Naegleria fowleri: A Review of the Pathogenesis, Diagnosis, and Treatment Options

Similar documents
Case Study. Andrew L. Dunn, MD, 1 Tameika Reed, MT (ASCP), 2 Charlotte Stewart, MT (ASCP), 2 Rebecca A. Levy, MD 1,2 * ABSTRACT

Synergistic Activity of Azithromycin and Amphotericin B against Naegleria fowleri In ACCEPTED. Shannon M. Soltow* and George M.

Critical Masses. MIDG October 2015 Myra Hardy, Jeremy Carr, Tony He, Nigel Curtis

Downloaded from:

PARASITOLOGY CASE HISTORY 8 (HISTOLOGY) (Lynne S. Garcia)

Case Report Primary Amoebic Meningoencephalitis in an Iranian Infant

Section 9: Amebic Meningitis/Encephalitis

An Update in the Management of Candidiasis

Pathogenic Free-Living Amebae

FREE-LIVING PROTOZOA. PAM GAE; skin or lung lesions amebic keratitis; GAE; skin or lung lesions

LASOP Case Presentation. Andrea D Auria, D.O. University of Southern California Department of Pathology

Free living amoebae and human disease

Unusual Presentation of Multiple Nerve Palsies with Granulomatous encephalitis Due to Acanthamoeba Species

After Reading discuss the following: 1. Based on the article, will you change any of your swimming habits? Why or Why not?

Protozoa: Introduction and classification Amoebae I. Pathogenic amoebae: Entamoeba histolytica, Naegleria fowleri, Acanthamoeba spp.

Pathogenic amoebae and ciliate. Dr. Narissara Jariyapan Department of Parasitology Faculty of Medicine Chiang Mai University

B. Incorrect! Peginterferon α-2a is used for the treatment of chronic hepatitis B and may be preferable to interferon- α.

Induced encystment improves resistance to preservation and storage of Acanthamoeba castellanii

Successful Treatment of Granulomatous Amoebic Encephalitis with Combination Antimicrobial Therapy

Factors Affecting the Virulence of Naegleria fowleri for Mice

Protozoa: Introduction and classification Amoebae I. Pathogenic amoebae: Entamoeba histolytica, Naegleria fowleri, Acanthamoeba spp.

Arousal of a New, Life-Taking Threat to the World.. Naegleria Fowleri!!

CNS parasitic infections. Jarmila Klieščiková, MD 1.LF UK

LAB DETECTION OF NAEGLERIA FOWLERI: CONTINUING EDUCATION VIDEO

SCREENING OF ANTI-AMOEBIC ACTIVITY ON NAEGLERIA FOWLERI IN EXTRACTS OF THAI MEDICINAL PLANTS

MINI REVIEW PRIMARY MENINGOENCEPHALITIS CAUSED BY NAEGLERIA FOWLERI: A MINI REVIEW

Dose-dependent effects of tobramycin in an animal model of Pseudomonas sinusitis Am J Rhino Jul-Aug; 21(4):423-7

Cigna Drug and Biologic Coverage Policy

Translational PK M&S for the assessment of duration of contraceptive cover after use of miltefosine for the treatment of visceral leishmaniasis

Granulomatous amebic encephalitis in a child with acute. lymphoblastic leukemia successfully treated with

Pharmacologyonline 1: (2010) ewsletter Singh and Kochbar. Optimizing Pharmacokinetic/Pharmacodynamics Principles & Role of

ICU Volume 11 - Issue 3 - Autumn Series

MENINGITIS CRYPTOCOCCAL. learn about the symptoms, diagnosing and treating this disease

Ameba has two stages of development: cyst and trophozoite

Intranasal Medications

Aurora Health Care South Region EMS st Quarter CE Packet

Educational Introduction and Protocol for Molecular diagnosis of Acanthamoeba infection

ROSOBAC-1GM / ROSOBAC-FORT

PHARMACOKINETIC & PHARMACODYNAMIC OF ANTIBIOTICS

Drug Use Criteria: Ivacaftor (Kalydeco ) and Lumacaftor/Ivacaftor (Orkambi )

by author ESCMID Online Lecture Library Steroids in acute bacterial meningitis

Free Living Ameba Case Report

Isolation and Genotyping of Acanthamoeba Strains from Corneal Scraps

Antifungal drugs Dr. Raz Muhammed

When choosing an antiepileptic ... PRESENTATION... Pharmacokinetics of the New Antiepileptic Drugs. Based on a presentation by Barry E.

