Opportunistic Amoebae: Challenges in Prophylaxis and Treatment

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Drug Resistance Updates 7 (2004) 41–51

Review
Opportunistic amoebae: challenges in prophylaxis and treatment
Frederick L. Schuster a,∗ , Govinda S. Visvesvara b
a Viral and Rickettsial Disease Laboratory, California Department of Health Services, Richmond, CA 94804, USA
b Division of Parasitic Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA

Received 29 December 2003; received in revised form 10 January 2004; accepted 10 January 2004

Abstract
This review focuses on free-living amoebae, widely distributed in soil and water, causing opportunistic and non-opportunistic infections
in humans: Acanthamoeba spp., Balamuthia mandrillaris, Naegleria fowleri, and Sappinia diploidea. Diseases include primary amoebic
meningoencephalitis (N. fowleri), granulomatous amoebic encephalitis, cutaneous and nasopharyngeal infections (Acanthamoeba spp.,
Balamuthia mandrillaris, S. diploidea), and amoebic keratitis (Acanthamoeba spp). Acanthamoeba, Balamuthia, and Naegleria have been
repeatedly isolated; S. diploidea has been reported only once, from a brain infection. Antimicrobial therapy for these infections is gener-
ally empirical and patient recovery often problematic. N. fowleri is highly sensitive to the antifungal agent amphotericin B, but delay in
diagnosis and the fulminant nature of the disease result in few survivors. Encephalitis and other infections caused by Acanthamoeba and
Balamuthia have been treated, more or less successfully, with antimicrobial combinations including sterol-targeting azoles (clotrimazole,
miconazole, ketoconazole, fluconazole, itraconazole), pentamidine isethionate, 5-fluorocytosine, and sulfadiazine. The use of drug com-
binations addresses resistance patterns that may exist or develop during treatment, ensuring that at least one of the drugs may be effective
against the amoebae. Favorable drug interactions (additive or synergistic) are another potential benefit. In vitro drug testing of clinical
isolates points up strain and species differences in sensitivity, so that no single drug can be assumed effective against all amoebae. Another
complication is risk of activation of dormant cysts that form in situ in Acanthamoeba and Balamuthia infections, and which can lead to
patient relapse following apparently effective treatment. This is particularly true in Acanthamoeba keratitis, a non-opportunistic infection
of the cornea, which responds well to treatment with chlorhexidine gluconate and polyhexamethylene biguanide, in combination with
propamidine isothionate (Brolene), hexamidine (Désomodine), or neomycin. Acanthamoeba spp. may also be carriers of endosymbiotic
bacteria (Legionella and Legionella-like pathogens) and have been implicated in outbreaks of pneumonias in debilitated hosts. As with
other infectious diseases, recovery is dependent not only on antimicrobial therapy, but also on patient’s immune status, infective dose and
virulence of the ameba strain, and on how early the disease is diagnosed and drug therapy initiated.
© 2003 Elsevier Ltd. All rights reserved.
Keywords: Amoebic encephalitis; Amoebic keratitis; Acanthamoeba spp.; Balamuthia mandrillaris; Naegleria fowleri; Sappinia diploidea

1. Introduction granulomatous amoebic encephalitis, as well as dissemi-


nated, cutaneous and nasopharyngeal infections, primarily
Free-living amoebae belonging to the genera Acan- in immunocompromised individuals; they also cause amebic
thamoeba, Balamuthia, and Naegleria are responsible for keratitis in immunocompetent persons. Balamuthia man-
opportunistic and non-opportunistic infections in humans drillaris, a close relative of Acanthamoeba, causes similar
and other animals (Martinez and Visvesvara, 1997). These infections in both immunocompromised and immunocom-
organisms can be isolated from soil and water, and are petent hosts, the latter being mostly children. Naegleria
widely spread in the outdoor and home (flowerpots, aquaria, fowleri is the causal agent of primary amoebic menin-
water taps and sink drains, humidifiers) environments. In goencephalitis, a non-opportunistic infection in otherwise
the soil habitat, they feed on bacteria, except for Balamuthia healthy children and young adults. There is no universal
which probably feeds on other amoebae. Persons devel- cure for these amoebic infections. With few exceptions, an-
oping infections include both immunocompromised and timicrobial therapy is largely empirical, and optimal therapy
immunocompetent individuals. Acanthamoeba spp. cause has yet to be determined. The mouse serves as a useful ani-
mal model for study of amoebic encephalitides; the animal
∗ Corresponding author. Tel.: +1-510-307-8651.
is inoculated intranasally or intracranially with a suspension
E-mail addresses: fschuste@dhs.ca.gov (F.L. Schuster), of amoebae and the murine disease parallels the human
gsv1@cdc.gov (G.S. Visvesvara). infection (Janitschke et al., 1996; Jarolim et al., 2000;

1368-7646/$ – see front matter © 2003 Elsevier Ltd. All rights reserved.
doi:10.1016/j.drup.2004.01.002
42 F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51

Martinez et al., 1973). Most drug testing, however, is carried of chlorhexidine gluconate and ketoconazole cream. Lo-
out in vitro. calized infections (e.g. cutaneous lesions) can develop into
disseminated infections through hematogenous spread. In
spite of several recoveries following drug therapy, the out-
2. Acanthamoeba spp. look for most patients remains grim, especially those weak-
ened by multiple opportunistic infections. Some successful
The Acanthamoeba life-cycle consists of trophic and cys- treatment regimens have included pentamidine isethion-
tic stages, with the latter being more resistant to antimi- ate, flucytosine, itraconazole, topical chlorhexidine and
crobials. Approximately 17 species of Acanthamoeba are ketoconazole cream (for skin lesions) in a lung transplant
recognized based on morphology and genomic sequencing patient with disseminated acanthamebiasis without central
(Stothard et al., 1998), several of which cause opportunis- nervous system (CNS) involvement (Oliva et al., 1999);
tic infections (Marciano-Cabral and Cabral, 2003). Many of trimethoprim-sulfamethoxazole, rifampin, and ketoconazole
the infections have been in HIV/AIDS and organ-transplant in two pediatric patients with CNS infections (Singhal et al.,
patients, or persons in a debilitated state of health. 2001); cotrimoxazole, flucytosine, sulfadiazine in a pediatric
A broad array of drugs has been used to treat acan- patient with CNS infection (Karande et al., 1991); intra-
thamoebic infections and some successes have been reported venous pentamidine, topical chlorhexidine, and ketocona-
(Schuster and Visvesvara, 2003). Most cases, however, es- zole cream in a renal transplant patient with disseminated
pecially in hosts weakened by opportunistic infections or infection (Slater et al., 1994); sulfadiazine, and fluconazole
made vulnerable by immunosuppressive medications, are fa- in an HIV/AIDS patient with granulomatous encephalitis
tal. For amoebic encephalitis, the problem of finding opti- (Martinez et al., 2000); flucytosine therapy following inef-
mal antimicrobial therapy is compounded by difficulty in fective ketoconazole treatment in an HIV/AIDS patient with
diagnosing the infection. Lacking specific pathognomonic cutaneous infection (Helton et al., 1993); multidrug therapy
symptoms, recognition of the infection is often at autopsy. for treatment of amoebic rhino-sinusitis in an HIV/AIDS pa-
Even then, infections may be missed in routine histopatho- tient (Rivera and Padhya, 2002). A combination of penicillin
logical reviews due to lack of familiarity with the amoebae. and chloramphenicol was apparently successful in treating
In contrast to systemic infections caused by Acan- a patient with meningitis in which Acanthamoeba culbert-
thamoeba which have an insidious and subclinical course, soni was recovered from the CSF (Lalitha et al., 1985); no
amoebic keratitis, a non-opportunistic infection, will cause post-isolation in vitro testing of the amoeba was performed
the patient to seek help because of pain, tearing, dimin- to confirm sensitivity. A chronic CNS infection with Hart-
ished vision, and photophobia. Diagnosis is by corneal mannella (Acanthamoeba) rhysodes improved but was not
scraping and staining to visualize amoebae, or by culti- cured following sulfamethazine treatment (Cleland et al.,
vation of the scrapings for isolation and identification of 1982). Acanthamoeba castellanii, isolated from skin nodules
the amoeba (Schuster, 2002). Unlike protocols for bacte- of a fatal cutaneous infection in an HIV/AIDS patient, was
rial susceptibility testing, there are no reference strains of resistant in vitro to metronidazole, amphotericin B, rifampin,
Acanthamoeba or other amoebae to be used as standards pentamidine, 5-fluorcystosine, and itraconazole (Hunt et al.,
for testing amoebic susceptibility. During in vitro testing, it 1995). Amphotericin B was found ineffective in treating a
is also important to distinguish between amoebastatic and fatal case of disseminated acanthamoebiasis (Gullet et al.,
amoebicidal drugs. With the former, amoebae that appear 1979). Recurrence of disease in cases that were success-
to be non-dividing or dead upon microscopic examination fully treated with antimicrobials has not been reported, but
recover following transfer to drug-free medium. Drugs must the possibility exists that cysts in brain and other tissues
also be cysticidal to prevent recurrence of infection from might reactivate to give rise to trophic amoebae. Table 1
activation of dormant cysts that have survived antimicrobial provides a summary of selected successful antimicrobial
treatment. Because of variable sensitivities, each isolate re- therapies that have been used in systemic Acanthamoeba
quires individual testing for susceptibility to drugs and drug infections.
concentrations, leading to sometimes contradictory results A number of other antimicrobials have been tested in
from different laboratories. vitro for activity against Acanthamoeba isolates, but have
not yet been tried in the clinic. Azithromycin was inhibitory
2.1. Antimicrobial treatment for systemic Acanthamoeba for up to 98% of brain, skin, and corneal isolates, while the
infections related macrolides clarithromycin and erythromycin were
ineffective (Schuster and Visvesvara, 1998). Several phe-
Drugs that have been used in treating nasopharyngeal, nothiazines (e.g. chlorpromazine, trifluoperazine) showed
disseminated, and central nervous system infections in- activity against Acanthamoeba clinical isolates (Schuster
clude ketoconazole, fluconazole, itraconazole, pentamidine and Visvesvara, 1998). Miltefosine (hexadecylphospho-
isethionate, trimethoprim-sulfamethoxazole (cotrimoxa- choline), an anti-cancer drug used for treating visceral
zole), sulfadiazine, and 5-fluorocytosine (flucytosine). Cuta- leishmaniasis (Sundar et al., 2002) is also active against En-
neous infections have been treated with topical applications tamoeba histolytica (Seifert et al., 2001) and Acanthamoeba
F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51 43

