS, Doft BS, Kennerdell JS: Cryptococcal endophthalmitis and meningitis associated with acute psychosis and exudative retinal detachment. Am J Ophthalmol 104:438, 1987

225. Crump JRC, Elner SG, Elner VM, Kauffman CA: Cryptococcal endophthalmitis: Case report and review. Clin Infect Dis 14:1069, 1992

226. Charle NC, Boxrud CA, Small EA: Cryptococcosis of the anterior segment in acquired immune deficiency syndrome. Ophthalmology 99:813, 1992

227. Kwon-Chung Kj, Bennett JE: Medical Mycology, p 397. Philadelphia: Lea & Febiger, 1992

228. Hiss PW, Shields JA, Augsburger JJ: Solitary retinovitreal abscess as the initial manifestation of cryptococcosis. Ophthalmology 95:162, 1988

229. Sheu SJ, Chen YC, Kuo NW et al: Endogenous cryptococcal endophthalmitis. Ophthalmology 105:377, 1998

230. Spickard A, Butler WT, Anriole V, Utz JP: The improved prognosis of cryptococcal meningitis with amphotericin B therapy. Ann Intern Med 58:66, 1963

231. Dismukes WE: Management of cryptococcosis. Clin Infect Dis 17 (Suppl 2):S507, 1993

232. Bennett JE, Dismukes WE, Duma RJ et al: A comparison of amphotericin B alone and combined with flucytosine in the treatment of cryptococcal meningitis. N Engl J Med 301:126, 1979

233. Saag MS, Powderly WG, Cloud G et al: Comparison of amphotericin B with fluconazole in the treatment of acute AIDS-associated cryptococcal meningitis. N Engl J Med 326:83, 1992

234. Bennett JE: The deep mycoses. Cryptococcosis, blastomycosis, histoplasmosis, and coccidioidomycosis. In Petersdorf RG, Adams RD, Braunwald E et al (eds): Harrison's Principles of Internal Medicine, pp 1056–1060. 10th ed. New York: McGraw-Hill, 1983

235. Smith CE: Epidemiology of acute coccidioidomycosis with erythema nodosum (San Juan or Valley fever). Am J Public Health 30:600, 1940

236. Drutz DJ, Catanzaro A: Coccidioidomycosis. Am Rev Respir Dis 117:559, 1978

237. Stevens DA (ed): Coccidioidomycosis: A Text. New York: Plenum Press, 1980

238. Ampel NM, Wieden MA, Galgiani JN: Coccidioidomycosis: Clinical update. Rev Infect Dis 11:897, 1989

239. Centers for Disease Control. Coccidioidomycosis, United States, 1991-1992. MMWR 42:21, 1993

240. Galgiani JN. Coccidioidomycosis: Changes in clinical expression, serological diagnosis, and therapeutic options. Clin Infect Dis 14(Suppl 1):S100, 1992

241. Warlick MA, Quan SF, Sobonya RE: Rapid diagnosis of pulmonary coccidioidomycosis. Cytology with potassium hydroxide preparations.Arch Intern Med 143:723, 1983

242. Kaufman L, Standard P: Improved version of the exoantigen test for identification of Coccidioides immitis and Histoplasma capsulatum cultures. J Clin Microbiol 8:42, 1978

243. Levitt JM: Ocular manifestations of coccidioidomycosis. Am J Ophthalmol 31:1626, 1948

244. Brown WC, Kellenberger RE, Hudson KE: Granulomatous uveitis associated with disseminated coccidioidomycosis. Am J Ophthalmol 45:102, 1958

245. Hagele AJ, Evans DJ, Larwood TR: Primary endophthalmic coccidioidomycosis: Report of a case of exogenous primary coccidioidomycosis of the eye diagnosed prior to enucleation. In Aiello L (ed): Coccidioidomycosis, pp 37–39. Tucson: University of Arizona Press, 1967