Granulomatous brain tumor caused by Acanthamoeba

Protein Synthesis Inhibitors. Ass Prof. Dr. Naza M. Ali 15 Nov 2018 Lec 8

Magnetic Resonance Imaging Findings of Eosinophilic Meningoencephalitis Caused by Angiostrongyliasis

Micafungin and Candida spp. Rationale for the EUCAST clinical breakpoints. Version February 2013

CD101: A Novel Echinocandin

Section 5.2: Pharmacokinetic properties

1. Pre-emptive therapy. colonization, colonization, pre-emptive therapy. , ICU colonization. colonization. 2, C. albicans

Risk for Transmission of Naegleria fowleri From Solid Organ Transplantation

Valproic acid. Brand Name: Depakene Drug Class: Opportunistic Infection and Other Drugs

Aspirin antagonism in isonizaid treatment of tuberculosis in mice ACCEPTED. Department of Molecular Microbiology & Immunology, Bloomberg School of

DISTRIBUTION OF PATHOGENIC NAEGLERIA SPP IN THAILAND

Antibiotics.. Tetracyclines. Aminoglycoside. Macrolides. Chloramphenicol

BIOCYT. Revista BIOCYT es editada en la Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México

%T MIC MIC. Pharmacokinetics PK: Cmax AUC T1/2 Pharmacodynamics PD: MIC: minimum inhibitory concentration time-killing-curve 1990.

Pharmaceutical Chemistry II. Antifungal Agents. = Antimycotics. Tutorial 1

Continuous Infusion of Antibiotics In The ICU: What Is Proven? Professor of Medicine Vice-Chairman, Department of Medicine SUNY at Stony Brook

Voriconazole. Voriconazole VRCZ ITCZ

Fungi are eukaryotic With rigid cell walls composed largely of chitin rather than peptidoglycan (a characteristic component of most bacterial cell

In vitro assessment of dual drug combinations to inhibit growth of Neisseria gonorrhoeae

ORIGINAL ARTICLE DOI: (e) ISSN Online: (p) ISSN Print: Anand Kumar Singh 1, Poonam Verma 2. Sciences, Dehradun

Synergists from Portulaca oleracea with macrolides against methicillin-resistant Staphylococcus aureus and related mechanism

Asian Journal of Pharmaceutical Analysis and Medicinal Chemistry Journal home page:

Macrolides & Ketolides. Objectives. Protein Synthesis

granulomatous infectious disease in an immunosuppressed patient a case report with radiological and pathological correlation

In vitro and Intracellular Activities of Peptide Deformylase. Inhibitor GSK against Legionella pneumophila Isolates

SMe. Me NITROIMIDAZOLES FOR VISCERAL LEISHMANIASIS FEXINIDAZOLE AND VL-2098 SHYAM SUNDAR

Brain-eating amoebae: Predilection sites in the brain and disease outcome. Timothy Yu Yee Ong,^ Naveed Ahmed Khan,^* Ruqaiyyah Siddiqui

Pharmacotherapy Issues in the Pediatric Population

Challenges in Therapeutic Drug Monitoring:

Q-FEVER Q FEVER. CPMP/4048/01, rev. 3 1/7 EMEA 2002

BUGS and DRUGS Part 2 March 13, 2013 Marieke Kruidering- Hall

Mesporin TM. Ceftriaxone sodium. Rapid onset, sustained action, for a broad spectrum of infections

Antifungal Treatment in Neonates

104 MMWR December 17, 2004

Xifaxan. Xifaxan (rifaximin) Description

CNS Infections in the Pediatric Age Group

Challenges and controversies of Invasive fungal Infections

Non-rifampin rifamycins in TB/HIV

Upper Respiratory Tract Infections

Waterborne Pathogens from Non-Human Sources and their Public Health Implications

Clarithromycin significantly increases linezolid serum concentrations

Nancy Nazaire-Bermal,MD Fellow PPS, PIDSP

1. Developed January Revised September 2017; August 2015; December 2013; February 2012; April 2010; March 2007.

Counties in the top and bottom two quintiles of both diabetes and obesity, Age-adjusted percentage of adults aged 20 years who are obese, 2007

Vancomycin: Class: Antibiotic.

Ideas for Educating Clinicians Ideas for Future Clinical Trials

ESCMID Online Lecture Library. by author

WHITE PAPER: A GUIDE TO UNDERSTANDING SUBARACHNOID HEMORRHAGE

Antifungal Update 2/22/12. Which is the most appropriate initial empirical therapy in a candidemic patient?