Table 1
in halting an infection is determined by a number of factors,
Examples of successful antimicrobial treatments of Acanthamoeba infec-
tions including the immune status of the host, the progression of
the disease and the time at which drug intervention is initi-
Antimicrobials Dosage Reference
ated, the infectious dose of amoebae, and the virulence and
Amebic encephalitis drug sensitivity profile of the particular strain causing the
Sulfamethazine 1 g qid Cleland et al. (1982)
infection.
Cotrimoxazole 75 mg/kg q12h IV Karande et al. (1991) Variations in both virulence and antibiotic sensitivity are
5-Fluorocytosine 150 mg/kg q6h encountered among clinical isolates of Acanthamoeba, even
Sulfadiazine 150 mg/kg q6h
between strains of a single species. With regard to differ-
Penicillin G 2 × 106 U q3h IV Lalitha et al. (1985) ences in virulence, some species and/or strains produce a
Chloramphenicol 500 mg q6h po
higher mortality in the mouse model or cause a more pro-
Sulfadiazine 500 mg qid Martinez et al. (2000)a nounced cytopathic effect in tissue cultures (De Jonckheere,
Pyrimethamine 50 mg qd 1980). A brain isolate (A. healyi) was more virulent than
Fluconazole 200 mg bid
isolates grown in tissue culture (A. culbertsoni) or those
Ketoconazole 5 mg/kg qd Singhal et al. (2001) from the environment when tested in an immunodeficient
Rifampin 10 mg/kg qd
mouse model (Kong et al., 1998). Likewise, mouse brain
TMP-SMX 20 mg/kg qd
and keratitis isolates were more cytopathogenic in tissue
Cutaneous infection cultures than isolates from tap water or contact lens cases
Ketoconazole 200 mg q8h IV Helton et al. (1993)a
5-Fluorocytosine 40 mg/kg q8h
(Walochnik et al., 2000). With regard to differences in
antimicrobial susceptibility, a comparative study of four
Pentamidine 4 mg/kg q24h IV Slater et al. (1994)
species of Acanthamoeba, found variations in sensitiv-
Topical chlorhexidine Cleansing bid
Ketoconazole cream 2% following ity to clotrimazole, with Acanthamoeba polyphaga being
cleansing most resistant and A. rhysodes most sensitive to the drug
Nasopharyngeal infection
(Stevens and Willaert, 1980). In a comparative study of
Pentamidine 4 mg/kg qd IV Rivera and Padhya azoles, clotrimazole showed the best inhibition against a
Levofloxacin 500 mg qd IV (2002)a different strain of A. polyphaga: clotrimazole (up to 99% in-
Amphotericin B 550 mg qd IV hibition) > bifonazole > ketoconazole > itraconazole > flu-
5-Fluorocytosine 2 gm q6h conazole (no inhibition) (Schuster, 1993). Even so, at the
Rifampin 600 mg bid po
Itraconaole 200 mg bid po
concentrations tested, the azoles were amoebastatic but not
amoebacidal.
Disseminated infection
The cell membrane is the target of azole compounds and
Pentamidine Dosages not given Oliva et al. (1999)
5-Fluorocytosine of amphotericin B (AMB). The azoles interfere with syn-
Itraconazole thesis of sterols that are incorporated into the membrane,
Topical chlorhexidine leading to defective membrane architecture, increased per-
Ketoconazole cream meability, and leakage of ions from the cell (Sande and
Abbreviations: qxh, every x hours; qd, every day; bid, twice a day; qid, Mandell, 1985). Ergosterol and 7-dehyrostigmasterol are
four times each day; po, oral administration; IT, intrathecal administration; major sterol membrane components of the non-pathogenic
IV, intravenous administration. A. castellanii (Smith and Korn, 1968) and the pathogenic A.
a HIV/AIDS patient.
culbertsoni strain A-1 (Mehdi et al., 1988). Cycloartenol is
the ergosterol precursor for A. polyphaga, whose conversion
isolates (Walochnik et al., 2002). Although not tested clini- is presumably blocked by azole activity (Raederstorff and
cally against Acanthamoeba, miltefosine may have potential Rohmer, 1985). The imidazoles (clotrimazole, bifonazole,
for treatment of systemic infections as well as amoebic ketoconazole) are more effective against Acanthamoeba than
keratitis (Seal, 2003). Ability of antimicrobials to enter the triazoles (itraconazole, fluconazole) (Schuster, 1993).
the brain and CSF are also important considerations. Mil- The polyene AMB acts by preferentially binding to ergos-
tefosine and the phenothiazines penetrate the blood-brain terol and producing pores in the cell membrane (Sande and
barrier as does azithromycin, although accumulation of the Mandell, 1985). It is not the drug of choice for acanthamoe-
latter in cerebrospinal fluid is reportedly low (0.015 ␮g/ml) bic infections, since many isolates are neither inhibited
(Jaruratanasirikul et al., 1996). nor killed by it (Duma and Finley, 1976). The pathogenic
Because of a multidrug treatment approach, there is lit- N. fowleri, however, is highly sensitive to AMB (see
tle or no evidence of clinical resistance to any of the drugs below).
used in treating Acanthamoeba infections. Drug combina-
tions might give rise to synergistic or additive effects that are 2.2. Antimicrobial treatment for Acanthamoeba keratitis
not apparent from in vitro testing of individual drugs. Drug
testing in the mouse model, simulating a clinical situation, Amoebic keratitis can result from corneal trauma or, more
is infrequent. The value of a drug or combination of drugs likely, the transfer of Acanthamoeba spp. either in their
44 F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51