246. Petit TH, Learn RN, Foos RY: Intraocular coccidioidomycosis. Arch Ophthalmol 77:655, 1967

247. Alexander PB, Coodley EL: Disseminated coccidioidomycosis with intraocular involvement. Am J Ophthalmol 64:283, 1967

248. Green WR, Bennett JE: Coccidioidomycosis: Report of a case with clinical evidence of ocular involvement. Arch Ophthalmol 77:337, 1967

249. Boyden BS, Yee DS: Bilateral coccidioidal choroiditis: A clinicopathologic case report. Trans Am Acad Ophthalmol Otolaryngol 75:1006, 1971

250. Bell R, Font RL: Granulomatous anterior uveitis caused by Coccidioides immitis. Am J Ophthalmol 74:93, 1972

251. Cutler JE, Binder PS, Paul TO, Beamis JF: Metastatic coccidioidal endophthalmitis. Arch Ophthalmol 96:689, 1978

252. Rainin EA, Little HL: Ocular coccidioidomycosis: A clinicopathologic case report. Trans Am Acad Ophthalmol Otolaryngol 76:645, 1972

253. Chandler JW, Kalina RE, Milam DF: Coccidioidal choroiditis following renal transplantation. Am J Ophthalmol 74:1080, 1972

254. Zakka KA, Foos RY, Brown WJ: Intraocular coccidioidomycosis. Surv Ophthalmol 22:313, 1978

255. Blumenkranz MS, Stevens DA: Endogenous coccidioidal endophthalmitis. Ophthalmology 87:974, 1980

256. Glasgow BJ, Brown HH, Foos RY: Miliary retinitis in coccidioidomycosis. Am J Ophthalmol 104:24, 1987

257. Rodenbiker HT, Ganley JP, Galgiani JN, Axline SG: Prevalence of chorioretinal scars associated with coccidioidomycosis. Arch Ophthalmol 99:71, 1981

258. Stone JL, Kalina RE: Ocular coccidioidomycosis. Am J Ophthalmol 116:249, 1993

259. Rodenbiker HT, Ganley JP: Ocular coccidioidomycosis. Surv Ophthalmol 24:263, 1980

260. Moorthy RS, Narsing AR, Sidikaro Y, Foos RY: Coccidioidomycosis iridocyclitis. Ophthalmology 101:1923, 1994

261. Stevens DA, Stiller RL, Williams PL, Sugar AM: Experience with ketoconazole in three major manifestations of progressive coccidioidomycosis. Am J Med 74:58, 1983

262. Cantanzaro A, Friedman PJ, Schillaci R et al: Treatment of coccidioidomycosis with ketoconazole: An evaluation utilizing a new scoring system. Am J Med 74:64, 1983

263. Galgiani JN, Catanzaro A, Cloud GA et al: Fluconazole therapy for coccidioidal meningitis: The NIAID Mycoses Study Group. Ann Intern Med 119:28, 1993

264. Tucker RM, Denning DW, Arathoon EG et al: Itraconazole therapy for nonmeningeal coccidioidomycosis: Clinical and laboratory observations. J Am Acad Dermatol 23:593, 1990

265. Winn RE: Systemic fungal infections: Diagnosis and treatment. 1. Sporotrichosis. Infect Dis North Am 2:899, 1988

266. Wilson DE, Mann JJ, Bennett JE, Utz JP: Clinical features of extracutaneous sporotrichosis. Medicine 46:265, 1967

267. Lynch PJ. Voorhees JJ, Harrell ER: Systemic sporotrichosis. Ann Intern Med 73:23, 1970

268. Chang AC, Destouet JM, Murphy WA: Musculoskeletal sporotrichosis. Skeletal Radiol 12:23, 1985

269. Satterwhite TK, Kagler WV, Conklin RH et al: Disseminated sporotrichosis. JAMA 240:771, 1978

270. Michelson E: Primary pulmonary sporotrichosis. Ann Thorac Surg 24:83, 1977

271. Matthay RA, Greene WH: Pulmonary infections in the immunocompromised patient. Med Clin North Am 64: 529, 1980

272. Bibler MR, Luber HJ, Glueck HI, Estes SA: Disseminated sporotrichosis in a patient with HIV infection after treatment for acquired factor VIII inhibitor. JAMA 256:3215, 1986