Parasitic Protozoa, Helminths, and Arthropod Vectors

Antifungal Pharmacodynamics A Strategy to Optimize Efficacy

Giardiasis Surveillance Protocol

GUIDELINE FOR THE MANAGEMENT OF MENINGITIS. All children with suspected or confirmed meningitis

Transcription:

AAC Accepted Manuscript Posted Online 10 August 2015 Antimicrob. Agents Chemother. doi:10.1128/aac.01293-15 Copyright 2015, American Society for Microbiology. All Rights Reserved. 1 2 Naegleria fowleri: A Review of the Pathogenesis, Diagnosis, and Treatment Options 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 Eddie Grace, Pharm.D. ψ, Scott Asbill, Ph.D., and Kris Virga, Ph.D. ψ Eddie Grace, Pharm.D. (Alternate corresponding author) Associate Professor of Pharmacy Practice Presbyterian College School of Pharmacy 307 N. Broad Street Clinton, SC 29325 EEGrace@presby.edu Fax: (864)938-3903 Scott Asbill, Ph.D., Chair and Professor of Pharmaceutical and Administrative Sciences Presbyterian College School of Pharmacy 307 N. Broad Street Clinton, SC 29325 CSAsbill@presby.edu Fax: (864)938-3903 Kristopher G. Virga, Ph.D., (Corresponding author) Associate Professor of Pharmaceutical Sciences Presbyterian College School of Pharmacy 1

24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 307 N. Broad Street Clinton, SC 29325 KGVirga@presby.edu Fax: (864)938-3903 Key Words: Naegleria fowleri, primary amoebic meningoencephalitis, waterborne parasite Conflict of interest: None of the authors have any conflict of interest to disclose Funding Sources: None Disclaimer: All authors have seen and approved the manuscript, contributed significantly to the work. The manuscript has not been previously published nor is not being considered for publication elsewhere. Summary: This article will review the microbiology, pathophysiology, and treatment of Naegleria fowleri. 2

46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 Abstract: Naegleria fowleri has generated tremendous media attention over the last five years due to several high profile cases. Several of these cases were followed very closely by the general public. N. fowleri is a eukaryotic, free-living amoeba belonging to the phylum Percolozoa. They are ubiquitous in the environment, being found in soil and bodies of freshwater, and feed on bacteria found in those locations. While N. fowleri infection appears to be quite rare when compared to other diseases, the clinical manifestations of primary amoebic meningoencephalitis are devastating and nearly always fatal. Due to the rarity of N. fowleri infections in humans, there are no clinical trials to date that assess the efficacy of one treatment regimen over another. Most of the information regarding medication efficacy is based either on case reports or in vitro studies. This review will discuss the pathogenesis, diagnosis, pharmacotherapy, and prevention of N. fowleri infections in humans including a brief review of all survivor cases in North America. Introduction: Naegleria fowleri has generated tremendous media attention over the last five years due to several high profile cases. Several of these cases were followed very closely by the national news outlets, as well as the general public. Unfortunately, much of the media attention served to generate public fear over public education. In this review, we will discuss the etiology, pathogenesis, case studies and treatment options for N. fowleri. Pathogenesis: N. fowleri is an amphizoic amoeba as it can survive in a free-living state in water, soil, or in the host which can be the human central nervous system (CNS) (1). N. fowleri infections have been documented in healthy children and adults following recreational water activities including swimming, diving, and water skiing. N. fowleri has been thought to infect the human body by entering the host through the nose when water is splashed or forced into the nasal cavity. Infectivity occurs first through attachment 3

70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 to the nasal mucosa followed by locomotion along the olfactory nerve, through the cribriform plate (which is more porous in children and young adults), and finally reaching the olfactory bulbs within the CNS (2). Once N. fowleri reaches the olfactory bulbs, it elicits a significant immune response through activation of the innate immune system including macrophages and neutrophils (3, 4). N. fowleri enters the human body in the trophozoite form. Structures on the surface of trophozoites known as food-cups enable the organism to ingest bacteria, fungi, and human tissue(3). In addition to tissue destruction by the food cup, the pathogenicity of N. fowleri is dependent upon the release of cytolytic molecules including, acid hydrolases, phospholipases, neuraminidases, and phospholipopolytic enzymes that play a role in host-cell and nerve destruction (1). The combination of the pathogenicity of N. fowleri and the intense immune response resulting from its presence results in significant nerve damage and subsequent CNS tissue damage, which often result in death. Diagnosis: Clinical symptoms and signs of infection with N. fowleri usually present within 2-8 days of infectivity, though some have been reported within 24 hours (5, 6). Despite the absence of specific signs and symptoms indicating N. fowleri infection, the most common symptoms include severe headache, fever, chills, positive Brudzinski sign, positive Kernig sign, photophobia, confusion, seizures, and possible coma. In addition, cardiac rhythm abnormalities and myocardial necrosis have been observed in some cases (7). Perhaps most importantly, increases in intracranial pressure and cerebral spinal fluid (CSF) pressure have been directly associated with death. CSF pressures of 600 mmh 2 O have been observed in patients with N. fowleri infection (5). CSF analysis has shown various abnormalities in color ranging from gray in the early stages of infection to red in late stage disease due to a significant increase in red blood cells (8, 9). Additional increases are seen in polymorphonuclear concentrations (as high as 26000 mm 3 ), as well as the presence of trophozoites in the CSF (using Trichome or Giemsa stains) (5, 7). Magnetic resonance 4