Table 2
vegetative state or as dormant cysts to the corneal surface
Successful antimicrobial treatment regimens for Acanthamoeba keratitis,
through the use of improperly or poorly maintained contact listed chronologically
lenses. The contact lens storage case, for lack of proper
Antimicrobials Dosage Reference
cleaning, can support growth of bacterial biofilms, which
can turn the case into a culture vessel for the bacteria-eating Ketoconazole 200 mg bid po Hirst et al. (1984)
amoebae. Amoebae are transferred to the eye when the con- Miconazole drops 10 mg/ml qh
Gentamicin drops qid
tact lens is placed upon the corneal surface. Once established
there, they penetrate the stroma, from which they are difficult Propamidine isethionate 0.1% qh Wright et al. (1985)
to eradicate. Because there is a greater likelihood for diagno- Dipropamidine ointment 0.15% q4h
Neomycin q4h
sis of amoebic keratitis than for systemic infections, antimi-
crobial treatment can be initiated early in the course of infec- Brolenea eyedrops qds Yeoh et al. (1987)
Brolene ointment qds
tion and, with compliant and intensive drug use (often hourly
applications), prospects for recovery are excellent. Drug use, Clotrimazole drops 1% qh Driebe et al. (1988)
however, can induce encystment (Kilvington et al., 1990) Propamidine qh
Ketoconazole 200 mg bid po
and once drug levels have declined, can lead to excystation Topical N-PB-Gb qh
and flare-up of the infection. In order to preclude recurrence,
Itraconazole 50 mg qd po Ishibashi et al. (1990)
treatment must be continued for 3–4 months (Kosrirukvongs
Topical miconazole
et al., 1999; Ficker et al., 1990). Some of the drugs initially
used in treating these infections were marginally amoe- Neosporinc drops qid Sharma et al. (1990)
Miconazole drops 10 mg/ml qh
bacidal or cysticidal and allowed eventual recovery from
inhibition. The cyst is more likely to survive treatment ow- PHMBd 0.02% qh to q3h Larkin et al. (1992)
ing to its thick wall and impermeability (Aksozek et al., Chlorhexidine 0.02% in saline qh Seal et al. (1995)
2002). Distinctly higher drug concentrations are required Propamidine 0.1% qh
to destroy cysts than are needed to destroy trophic amoe- PHMB 0.02% qh to qid Murdoch et al. (1998)
bae (Kilvington et al., 1990; Turner et al., 2000), and the ±Propamidine 0.1% qh
process of encystation is accompanied by development of ±Chlorhexidine 0.1% qid
±Désomedinee
drug resistance (Lloyd et al., 2001; Larkin et al., 1992; Lim
et al., 2000; Turner et al., 2000; Wright et al., 1985). Re- Chlorhexidine 0.006% qh Kosrirukvongs
currence of infection from excysting amoebae may lead to et al. (1999)
repeated corneal transplants (keratoplasty) in attempts to PHMB 0.02% qh Pérez-Santonja
eliminate or reduce numbers of amoebae and restore vision. ±Chlorhexidine 0.02% qh et al. (2003)
±Brolene 0.1% qh
In more chronic situations or where stubborn drug resis-
±Désomedine 0.1% qh
tance has been encountered, enucleation (surgical removal
of the eye from its socket) is a last resort to halting the The dosages and scheduling indicated in the table are generally for initial
treatment, and drug applications taper off over time and with improvement.
infection. In some of the referenced papers, several case histories are presented with
Substantially more literature exists on the drug sensi- minor differences in treatments.
tivity of Acanthamoeba strains and species involved in a Brolene: propamidine isethionate and dipropamidine.
b N-PB-G: neomycin–polymyxin B–gramicidin.
amoebic keratitis, than in systemic infections. Some of the
c Neosporin: neomycin–polymyxin B–bacitracin.
earlier chemotherapies employed were topical neomycin– d PHMB: polyhexamethylene biguanide.
polymyxin B–bacitracin (Neosporin) with or without mi- e Désomodine (Desmodine): hexamidine.
conazole or ketoconazole (Sharma et al., 1990); oral itra-
conazole and topical miconazole (Ishibashi et al., 1990);
systemic ketoconazole and topical miconazole (Hirst 1993). Its use, however, is discouraged because of resis-
et al., 1984); topical clotrimazole plus other antimicro- tance of cysts, and possible neurotoxicity (Murdoch et al.,
bials (Driebe et al., 1988). Brolene eyedrops (an over-the 1998; Seal, 2003). Selected examples of successful an-
counter pharmaceutical containing propamidine isethionate timicrobial regimens for treatment of amoebic keratitis are
and dibromopropamidine) and ointment, available in the presented in Table 2. Anti-inflammatory agents (topical
United Kingdom, have been used successfully in treating steroids) are generally added to the anti-amoebic treatment
keratitis cases (Yeoh et al., 1987), but are not well-tolerated regimens (Larkin et al., 1992; Pérez-Santonja et al., 2003),
in the eye when used over long periods of time (Murdoch but caution has been suggested in their use because of sup-
et al., 1998; Wright et al., 1985; Yeoh et al., 1987). Re- pression of macrophage activity (Berger et al., 1990; Seal,
sistance to propamidine was found in six out of eight 2003).
clinical isolates of Acanthamoeba (minimal inhibitory A major improvement in treating amoebic keratitis came
concentration ≥ 500 ␮g/ml) (Pérez-Santonja et al., 2003). about with the use of two cationic antiseptics, chlorhexi-
Neomycin was effective against trophic amoebae in vitro, dine gluconate and polyhexamethylene biguanide (PHMB;
but less so against cysts (Hay et al., 1994; Varga et al., Bacquacil) (Elder and Dart, 1995; Larkin et al., 1992).
F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51 45

These have become the drugs of choice for treating ker- Ficker et al., 1990; Lim et al., 2000; Pérez-Santonja et al.,
atitis. Chlorhexidine may have an advantage over PHMB 2003; Seal, 2003).
(Seal et al., 1995; but see Elder and Dart, 1995 for a dif- Animal models have been developed to study amoebic
ferent view). Used at a concentration of 0.02% or lower, keratitis, but they have not been used to evaluate antimicro-
the two drugs have been shown to be clinically effective bial efficacy (Badenoch et al., 1990; He et al., 1992; van
against both vegetative amoebae and dormant cysts, and Klink et al., 1993). A readily available model for in vivo
are well-tolerated in the eye (Kosrirukvongs et al., 1999). testing has yet to be established. Corneal and other isolates
PHMB is a more potent cysticidal agent than propami- of amoebae produce cytopathogenicity in tissue cultures,
dine (Larkin et al., 1992; Varga et al., 1993). PHMB was which can be used as an index of strain virulence (Badenoch
used with or without other antimicrobials (chlorhexidine, et al., 1995; Niszl et al., 1998). The ability of antimicrobials
Brolene, and hexamidine, a diamidine compound marketed to block cytopathic destruction of tissue culture monolayers
in France as Désomedin) (Pérez-Santonja et al., 2003). has, in turn, been used as a technique for drug evaluation
Chlorhexidine and PHMB have been used alone, or in (Schuster and Visvesvara, 1998).
combination with propamidine isethionate and/or neomycin
(Hay et al., 1994; Lindquist, 1998), or with 0.1% hexami- 2.3. Acanthamoeba as carrier of pathogenic bacteria
dine (Murdoch et al., 1998); they were also used in com-
bination (Murdoch et al., 1998), although the two together Acanthamoeba isolates from environmental water and soil
might prove to be toxic (Seal, 2003). samples (Newsome et al., 1998) as well as clinical samples
Development of drug resistance during prolonged treat- (Fritsche et al., 1993; Rowbotham, 1983) have been found
ment of infections is understandably a concern. Unlike the to harbor endosymbiotic bacteria. The most prominent of
situation for systemic infections where it is difficult to as- these are Legionella spp. or Legionella-like bacteria, which
sess drug resistance, it is more readily apparent in keratitis have had a role in community-acquired and nosocomial
when treatment does little to alleviate symptoms. Repeated pneumonias. Amoebae found in tap water, ventilating and
amoeba isolation from a persistent keratitis case demon- cooling systems and humidifiers are lysed by these bacteria,
strated resistance development to Brolene (propamidine) and and the bacteria are subsequently released in aerosols. Oc-
to an experimental arsenic compound (R6/56) in the course currences in hospitals and office buildings of legionellosis,
of the infection, as shown by an increase in the minimal in- Pontiac and humidifier fevers, and atypical pneumonias have
hibitory drug concentration for the two compounds (Ficker been associated with Acanthamoeba spp. and pose a partic-
et al., 1990). Two keratitis cases were reported to be re- ular problem for elderly or immunocompromised patients.
fractory to PHMB, even though the isolated amoebae were Endosymbionts may also influence outcomes of cases of
demonstrably sensitive to the compound in vitro (Murdoch amoebic keratitis (Murdoch et al., 1998). In addition to Le-
et al., 1998). The rationale for using combination drug ther- gionella spp., bacteria with disease potential found naturally
apy is to minimize risk of selection for resistance during in Acanthamoeba and other amoebae are Parachlamydia
treatment of an infection. acanthamoeba (Maurin et al., 2002), Neochlamydia hart-
Other drugs for use in amoebic keratitis have been mannellae (Horn et al., 2000), and Rickettsia-like organisms
tested in vitro. Magainins, a group of natural and syn- (Fritsche et al., 1999). Furthermore, Acanthamoeba can
thetic membrane-active peptides, tested favorably when be readily infected in vitro with bacteria including Afipia
combined with silver nitrate (Schuster and Jacob, 1992). felis, Burkholderia cepacia, Escherichia coli O157, Liste-
Povidone-iodine (Betadine) was reported to be a better ria monocytogenes, Mycobacterium avium, Mycobacterium
amoebicidal agent than chlorhexidine when tested against bovis, Simkania negevensis, and Vibrio cholerae.
corneal isolates of Acanthamoeba in vitro (Gatti et al., 1998). Little has been done to determine antimicrobial sensi-
Activity of seven different diamidine compounds, including tivities of naturally occurring endosymbionts, despite their
propamidine and hexamidine, against Acanthamoeba was potential as etiologic agents of pneumonia. This is due in
shown to be proportional to the length of the alkyl chain part to the difficulty in culturing some of the endosymbiotic
of the diamidine molecule, relating to its lipophilicity and bacteria. In a survey of patients with community-acquired
its ability to penetrate the amoeba cell membrane (Perrine pneumonia, antibodies were detected to Legionella-like
et al., 1995). Hexamidine was found to be more effec- bacterial and Parachlamydia strains associated with Acan-
tive than propamidine, though but this has been recently thamoeba. Erythromycin plus a second antimicrobial (ce-
disputed (Seal, 2003). Myristamidopropyl dimethylamine furoxime or ceftazidime) were effective in patient treatment
(MAPD) has been found to be effective in vitro against (Marrie et al., 2001). An in vitro study of antimicrobial
both trophic and cystic stages of Acanthamoeba (Kilvington susceptibility of two strains of P. acanthamoeba derived
et al., 2002), and did not select for resistance in amoeba from amoebae compared the amoeba isolates with other
populations that survived exposure to low concentrations of Chlamydia spp. The acanthamoebal isolates were resistant
MAPD (Schuster et al., 2003). But for many drugs tested to ␤-lactam and fluoroquinolone antibiotics, but were sen-
against amoeba isolates, in vitro activity was no guarantee sitive to aminoglycosides and cotrimoxazole (Maurin et al.,
of in vivo efficacy in amoebic keratitis (Elder et al., 1994; 2002).
46 F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51