273. Schenck BR: On refractory subcutaneous abscesses caused by a fungus related to the sporotricha. Bull Johns Hopkins Hosp 9:286, 1998

274. DeBeurmann CL, Gougerot H: Sporotrichose cachectisante mortelle. Bull Mem Soc Med Hop Paris 26:1046, 1909

275. Francois J. Rysselaere M: Oculomycoses, pp 188–189, 370–380. Springfield, IL: Charles C Thomas, 1972

276. Levy JH: Intraocular sporotrichosis: Report of a case. Arch Ophthalmol 85:574, 1971

277. Cassady JR, Foerster HC: Sporotrichum schenckii endophthalmitis. Arch Ophthalmol 85:71, 1971

278. Font RL, Jakobiec FA: Granulomatous necrotizing retinochoroiditis caused by Sporotrichum schenckii: Report of a case including immunofluorescence and electron microscopical studies. Arch Ophthalmol 94:1513, 1976

279. Kurosawa A, Pollack SC, Collins MP et al: Sporothrix schenckii endophthalmitis in a patient with human immunodeficiency virus infection. Arch Ophthalmol 106:376, 1988

280. Agger WA, Caplan RH, Maki DG: Ocular sporotrichosis mimicking mucormycosis in a diabetic. Ann Ophthalmol 10:767, 1978

281. Shelley WB, Sica PA Jr: Disseminated sporotrichosis of skin and bone cured with 5-fluorocytosine: Photosensitivity as a complication. J Am Dermatol 8:229, 1983

282. Rippon JW: A new ere in antimycotic agents. Arch Dermatol 122:399, 1986

283. Sharkey-Mathis PK, Kauffman CA, Graybill JR et al: Treatment of sporotrichosis with itraconazole: NIAID Mycoses Study Group. Am J Med 95:279, 1993

284. Kauffman CA: Newer developments in therapy for systemic mycoses. Clin Infect Dis 19S:S28, 1994

285. Gilchrist TC, Protozoan dermatitis. J Cutan Dis 12:496, 1894

286. Sarosi GA, Davies SF: Blastomycosis. Am Rev Respir Dis 120:911, 1979

287. Schwarz J, Salfelder K: Blastomycosis: A review of 152 cases. Curr Top Pathol 65:165, 1977

288. Bradsher RW: Blastomycosis. Infect Dis Clin North Am 2:877, 1988

289. Papas PG, Thelkeld MG, Bedsole GD et al: Blastomycosis in immunocompromised patients. Medicine 72:311, 1993

290. Churchill T, Stober AM: A case of systemic blastomycosis. Arch Intern Med 13:568, 1914

291. Schwartz VJ: Intraocular blastomycosis. Arch Ophthalmol 5:581, 1931

292. Casady JV: Uveal blastomycosis. Arch Ophthalmol 35:84, 1946

293. Sinskey RM, Anderson WB: Miliary blastomycosis with metastatic spread to posterior uvea of both eyes. Arch Ophthalmol 54:602, 1955

294. Font RL, Spaulding AG, Green WR: Endogenous mycotic panophthalmitis caused by Blastomyces dermatitidis: Report of a case and a review of the literature. Arch Ophthalmol 77:217, 1967

295. Bond WI, Sanders CV, Joffe L, Franklin RM: Presumed blastomycosis endophthalmitis. Ann Ophthalmol 14:1183, 1982

296. Lewis H, Aaberg TM, Fary DRB, Stevens TS: Latent disseminated blastomycosis with choroidal involvement. Arch Ophthalmol 106:527, 1988