93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 imaging (MRI) of the brain often shows abnormalities in various regions of the brain including the midbrain and subarachnoid space (5, 7). Treatment Options: Due to the rarity of N. fowleri infections in humans, there are no clinical trials to date that assess the efficacy of one treatment regimen over another. Most of the information regarding medication efficacy is based either on case reports or in vitro studies. Perhaps the most agreed upon medication for the treatment of N. fowleri infection is amphotericin B, which has been studied in vitro and also used in several case reports. Other anti-infectives, which have been used in case reports include, fluconazole, miconazole, miltefosine, azithromycin, and rifampin. Various other agents have been studied in vitro and/or in vivo, including hygromycin, rokitamycin, clarithromycin, erythromycin, roxithromycin, and zeocin (10). Amphotericin B Amphotericin B has a minimal amoebicidal concentration of 0.01 mcg/ml against N. fowleri (11, 12). However, in vitro studies have shown that an amphotericin B concentration of at least 0.1 mcg/ml was needed to suppress greater than 90% of growth, while 0.39 mcg/ml were needed to completely suppress amoeba proliferation (11). The associated minimum inhibitory concentration (MIC) to kill 100% of the organisms of amphotericin B in the in vitro studies was 0.78 mcg/ml (12). Based on these findings and the limited data from survival cases of N. fowleri, amphotericin B, whether intravenously or intrathecally, is the cornerstone of therapy and should be used with or without other adjunctive therapies. N. fowleri studies in mice have shown that amphotericin B at doses of 7.5 mg/kg/day are needed to improve survival in mice infected with N. fowleri (11). Intravenous doses of amphotericin B of 0.25-1.5 mg/kg/day are recommended in adults, while doses ranging from 0.5-0.7 mg/kg/day are recommended in pediatrics. (13) The recommended duration of therapy with amphotericin B for the treatment of N. fowleri is 10 days (14, 15). The Centers for Disease Control and Prevention (CDC) 5

117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 recommends conventional amphotericin B formulation over liposomal or amphotericin B methyl-ester as both these agents have shown to have a significantly higher MIC against N. fowleri than conventional amphotericin B (13, 16). The CDC recommends intravenous conventional amphotericin B at doses of 1.5 mg/kg/day in 2 divided doses for 3 days followed by 1 mg/kg/day once daily for an additional 11 days (total 14 days of therapy) (13). Intrathecal amphotericin B should also utilize the conventional amphotericin B formulation. The CDC recommended dose of conventional amphotericin B intrathecally is 1.5 mg/day for 2 days followed by 1 mg/day for an additional 8 days (total of 10 days of therapy) (13). Although amphotericin B has become the primary drug of choice for treating primary amebic meningoencephalitis (PAM), its use is associated with multiple side-effects including use-limiting renal toxicity (17). Much of the problems with amphotericin B can be linked to its lack of aqueous solubility, which affects dissolution, compartmental concentration, and clearance. Recently, corifungin, which is described as a new drug entity, was granted orphan drug status for the treatment of PAM (18). Initial reports for the in vivo efficacy of corifungin in a mouse model of PAM showed superior activity to that of amphotericin B at equivalent dosing (18). Chemically, corifungin is the sodium salt of amphotericin B with excellent aqueous solubility (> 100 mg/ml) (18). The increased solubility of corifungin is likely to account for the described increase in activity. Unfortunately, human studies have not yet been reported to determine whether the increased solubility will translate into therapeutic benefits such as enhanced blood-brain-barrier penetration and/or reduced renal toxicity. Miltefosine In 2013, two patients infected with N. fowleri survived after being treated with miltefosine (discussed in further detail in a later section). While these results are limited, they show promise for the use of miltefosine when early diagnosis is made. Chemically, miltefosine is a phospholipid with an attached choline; an alkylphosphocholine (figure 1). The overall molecule is amphiphilic, with a polar phosphocholine head region and an aliphatic tail. It also exists in the zwitterionic form resulting from 6