Table 3
3. Naegleria fowleri
Successful therapy in treatment of primary amoebic meningoencephalitis
caused by Naegleria fowleri
Over 30 species of Naegleria have been identified on the
Antimicrobials Dosage Reference
basis of sequencing data (De Jonckheere, 2002). Only one
species, N. fowleri, has been isolated from human infections, Amphotericin B 0.75 mg/kg qd IV Apley et al. (1970)
although several others are pathogenic in the mouse model. Amphotericin B 0.1 mg 5 times Brown (1991)
The amoeba has a life-cycle that includes amebic and cys- per day IT
tic stages and, for most species, a transient flagellate stage Rifampin 600 mg q12h
that develops from the vegetative ameba. The organism is Amphotericin B 1 mg/kg qd Jain et al. (2002)
the causal agent of primary amoebic meningoencephaltis Rifampin 450 mg qd po
(PAM) in immunocompetent children and young adults. The Ornidazole 500 mg q8h
infection is acquired by swimming or bathing in warm fresh Amphotericin B 1.5 mg/kg q12h IV Seidel et al. (1982)
waters, such as lakes, ponds, hot springs, and thermally pol- 1.5 mg per day IT
Miconazole 350 mg/m2 q8h IV
luted rivers or streams. Trophic amoebae, cysts (or even
10 mg per day IT
flagellates; Singh and Das, 1972) in the water enter into the Rifampin 10 mg/kg q8h po
nasal passages, penetrate the nasal epithelium, and migrate Sulfisoxazole 1 g q6h IV
along the olfactory nerves to the brain. PAM is a fulminant Amphotericin B 60 mg qd Wang et al. (1993)
disease, almost invariably fatal in a matter of days. Because Rifampin 450 mg qd
the disease has a short incubation period, and runs its course Chloramphenicol 1 g qid
within 7–10 days, early diagnosis and initiation of antimi- Dosages noted in table are initial drug levels used in treatment and were
crobial therapy are critical to patient survival. Diagnosis of reduced with time.
PAM is by microscopic identification and/or cultivation of
amoebae from freshly drawn CSF (Schuster, 2002).
treating PAM follow. In a rare instance of PAM in an
3.1. Antimicrobial therapy for PAM immunocompromised patient with systemic lupus erythe-
matosus, high concentrations of IV and IT AMB started on
The drug of choice in treating PAM is the antifungal hospital day 9 (IV at 0.5, and IT at 1.0 mg/kg) cleared the
polyene antibiotic AMB. Naegleria amoebae are highly CSF of amoebae, but the patient died on hospital day 40
sensitive to the drug, with a minimal amoebicidal concen- with amoebae present in the CSF (Shrestha et al., 2003).
tration of 0.026–0.078 ␮g/ml (Duma et al., 1971). But drug Similar clearing of amoebae from the CSF of an adult male
treatment must be started early in order to be effective and followed IT and IV administration of AMB not until hospi-
there are only a handful of recoveries published in the litera- tal day 3, and the patient expired on day 9 with numerous
ture out of an estimated 200 cases (Anderson and Jamieson, amoebae having reappeared in the CSF (Lawande et al.,
1972; Apley et al., 1970; Brown, 1991; Jain et al., 2002; 1980). Intravenous AMB (0.6 mg/kg of body weight) and
Seidel et al., 1982; Wang et al., 1993). In a well-documented ceftriaxone (100 mg/kg) were used unsuccessfully to treat a
recovery, the patient was given intravenous (IV) and in- pediatric case of PAM (Shenoy et al., 2002). In none of these
trathecal (IT) AMB, IV and IT miconazole, and oral ri- three cases was follow-up in vitro testing of the amoeba
fampin (Seidel et al., 1982). In vitro testing of the isolate done.
from the patient indicated a synergistic or additive effect of In addition to AMB, other antifungal drugs have been
AMB and miconazole, but found no efficacy for rifampin tested against Naegleria isolates in vitro and in mice in-
(Seidel et al., 1982). Amphotericin B, rifampin, and ornida- fected with the amoebae. Miconazole and ketoconazole were
zole were used successfully to treat a suspected PAM case shown in vitro to be active against N. fowleri, but not in the
(Jain et al., 2002). Unlike Acanthamoeba (and Balamuthia, mouse model (Elmsly et al., 1980). As noted above, micona-
see following section), which encyst in tissues, Naegleria zole, which is not available in the United States, was used
remains in the trophic form in brain tissue and, once amoe- in conjunction with AMB in successful treatment of PAM
bae are destroyed by effective antimicrobial treatment, the (Seidel et al., 1982), but it is not clear if it was the combi-
infection should not recur. Antimicrobial regimens for sev- nation of the two drugs or AMB alone that was effective.
eral cases of successful treatment of PAM are summarized in AMB, clotrimazole, and miconazole were found effective in
Table 3. vitro against N. fowleri (Duma and Finley, 1976). A com-
In cases where intensive AMB therapy was employed, parison of AMB with ketoconazole, fluconazole, and itra-
death of the patient is assumed to be due to the pathophys- conazole in vitro, found clinical isolates of N. fowleri to be
iology of disease or to delay in starting therapy, and not to most sensitive to ketoconazole and AMB, but less so to the
resistance to AMB. Naegleria isolates from deceased PAM triazoles itraconazole and fluconazole (Tiewcharoen et al.,
patients have all shown sensitivity to amphotericin B when 2002). A drawback in the use of the AMB is the risk of
tested in vitro (Duma and Finley, 1976; Tiewcharoen et al., impaired kidney function following drug use. In comparing
2002). Several examples of unsuccessful use of AMB in AMB with liposomal AMB, a less toxic form of the drug,
F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51 47