297. Gottlieb JL, McAllister IL, Guttman FA, Vine AK: Choroidal blastomycosis: A report of 2 cases. Retina 15:248, 1995

298. Li S, Perlman JI, Edward DP, Weiss R: Unilateral Blastomyces dermatitidis endophthalmitis and orbital cellulitis. Ophthalmology 105:1466, 1998

299. Safneck JR, Hogg GA, Napier LB: Endophthalmitis due to Blastomyces dermatitidis: Case report and review of the literature. Ophthalmology 97:212, 1990

300. Vida L, Moel SA: North American blastomycosis with orbital involvement. Am J Ophthalmol 77:240, 1974

301. McManus EJ, Jones JM: The use of ketoconazole in the treatment of blastomycosis. Am Rev Respir Dis 133:141, 1986

302. Dismukes WE, Bradsher RW, Cloud GA et al: Itraconazole therapy for blastomycosis and histoplasmosis. Am J Med 93:489, 1992

303. Rippon JW: Medical mycology: The pathogenic fungi and the pathogenic actinomycetes. In Freeman BA (ed): Burrow's Textbook of Microbiology, pp 760–774. 21st ed. Philadelphia: WB Saunders, 1979

304. Goodwin RA Jr, Desperez RM: Histoplasmosis: State of the art. Am Rev Respir Dis 117:929, 1978

305. Kauffman CA, Isreal AS et al: Histoplasmosis in immunosuppressed patients. Am J Med 64:923, 1978

306. Specht CS, Mitchell KT, Baum AE, Gupta M: Ocular histoplasmosis with retinitis in a patient with the acquired immune deficiency syndrome. Ophthalmology 98:1356, 1991

307. Macher A, Rodrigues MM, Kaplan W et al: Disseminated bilateral chorioretinitis due to Histoplasma capsulatum in a patient with the acquired immunodeficiency syndrome. Ophthalmology 92:1159, 1985

308. Goldstein BG, Buettner H: Histoplasmic endophthalmitis: A clinicopathologic correlation. Arch Ophthalmol 101: 774, 1983

309. Klintworth GK, Hollingsworth AS, Lusman PA, Bradford WD: Granulomatous choroiditis in a case of disseminated histoplasmosis. Histopathologic demonstration of Histoplasma capsulatum in choroidal lesions. Arch Ophthalmol 90:45, 1973

310. Scholz R, Green WR, Kutys R et al: Histoplasma capsulatum in the eye. Ophthalmology 91:1100, 1984

311. Schwartz J, Salfelder K, Viloria JE: Histoplasma capsulatum in vessels of the choroid. Ann Ophthalmol 9:633, 1977

312. Schwartz J: Granulomatous histoplasmosis of the eye: A rare disease. Int Ophthalmol Clin 15:65, 1975

313. Weingeist TA, Watzke RC: Ocular involvement by Histoplasma capsulatum. Int Ophthalmol Clin 23:33, 1982

314. Ganley JP: Epidemiologic characteristics of presumed ocular histoplasmosis. Acta Ophthalmol Scand Suppl 119:1, 1973

315. Katz BJ, Scott WE, Folk JC: Acute histoplasmosis choroiditis in 2 immunocompetent brothers. Arch Ophthalmol 115:1470, 1997

316. Davies SF: Serodiagnosis of histoplasmosis. Semin Respir Infect 1:9, 1986

317. Slama TG: Treatment of disseminated and progressive cavitary histoplasmosis with ketoconazole. Am J Med 70, 1983

318. Sarosi GA Davies SF: Therapy for fungal infections. Mayo Clin Proc 69:1111, 1994

319. Goodwin RA Jr, Shapiro JL, Thurman GH et al: Disseminated histoplasmosis: Clinical and pathologic correlations. Medicine 59:1, 1980