the MIC for AMB was 0.1 ␮g/ml, while that of liposomal boon that had died in a zoological park in ∼1985, and was
AMB was 1 ␮g/ml (Goswick and Brenner, 2003b). Liposo- soon recognized in human and other animals as the cause
mal AMB, however, was found less effective in the mouse of cutaneous, nasopharyngeal, and central nervous system
model and in vitro testing than the more toxic form of AMB. infections (Martinez and Visvesvara, 1997). All isolates are
AMB methyl ester, another AMB derivative less toxic than morphologically and genomically alike, and are members of
the parent compound, was also found to be less effective in a single species. The earliest reported cases were in immuno-
the mouse model (Ferrante, 1982). suppressed, immunocompromised, or debilitated hosts, but
AMB targets the plasma membrane, as noted in the sec- later cases have occurred in immunocompetent children. We
tion on Acanthamoeba. The drug acts by binding to ergos- are aware of more than 100 cases on record at the Centers
terol in the membrane, producing pores and causing loss of for Disease Control and in the literature since the organism’s
small molecules (Sande and Mandell, 1985). Few studies recognition, all but a few being fatal. Like acanthamoebi-
have been done on the sterol components of the Naegleria asis, balamuthiasis is an insidious disease taking up to 2
membrane, and none on the membrane of N. fowleri. Cy- years to develop in the host. Diagnosis is difficult and is
cloartenol is the ergosterol precursor in N. lovaniensis; in based upon finding amoebae in biopsied brain and other tis-
N. gruberi, the precursors are cycloartenol, lanosterol, and sues, or by immunofluorescence staining for Balamuthia an-
parkeol (Raederstorff and Rohmer, 1987). In an ultrastruc- tibodies in patient serum (Schuster et al., 2001; Visvesvara
tural study of the effect of AMB on N. fowleri and N. gruberi, et al., 1993). The amoeba has also been isolated from biop-
the drug produced distortions of nuclear shape, increase in sied brain tissue into tissue culture using monkey kidney or
cytoplasmic membranes (rough and smooth varieties), mito- other cell-types as a feeder layer (Schuster, 2002). Because
chondrial abnormalities, appearance of autophagic vacuoles, of lack of familiarity with the amoeba, it is likely that cases
and plasma membrane blebbing (Schuster and Rechthand, are undiagnosed and the disease underreported. Most diag-
1975). These changes in the cell became more pronounced noses are made postmortem.
with duration of exposure and increased concentration of
AMB. 4.1. Antimicrobial therapy for Balamuthia infections
The macrolide azithromycin has both in vitro and in
vivo (mouse model) efficacy against pathogenic Naegleria There are few successful treatments for Balamuthia
(Goswick and Brenner, 2003a; Schuster et al., 2001). Clar- amoebic encephalitis (BAE). If the infection can be rec-
ithromycin was less effective than azithromycin in vitro, ognized early in its course, there is an opportunity for
and erythromycin was minimally effective (Schuster et al., antimicrobial intervention. Two such cases reported in the
2001). As noted earlier, azithromycin levels in the cere- literature had successful outcomes, both having used sim-
brospinal fluid have been found to be low (Jaruratanasirikul ilar combinations of antimicrobial agents (Deetz et al.,
et al., 1996). 2003). The drugs used included flucytosine, pentamidine
Like clinical Acanthamoeba isolates, variations in viru- isethionate, fluconazole, sulfadiazine, a macrolide antibi-
lence exist among N. fowleri isolates. Cytopathogenicity for otic (azithromycin or clarithromycin), and phenothiazines
tissue culture cells is often regarded as an indicator of viru- (thioridazide or trifluoperazine). Amoebae, however, were
lence (John and John, 1989, 1994). Differences in virulence not isolated from either patient for in vitro drug sensitiv-
are also reflected in mortality in the mouse model (Wong ity testing. In vitro results on drug activity against other
et al., 1977). The amoeba isolated from the successfully clinical isolates of Balamuthia found pentamidine isethion-
treated PAM case mentioned above (Seidel et al., 1982), was ate > azithromycin > amphotericin B to be most effective in
found to be among the least cytopathic for monkey kidney that order, while fluconazole, flucytosine, and sulfadiazine
cell cultures among several N. fowleri isolates, with 7 days had limited efficacy (Schuster and Visvesvara, unpublished
required for destruction of the tissue culture monolayer as observations). It may be that in vivo the drugs have syner-
compared to 2–6 days for others (John and John, 1989). gistic effects that are not apparent in vitro. A concern in
Furthermore, it was also the most sensitive to phenothiazine these two cases was for recurrence of the infection as a
compounds of several strains tested in vitro (Schuster and result of reactivation of dormant cysts in brain lesions. Both
Mandel, 1984). A possible conclusion is that a combination patients, however, have shown no evidence of reactivation
of low virulence and greater drug sensitivity of the isolate of disease over periods of about 5 and 7 years, although they
were factors in survival of the patient in that particular remain on fluconazole plus sulfadiazine or clarithromycin
case. (Deetz et al., 2003). The drug regimens for the two cases, a
64-year-old male and a 5-year-old female, are summarized
in Table 4.
4. Balamuthia mandrillaris Pentamidine isethionate has been used in treatment of
CNS infections by Acanthamoeba (see above) as well as
Balamuthia, like Acanthamoeba, has trophic and cystic Balamuthia. While demonstrably effective in vitro against
stages in its life-cycle (Visvesvara et al., 1993). The amoeba Balamuthia amoebae, it was reported to have relatively poor
was first isolated from the brain of a pregnant mandrill ba- penetration into the CNS when tested in HIV/AIDS patients
48 F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51

Table 4
5. Sappinia diploidea
Successful treatment of two patients with confirmed Balamuthia amoebic
encephalitis
A unique case of CNS infection with S. diploidea in an im-
Antimicrobials Dosage
munocompetent individual was treated with azithromycin,
Patient: 64-year-old male pentamidine, itraconazole, and flucytosine following surgi-
Flucytosine 2 g q6h cal excision of the necrotic lesion. The infection may have
Fluconazole 400 mg qd
Pentamidine isethionatea 4 mg/kg qd IV
developed from an earlier sinus infection. The patient re-
Sulfadiazine 1.5 g q6h covered, apparently without neurologic impairment (Gelman
Azithromycin, clarithromycinb 500 mg qd et al., 2003).
Trifluoperazine 10 mg q12h
Patient: 5-year-old female
Flucytosine 110 mg/kg qd 6. Conclusions
Fluconazole 14 mg/kg qd
Pentamidine isethionatea 1 mg/kg qd
Progress has been made in treatment of encephalitis and
Azithromycin, clarithromycinb 14 mg/kg qd
Thioridizine 1 mg/kg qd related infections caused by the free-living amoebae Acan-
thamoeba and Balamuthia. While new drugs and combina-
For detailed account of drug therapy and side effects, see Deetz et al.
tions of drugs have been tried in clinical situations, these
(2003).
a Discontinued because of developing hyperglycemia. amoebic diseases continue to have a high mortality. Part of
b Switched from azithromycin to clarithromycin because of concern the reason is due to difficulty in diagnosing the diseases,
about poor penetration into the CNS, and side effects (elevated creatinine and the resultant delay in initiating effective therapy. More
level, possible nephritis). often than not, diagnosis is made at autopsy. It is likely that
amoebic encephalitides are underreported, given the lack of
familiarity of physicians and pathologists with the diseases
(Donnelly et al., 1988). Its use had to be discontinued in and their causal agents. For the most part, combinations of
both cases cited because of side effects. In its favor, pen- drugs have been used in treatment and, while mortality re-
tamidine was shown to concentrate within hemoflagellate mains high, successful recoveries have been published in the
and malaria parasites through selective membrane transport literature. Any indications of drug resistance are hidden by
mechanisms and, despite extensive prophylactic use of the the multidrug approach used in treating infections. Naegleria
drug in parts of Africa, development of resistance was rare fowleri, the causal agent of meningoencephalitis, is sensi-
(Bray et al., 2003). Diamidines with improved ability to tive to amphotericin B and other antifungal compounds, but
cross the blood-brain barrier and lower toxicity would be the fulminant nature of the disease, coupled with diagnostic
highly desirable in treating amoebic and other CNS infec- delay, is often a critical factor in determining outcome.
tions (Bray et al., 2003). Acanthamoeba keratitis is a discrete infection that is more
There is no information on the membrane composition amenable to antimicrobial treatment than systemic infec-
of Balamuthia amoebae. Because of its close relationship tions. More readily recognized because of its immediate ef-
to Acanthamoeba (Booton et al., 2003), it may also have fect on the comfort and well-being of patients, the infection
similar membrane sterols (ergosterol and its precursor cy- responds favorably to treatment with cationic disinfectants
cloartenol) (Raederstorff and Rohmer, 1985). Clinical iso- (chlorhexidine, polyhexamethylene biguanide), which have
lates show variable in vitro susceptibility to amphotericin B, now become the drugs most frequently used in treatment.
but are relatively insensitive to the triazole compound, flu- Still, new compounds with amoebacidal and cysticidal ac-
conazole (Schuster, unpublished observations). More testing tivity are needed for use in the few situations where drug
with other azoles is needed to obtain a sensitivity profile of resistance is encountered. Myristamidopropyl dimethy-
these membrane-active compounds. lamine and hexadecylphosphosphocholine (miltefosine) are
By far the majority of cases (∼98%) on record, whether two compounds that show promise for use in recalcitrant
BAE, cutaneous or nasopharyngeal infections, have been keratitis infections, but they have yet to be tested clinically.
fatal. Even with antemortem diagnosis of BAE, initiation of The recognition of an unusual soil amoeba, S. diploidea,
effective antimicrobial therapy may come too late to help as the cause of a brain lesion indicates that other free-living
the patient (Bakardjiev et al., 2003). The use of flucytosine, amoebae are capable of causing infections in humans, and
fluconazole, pentamidine, and clarithromycin at regular or these can pose challenges to the development of effective
high dosages was ineffective in four Argentinean pediatric antimicrobial therapy.
BAE cases (Galarza et al., 2002). Miltefosine (hexade-
cylphosphocholine) was tested against clinical isolates of References
Balamuthia and has minimal inhibitory concentrations of
30–40 ␮M (Schuster, unpublished data). Its ability to cross Aksozek, A., McClellan, K., Howard, K., Niederkorn, J.Y., Alizadeh,
the blood-brain barrier would make it a potential drug for H., 2002. Resistance of Acanthamoeba castellanii cysts to physical,
treating BAE. chemical and radiological conditions. J. Parasitol. 88, 621–623.
F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51 49

Anderson, K., Jamieson, A., 1972. Primary amoebic meningoencephalitis. Fritsche, T.R., Horn, M., Seyedirashti, S., Gautom, R.K., Schleifer, K.-H.,
Lancet 1, 902–903. Wagner, M., 1999. In situ detection of novel bacterial endosymbionts of
Apley, J., Clarke, S.K.R., Roome, A.P.C.H., Sandry, S.A., Saygi, G., Silk, Acanthamoeba spp. phylogenetically related to members of the Order
B., Warhurst, D.C., 1970. Primary amoebic meningoencephalitis in Rickettsiales. Appl. Environ. Microbiol. 65, 206–212.
Britain. Br. Med. J. 1, 596–599. Galarza, M., Cuccia, V., Sosa, F.P., Monges, J.A., 2002. Pediatric
Badenoch, P.R., Adams, M., Coster, D.J., 1995. Corneal virulence, granulomatous cerebral amebiasis: a delayed diagnosis. Pediatr. Neurol.
cytophatic effect on human keratocytes and genetic characterization of 26, 153–156.
Acanthamoeba. Int. J. Parasitol. 25, 229–239. Gatti, S., Cevini, C., Bruno, A., Penso, G., Rama, P., Scaglia, M., 1998.
Badenoch, P.R., Johnson, A.M., Christy, P.E., Coster, D.J., 1990. In vitro effectiveness of povidone-iodine on Acanthamoeba isolates
Pathogenicity of Acanthamoeba and a corynebacterium in the rat from human cornea. Antimicrob. Agents Chemother. 42, 2232–2234.
cornea. Arch. Ophthalmol. 108, 107–112. Gelman, B.B., Popov, V., Chaljub, G., Nader, R., Rauf, S.J., Nauta, H.W.,
Bakardjiev, A., Azimi, P.H., Ashouri, N., Ascher, D.P., Janner, D., Visvesvara, G.S., 2003. Neuropatholgical and ultrastructural features
Schuster, F.L., Visvesvara, G.S., Glaser, C., 2003. Amebic encephalitis of amebic encephalitis caused by Sappinia diploidea. J. Neuropathol.
caused by Balamuthia mandrillaris: report of four cases. Pediatr. Infect. Exp. Neurol. 62, 990–998.
Dis. 22, 447–452. Goswick, S.M., Brenner, G.M., 2003a. Activities of azithromycin and
Berger, S.T., Mondino, B.J., Hoft, R.H., Donzis, P.B., Holland, G.N., amphotericin B against Naegleria fowleri in vitro and in a mouse
Farley, M.K., Levenson, J.E., 1990. Successful medical management model of primary amebic meningoencephalitis. Antimicrob. Agents
of Acanthamoeba keratitis. Am. J. Ophthalmol. 110, 395–403. Chemother. 47, 524–528.
Booton, G.C., Carmichael, J.R., Visvesvara, G.S., Byers, T.J., Fuerst, Goswick, S.M., Brenner, G.M., 2003b. Activities of therapeutic agents
P.A., 2003. Genotyping of Balamuthia mandrillaris based on nuclear against Naegleria fowleri in vitro and in a mouse model of primary
18S and mitochondrial 16S rRNA genes. Am. J. Trop. Med. Hyg. 68, amebic meningoencephalitis. J. Parasitol. 89, 837–842.
65–69. Gullet, J., Mills, J., Hadley, K., Podemski, B., Pitts, L., Gelber, R., 1979.
Bray, P.G., Barrett, M.P., Ward, S.A., de Kooning, H.P., 2003. Pentamidine Disseminated granulomatous Acanthamoeba infection presenting as an
uptake and resistance in pathogenic protozoa: past, present and future. unusual skin lesion. Am. J. Med. 67, 891–896.
Trends Parasitol. 19, 232–239. Hay, J., Kirkness, C.M., Seal, D.V., Wright, P., 1994. Drug resistance
Brown, R.L., 1991. Successful treatment of primary amebic meningo- and Acanthamoeba keratitis: the quest for alternative antiprotozoal
encephalitis. Arch. Intern. Med. 151, 1201–1202. chemotherapy. Eye 8, 555–563.
Cleland, P.G., Lawande, R.V., Onyemelukwe, G., Whittle, H.C., 1982. He, Y.G., McCulley, J.P., Alizadeh, H., Pidherney, M., Mellon, J.,
Chronic amebic meningoencephalitis. Arch. Neurol. 39, 56–57. Ubelaker, J.E., Stewart, G.L., Silvany, R.E., Niederkorn, J.Y., 1992. A
Deetz, T.R., Sawyer, T.H., Billman, G., Schuster, F.L., Visvesvara, pig model of Acanthamoeba keratitis: transmission via contaminated
G.S., 2003. Successful treatment of Balamuthia amoebic encephalitis. contact lenses. Invest. Ophthalmol. 33, 126–133.
Presentation of 2 casees. Clin. Infect. Dis. 37, 1304–1312. Helton, J., Loveless, M., White, Jr., C.R., 1993. Cutaneous Acanthamoeba
De Jonckheere, J., 1980. Growth characteristics, cytopathic effect in cell infection associated with leukocytoclastic vasculitis in an AIDS patient.
culture, and virulence in mice of 36 type strains belonging to 19 Am. J. Dermatopathol. 15, 146–149.
different Acanthamoeba spp. Appl. Environ. Microbiol. 39, 681–685. Hirst, L.W., Green, W.R., Merz, W., Kaufmann, C., Visvesvara, G.S.,
De Jonckheere, J.F., 2002. A century of research on the amoebofiagellate Jensen, A., Howard, M., 1984. Management of Acanthamoeba keratitis:
genus Naegleria. Acta Protozoologica 41, 309–342. a case report and review of the literature. Ophthalmology 1, 1105–
Donnelly, H., Bernard, E.M., Rothkotter, H., Gold, J.W.M., Armstrong, 1111.
D., 1988. Distribution of pentamidine in patients with AIDS. J. Infect. Horn, M., Wagner, M., Müller, K.-D., Schmid, E.N., Fritsche, T.R.,
Dis. 157, 985–989. Schleifer, K.-H., Michel, R., 2000. Neochlamydia hartmannellae gen.
Driebe, Jr., W.T., Stern, G.A., Epstein, R.J., Visvesvara, G.S., Adi, M., nov., sp. nov. (Parachlamydiaceae), an endoparasite of the amoeba
Komadina, T., 1988. Acanthamoeba keratitis. Potential role for topical Hartmannella vermiformis. Microbiology 146, 1231–1239.
clotrimazole in combination chemotherapy. Arch. Ophthalmol. 106, Hunt, S.J., Reed, S., Mathews, W.C., Torian, B., 1995. Cutaenous
1196–1201. Acanthamoeba infection in the acquired immunodeficiency syndrome:
Duma, R.J., Finley, R., 1976. In vitro susceptibility of pathogenic response to multidrug therapy. Cutis 56, 285–287.
Naegleria and Acanthamoeba species to a variety of therapeutic agents. Ishibashi, Y., Matsumoto, Y., Kabata, T., Watanabe, R., Hommura,
Antimicrob. Agents Chemother. 10, 370–376. S., Yasuraoka, K., Ishii, K., 1990. Oral itraconazole, and topical
Duma, R.J., Rosenblum, W.I., McGhee, R.F., Jones, M.M., Nelson, E.C., miconazole, with débridement for Acanthamoeba keratitis. Am. J.
1971. Primary amoebic encephalitis caused by Naegleria. Two new Ophthalmol. 109, 121–126.
cases, response to amphotericin B, and a review. Ann. Int. Med. 74, Jain, R., Prabhakar, S., Modi, M., Bhatia, R., Sehgal, R., 2002. Naegleria
923–931. meningoencephalitis: a rare survival. Neurol. India 50, 470–472.
Elder, M.J., Dart, J.K.G., 1995. Chemotherapy for Acanthamoeba keratitis. Janitschke, K., Martinez, A.J., Visvesvara, G.S., Schuster, F., 1996.
Lancet 345, 791–792. Animal model Balamuthia mandrillaris CNS infection: contrast
Elder, M.J., Kilvington, S., Dart, J.K.G., 1994. A clinicopathologic study and comparison in immunodeficient and immunocompetent mice: a
of in vitro sensitivity testing and Acanthamoeba keratitis. Invest. murine model of “granulomatous” amebic encephalitis. J. Neuropathol.
Ophthalmol. Vis. Sci. 35, 1059–1064. Exp. Neurol. 55, 815–821.
Elmsly, C.J., Donald, J.J., Brown, T.J., Keys, E.A., 1980. Chemotherapy Jarolim, K.L., McCosh, J.K., Howard, M.J., John, D.T., 2000. A light
of primary amoebic meningo-encephalitis (PAM). II. Miconazole and microscopy study of the migration of Naegleria fowleri from the nasal
R41,400 (ketoconazole). N. Z. J. Med. Lab. Technol. 34, 37–43. submucosa to the central nervous system during the early stage of
Ferrante, A., 1982. Comparative sensitivity of Naegleria fowleri to amebic meningoencephalitis in mice. J. Parasitol. 86, 50–55.
amphotericin B and amphotericin B methyl ester. Roy. Soc. Trop. Med. Jaruratanasirikul, S., Hortiwakul, R., Tantisarasart, T., Phuenpathom, N.,
Hyg. 76, 476–478. Tussanasunthornwong, S., 1996. Distribution of azithromycin into brain
Ficker, L., Seal, D., Warhurst, D., Wright, P., 1990. Acanthamoeba tissue, cerebrospinal fluid, and aqueous humor of the eye. Antimicrob.
keratitis—resistance to medical therapy. Eye 4, 835–838. Agents Chemother. 40, 825–826.
Fritsche, T.R., Gautom, R.K., Seyedirashti, S., Bergeron, D.L., Lindquist, John, D.T., John, R.A., 1989. Cytopathogenicity of Naegleria fowleri in
T.D., 1993. Occurrence of bacterial endosymbionts in Acanthamoeba mammalian cell cultures. Parasitol. Res. 76, 20–25.
spp. isolated from corneal and environmental specimens and contact John, D.T., John, R.A., 1994. Enhancement of virulence of Naegleria
lenses. J. Clin. Microbiol. 31, 1122–1126. fowleri by growth in Vero-cell cultures. J. Parasitol. 80, 149–151.
50 F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51