320. Pfeifer JD, Grande MG, Thomas MA et al: Endogenous Pseudallescheria boydii endophthalmitis: Clinicopathologic findings in two cases. Arch Ophthalmol 109:1714, 1991

321. Taravella MJ, Johnson DW, Petty JG et al: Infectious posterior scleritis caused by Pseudallescheria boydii. Clinicopathologic findings. Ophthalmology 104:1312, 1997

322. McGuire TW, Bullock JD, Bullock JD Jr et al: Fungal endophthalmitis: An experimental study with review of 17 human ocular cases. Arch Ophthalmol 109:1289, 1991

323. Lieberman TW, Ferry AP, Bottone EJ: Fusarium solani endophthalmitis without primary corneal involvement. Am J Ophthalmol 88:764, 1979

324. Cho CT, Vats TS, Lowman JT et al: Fusarium solani infection during treatment for acute leukemia. J Pediatr 83: 1028, 1973

325. Mayayo E, Guarro J, Pujol I: Endogenous endophthalmitis by Fusarium solani: An animal experimental model. Med Mycol 36:249, 1998

326. Glasgow BJ, Engstrom RE Jr, Holland GN et al: Bilateral endogenous Fusarium endophthalmitis associated with acquired immunodeficiency syndrome. Arch Ophthalmol 114:873, 1996

327. Gabriele P, Hutchins RK: Fusarium endophthalmitis in an intravenous drug abuser. Am J Ophthalmol 122:119, 1996

328. Louie T, Baba F, Shulman M, Jimenez-Lucho V: Endogenous endophthalmitis due to Fusarium: Case report and review. Clin Infect Dis 18:585, 1994

329. Pittke EC, Hertenstein B, Bachor R: Orbitale und okulare Mucormykose. Klin Monatsbl Augenheilkd 1900:530, 1987

330. Minogue MJ et al: Cure of Paecilomyces endophthalmitis with multiple intravitreal injections of amphotericin B. Arch Ophthalmol 107:1281, 1989

331. Lam DS et al: Endogenous fungal endophthalmitis caused by Paecilomyces variotii. Eye 13:113, 1999

332. Okhravi N et al: Paecilomyces lilacinus endophthalmitis with secondary keratitis: A case report and literature review. Arch Ophthalmol 115:1320, 1997

333. Swan SK, Wagner RA, Myers JP, Cinelli AB: Mycotic endophthalmitis caused by Pencillium sp after parenteral drug abuse. AM J Ophthalmol 100:408, 1985

334. Pavan PR et al: Endogenous endophthalmitis due to Bipolaris hawaiiensis in a patient with acquired immunodeficiency syndrome. Am J Ophthalmol 116:644, 1993

335. Walsh TJ, Orth DH, Shapiro CM et al: Metastatic fungal chorioretinitis developing during Trichosporon sepsis. Ophthalmology 89:152, 1982

336. Gregor RJ, Chong CA, Augsburger JJ et al: Endogenous Nocardia asteroides subretinal abscess diagnosed by transvitreal fine-needle aspiration biopsy. Retina 9:118, 1989

337. Ferry AP, Font RL, Weiberg RS et al: Nocardial endophthalmitis: Report of two cases studied histopathologically. Br J Ophthalmol 72:55, 1988

338. Mamalis N, Daily MJ, Ros D: Presumed intraocular nocardiosis in a cardiac-transplant patient. Ann Ophthalmol 20:271, 1988

339. Bullock JD: Endogenous ocular nocardiosis: A clinical and experimental study. Trans Am Ophthalmol Soc 81:451, 1983

340. Ishibashi Y, Watanabe R, Hommura S et al: Endogenous Nocardia asteroides endophthalmitis in a patient with systemic lupus erythematosus. Br J Ophthalmol 74:433, 1990

341. Wallace RJ JR, Steele LC, Sunter G, Smith JM: Antimicrobial susceptibility patterns of Nocardia asteroides. Antimicrob Agents Chemother 32:1776, 1988

Back to Top