Karande, S.C., Lahiri, K.R., Sheth, S.S., Nadkarni, U.S., Jain, M.K., Pérez-Santonja, J.J., Kilvington, S., Hughes, R., Tufail, A., Matheson, M.,
Shah, M.D., 1991. Acanthamoeba meningoencephaliltis complicating Dart, J.K., 2003. Persistently culture positive Acanthamoeba keratitis:
pyogenic meningitis. Indian Pediatr. 28, 794–797. in vivo resistance and in vitro sensitivity. Ophthalmology 110, 1593–
Kilvington, S., Hughes, R., Byas, J., Dart, J., 2002. Activities of 1600.
therapeutic agents and myristamidopropyl dimethylamine against Perrine, D., Chenu, J.P., Georges, P., Lancelot, J.C., Saturnino, C., Robba,
Acanthamoeba isolates. Antimicrob. Agents Chemother. 46, 2007– M., 1995. Amoebicidal efficiencies of various diamidines against two
2009. strains of Acanthamoeba polyphaga. Antimicrob. Agents Chemother.
Kilvington, S., Larkin, D.F.P., White, D.G., Beeching, J.R., 1990. 39, 339–342.
Laboratory investigation of Acanthamoeba keratitis. J. Clin. Microbiol. Raederstorff, D., Rohmer, M., 1985. Sterol biosynthesis de novo via
28, 2722–2725. cycloartenol by the soil amoeba Acanthamoeba polyphaga. Biochem.
Kong, H.H., Yee, S.T., Chung, D.I., 1998. Comparison of virulence by J. 231, 609–615.
Acanthamoeba strains in a murine model of acquired immunodeficiency Raederstorff, D., Rohmer, M., 1987. Sterol biosynthesis via cycloartenol
syndrome. Korean J. Parasitol. 36, 23–31 (in Korean with English and other biochemical features related to photosynthetic phyla in the
abstract). amoebae Naegleria lovaniensis and Naegleria gruberi. Eur. J. Biochem.
Kosrirukvongs, P., Wanachiwanawin, D., Visvesvara, G.S., 1999. 164, 427–434.
Treatment of Acanthamoeba keratitis with chlorhexidine. Ophthalmo- Rivera, M.A., Padhya, T.A., 2002. Acanthamoeba: a rare primary cause
logy 106, 798–802. of rhinosinusitis. Laryngoscope 112, 1201–1203.
Lalitha, M.K., Anandi, V., Srivastava, A., Thomas, K., Cherian, A.M., Rowbotham, T.J., 1983. Isolation of Legionella pneumophila from clinical
Chandi, S.M., 1985. Isolation of Acanthamoeba culbertsoni from a specimens via amoebae, and the interaction of those and other isolates
patient with meningitis. J. Clin. Microbiol. 21, 666–667. with amoebae. J. Clin. Pathol. 36, 978–986.
Larkin, D.F.P., Kilvington, S., Dart, J.K.G., 1992. Treatment of Sande, M.A. Mandell, G.L., 1985. Antimicrobial agents: antifungal
Acanthamoeba keratitis with polyhexamethylene biguanide. Ophthal- and antiviral agents. In: Gilman, A.G., Goodman, L.S., Rall, T.W.,
mology 99, 185–191. Murad, F. (Eds.), Goodman and Gilman’s The Pharmacological Basis
Lawande, R.V., Macfarlane, J.T., Weir, W.R.C., Awunor-Renner, C., 1980. of Therapeutics, seventh ed. Macmillan Publishing Co., New York,
A case of primary amebic meningoencephalitis in a Nigerian farmer. pp. 1219–1239.
Am. J. Trop. Med. Hyg. 29, 21–25. Schuster, F.L., 1993. Comparative effects of selected azole compounds
Lim, L., Coster, D.J., Badenoch, P.R., 2000. Antimicrobial susceptibility on trophic and cystic stages of Acanthamoeba polyphaga. J. Euk.
of 19 Australian corneal isolates of Acanthamoeba. Ophthalmology Microbiol. 40, 563–569.
28, 119–124. Schuster, F.L., 2002. Cultivation of pathogenic and opportunistic free-
Lindquist, T.D., 1998. Treatment of Acanthamoeba keratitis. Cornea 17, living ameas. Clin. Microbiol. Rev. 15, 342–354.
11–16. Schuster, F.L., Buck, S., Rosenthal, R.A., Schlech, B.A., 2003. Efficacy of
Lloyd, D., Turner, N.A., Khunkitti, W., Hann, A.C., Rurr, J.R., Russell,
myristamidopropyl dimethylamine (Aldox® ) against corneal isolates
A.D., 2001. Encystation in Acanthamoeba castellanii: development of
of Acanthamoeba spp. J. Euk. Microbiol. 50 (Supplement), 520–
biocide resistance. J. Euk. Microbiol. 48, 11–16.
521.
Marciano-Cabral, F., Cabral, G., 2003. Acanthamoeba spp. as agents of
Schuster, F.L., Dunnebacke, T.H., Visvesvara, G.S., 2001. Activity of
disease in humans. Clin. Microbiol. Rev. 16, 273–307.
Marrie, T.J., Raoult, D., La Scola, B., Birtles, R.J., de Carolis, E., 2001. selected antimicrobials against clinical isolates of pathogenic free-
Legionella-like and other amoebal pathogens as agents of community- living amebas. In: Billot-Bonef, S., Cabanes, P.A., Marciano-Cabral,
acquired pneumonia. Emerg. Inf. Dis. 7, 1026–1029. F., Pernin, P., Pringuez, E. (Eds.), Proceedings of IXth International
Martinez, A.J., Duma, R.J., Nelson, E.C., Moretta, F.L., 1973. Meeting on the Biology and Pathogencity of Free-Living Amoebae,
Experimental Naegleria meningoencephalitis in mice. Penetration of John Libbey Eurotext, Paris, France, pp. 45–48.
the olfactory mucosal epithelium by Naegleria and pathological Schuster, F.L., Glaser, C., Gilliam, S., Visvesvara, G.S., 2001. Survey of
changes produced: a light and electron microscope study. Lab. Invest. sera from encephalitis patients for Balalmuthia mandrillaris antibody.
29, 121–133. J. Euk. Microbiol. 48 (Suppl.), 10S–11S.
Martinez, A.J., Visvesvara, G.S., 1997. Free-living amphizoic and Schuster, F.L., Jacob, L.S., 1992. Effects of magainins on ameba and cyst
opportunistic amebas. Brain Pathol. 7, 583–598. stages of Acanthamoeba polyphaga. Antimicrob. Agents Chemother.
Martinez, M.S., Gonzalez-Mediero, G., Santiago, P., Rodriguez De Lope, 36, 1263–1271.
A., Diz, J., Conde, C., Visvesvara, G.S., 2000. Granulomatous amebic Schuster, F.L., Mandel, N., 1984. Phenothiazine compounds inhibit in
encephalitis in a patient with AIDS: isolation of Acanthamoeba sp. vitro growth of pathogenic free-living amoebae. Antimicrob. Agents
group II from brain tissue and successful treatment with sulfadiazine Chemother. 25, 109–112.
and fluconazole. J. Clin. Microbiol. 38, 3892–3895. Schuster, F.L., Rechthand, E., 1975. In vitro effects of amphotericin B on
Maurin, M., Bryskier, A., Raoult, D., 2002. Antibiotic susceptibilities growth and ultrastructure of the amoeboflagellates Naegleria gruberi
of Parachlamydia acanthamoeba in amoebae. Antimicrob. Agents and Naegleria fowleri. Antimicrob. Agents Chemother. 8, 591–605.
Chemother. 46, 3065–3067. Schuster, F.L., Visvesvara, G.S., 1998. Efficacy of novel antimicrobials
Mehdi, H., Garg, H.S., Garg, N.K., Bhakuni, D.S., 1988. Sterols of against clinical isolates of opportunistic amebas. J. Euk. Microbiol.
Acanthamoeba culbertsoni strain A-1. Steroids 51, 551–558. 45, 612–618.
Murdoch, D., Gray, T.B., Cursons, R., Parr, D., 1998. Acanthamoeba Schuster, F.L., Visvesvara, G.S., 2003. Amebic encephalitides and amebic
keratitis in New Zealand, including two cases with in vivo resistance to keratitis caused by pathogenic and opportunistic free-living amebas.
polyhexamethylene biguanide. Aust. N. Z. Ophthalmol. J. 26, 231–236. Curr. Treatment Options Infect. Dis. 5, 273–282.
Newsome, A.L., Scott, T.M., Benson, R.F., Fields, B.S., 1998. Isolation of Seal, D.V., 2003. Acanthamoeba keratitis update—incidence, molecular
an amoeba naturally harboring a distinctive Legionella species. Appl. epidemiology and new drugs for treatment. Eye 17, 893–905.
Environ. Microbiol. 64, 1688–1693. Seal, D.V., Hay, J., Kirkness, C.M., 1995. Chlorhexidine or polyhexa-
Niszl, I.A., Veale, R.B., Markus, M.B., 1998. Cytopathogenicity of clinical methylene biguanide for Acanthamoeba keratitis. Lancet 345, 136.
and environmental Acanthamoeba isolates for two mammalian cell Seidel, J.S., Harmatz, P., Visvesvara, G.S., Cohen, A., Edwards, J., Turner,
lines. J. Parasitol. 84, 961–967. J., 1982. Successful treatment of primary amebic meningoencephalitis.
Oliva, S., Jantz, M., Tiernan, R., Cook, D.L., Judson, M.A., 1999. N. Engl. J. Med. 306, 346–348.
Successful treatment of widely disseminated acanthamoebiasis. Seifert, K., Duchêne, M., Wernsdorfer, W.H., Kollaritsch, H., Scheiner, O.,
Southern Med. J. 92, 55–57. Wiedermann, G., Hottkowitz, T., Eibl, H., 2001. Effects of miltefosine
F.L. Schuster, G.S. Visvesvara / Drug Resistance Updates 7 (2004) 41–51 51

and other akylphosphocholines on human intestinal parasite Entamoeba Tiewcharoen, S., Junnu, V., Chinabut, P., 2002. In vitro effect of antifungal
histolytica. Antimicrob. Agents Chemother. 45, 1505–1510. drugs on pathogenic Naegleria spp. Southeast Asian J. Trop. Med.
Sharma, S., Srinivasan, M., George, C., 1990. Acanthamoeba keratitis in Public Health 33, 38–41.
non-contact lens wearers. Arch. Ophthalmol. 108, 676–678. Turner, N.A., Russell, A.D., Furr, J.R., Lloyd, D., 2000. Emergence
Shenoy, S., Wilson, G., Prashanth, H.V., Vidyalakshmi, K., Dhanashree, B., of resistance to biocides during differentiation of Acanthamoeba
Bharath, R., 2002. Primary meningoencephalitis by Naegleria fowleri: castellanii. J. Antimicrob. Chemother. 46, 27–34.
first reported case from Mangalore, South India. J. Clin. Microbiol. van Klink, F., Alizadeh, H., He, Y., Mellon, J.A., Silvany, R.E., McCulley,
40, 309–310. J.P., Niederkorn, J.Y., 1993. The role of contact lenses, trauma,
Shrestha, N.K., Khanal, B., Sharma, S.K., Dhakal, S.S., Kanungo, R., and Langerhans cells in a Chinese hamster model of Acanthamoeba
2003. Primary amoebic meningoencephalitis in a patient with systemic keratitis. Invest. Ophthalmol. Vis. Sci. 344, 1937–1944.
lupus erythematosus. Scand. J. Infect. Dis. 35, 514–516. Varga, J.H., Wolf, T.C., Jensen, H.G., Parmley, V.C., Rowsey, J.J., 1993.
Singh, B.N., Das, S.R., 1972. Intra-nasal infection of mice with flagellate Combined treatment of Acanthamoeba keratitis with propamidine,
stage of Naegleria aerobia and its bearing on the epidemiology of neomycin, and polyhexamethylene biguanide. Am. J. Ophthalmol. 115,
human meningo-encephalitis. Curr. Sci. 41, 625–628. 466–470.
Singhal, T., Bajpai, A., Kalra, V., Kabra, S.K., Samantaray, J., Satpathy, G., Visvesvara, G.S., Schuster, F.L., Martinez, A.J., 1993. Balamuthia
Gupta, A.K., 2001. Successful treatment of Acanthamoeba meningitis mandrillaris, N. G., N. Sp., agent of amebic meningoencephalitis in
with combination oral antimicrobials. Pediatr. Infect Dis. J. 20, 623– humans and other animals. J. Euk. Microbiol. 40, 504–514.
627. Walochnik, J., Obwaller, A., Aspöck, H., 2000. Correlations between
Slater, C.A., Sickel, J.Z., Visvesvara, G.S., Pabico, R.C., Gaspari, A.A., morphological, molecular biological, and physiological characteristics
1994. Brief report: successful treatment of disseminated Acanthamoeba in clinical and nonclinical isolates of Acanthamoeba spp. Appl.
infection in an immunocompromised patient. N. Eng. J. Med. 331, Environ. Microbiol. 66, 4408–4413.
85–87. Walochnik, J., Duchene, M., Seifert, K., Obwaller, A., Hottkowitz, T.,
Smith, F.R., Korn, E.D., 1968. 7-Dehyrostigmasterol and ergosterol: the Wiedermann, G., Eibl, H., Aspöck, H., 2002. Cytotoxic activities of
major sterols of an amoeba. J. Lipid Res. 9, 405–408. alkylphosphocholines against clinical isolates of Acanthamoeba spp.
Stevens, A.R., Willaert, E., 1980. Drug sensitivity and resistance of four Antimicrob. Agents Chemother. 46, 695–701.
Acanthamoeba species. Trans. Roy. Soc. Trop. Med. Hyg. 74, 806–808. Wang, A., Kay, R., Poon, W.S., Ng, H.K., 1993. Successful treatment of
Stothard, D.R., Schroeder-Diedrich, J.M., Awwad, M.H., Gast, R.J., Ledee, amoebic meningoencephalitis in a Chinese living in Hong Kong. Clin.
D.R., Rodriguez-Zaragoza, S., Dean, C.L., Fuerst, P.A., Byers, T.J., Neurol. Neurosurg. 95, 249–252.
1998. The evolutionary history of the genus Acanthamoeba and the Wong, M.M., Karr, Jr., S.L., Chow, C.K., 1977. Changes in the virulence
identification of eight new 18S rRNA gene sequence types. J. Euk. of Naegleria fowleri maintained in vitro. J. Parasitol. 63, 872–878.
Microbiol. 45, 45–54. Wright, P., Warhurst, D., Jones, B.R., 1985. Acanthamoeba keratitis
Sundar, S., Jha, T.K., Thakur, C.P., Engel, J., Sindermann, H., Fischer, successfully treated medically. Br. J. Ophthalmol. 69, 778–782.
C., Junge, K., Bryceson, A., Berman, J., 2002. Oral miltefosine for Yeoh, R., Warhurst, D.C., Falcon, M.G., 1987. Acanthamoeba keratitis.
Indian visceral leishmaniasis. N. Eng. J. Med. 347, 1739–1746. Br. J. Ophthalmol. 71, 500–503.

You might also like