141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 the permanently charged quaternary ammonium ion, as well as the anionic phosphate. The reported mechanism of action for miltefosine is inhibitory action against protein kinase B, PKB or Akt. This mechanism stands to reason miltefosine s interest and approval as an anticancer agent due to the role of phosphatidylinositol 3-kinase (PI3K) and PKB in cell survival (19). Miltefosine is still undergoing studies to determine absolute bioavailability in humans. It has demonstrated favorable (>80%) oral bioavailability in rodents and dogs (19). Evidence thus far, indicates both a passive and active transport mechanism of miltefosine absorption following oral administration (20). Miltefosine demonstrates a high level of plasma protein binding (>95%) and also has wide tissue distribution in rodent models with the highest concentration of drug being found in organ tissues; lung, adrenal glands, spleen, and liver. It is difficult to rationalize the CNS penetration of miltefosine, as a permanently charged species, without an active transport mechanism. Specific studies of miltefosine concentrations in brain have not been described, but its effective use in N. fowleri infection demonstrates some level of penetration. It is not clearly understood if the blood brain barrier of these patients has been in any way compromised due to the infection. Metabolism studies from human subjects have not been determined. However, miltefosine has been measured for oxidative metabolism against a panel of CYP450 isoforms, as well as the ability to induce isoforms of CYP3A. None of those evaluations predicted a significant degree of metabolism or potential for induction, suggesting the risk for drug-drug interactions would be low (21). It is actually suspected that the breakdown products of miltefosine result from the activity of phospholipases. The primary metabolites produced in animal studies have been choline, phosphocholine, and cetyl alcohol. The cetyl alcohol is oxidized to palmitic acid (see figure 2). Each of these metabolic byproducts is naturally occurring and likely gets utilized in biosynthetic processes (19). Miltefosine has shown in vitro efficacy against N. fowleri suggesting a possible benefit in treatment (22). In vitro studies have shown that N. fowleri amoebas have survived in miltefosine concentrations of 40 mcg but not 80 mcg (22). Using incremental increases in concentrations of miltefosine, a concentration 7

165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 of 40 mcg can be considered amoebostatic (representing the minimum inhibitory concentration) while concentrations of 55 mcg are amoebicidal (representing the minimum amoebicidal concentration), thus killing all amoebas exposed to this concentration of miltefosine (22). The CDC has made miltefosine available on a need basis through an investigational new drug (IND) protocol for the treatment of infections caused by free-living amoebas, which include N. fowleri, Balamuthia mandrillari, and Acanthamoeba species (23). The CDC recommends miltefosine at doses of 50 mg orally twice to three times daily (based on body weight) with a maximum dose of 1.5 mg/kg/day for a total of 28 days (23). Adjunctive Therapies Fluconazole Fluconazole, an azole antifungal agent, has been used in conjunction with Amphotericin B in the treatment of some cases of N. fowleri (24). Addition of fluconazole has shown to provide some additional benefit to amphotericin B therapy (25-27). Fluconazole s efficacy may be due to its increased penetration into the CNS. Fluconazole and amphotericin B in combination show synergistic effects on eradicating N. fowleri infection through recruitment of neutrophils (28). Based on these findings, fluconazole can be considered as an add-on therapy to amphotericin B in patients with suspected N. fowleri infections. The CDC recommends intravenous fluconazole at a dose of 10 mg/kg/day once daily (maximum dose of 600 mg/day) for a total of 28 days (13). Voriconazole, a broader spectrum azole antifungal agent, has been shown in vitro to effectively kill N. fowleri at concentrations 1 mcg/ml (22). Azithromycin Azithromycin, a macrolide antibiotic has been tested in vitro against N. fowleri and has shown to suppress greater than 90% of organism growth at concentrations of 10 and 100 mcg/ml (21). Azithromycin has shown to have an MIC of 10 mcg/ml against N. fowleri (11). In vivo studies in mice have shown that azithromycin dosed at 75 mg/kg/day was needed to prevent death in mice infected 8

188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 with N. fowleri. The CDC recommends intravenous azithromycin at doses of 10 mg/kg/day once daily (maximum 500 mg daily dose) for 28 days (13). Of note, other agents such as miconazole, hygromycin (aminoglycoside antibiotic; not available in the US), rokitamycin (macrolide antibiotic; not available in the US), clarithromycin (macrolide antibiotic), erythromycin (macrolide antibiotic), roxithromycin (macrolide antibiotic), chlorpromazine (antipsychotic), and rifampin (RNA polymerase inhibitor antibiotic) have been tested in vitro against N. fowleri showing either lack of efficacy or inconsistency with efficacy in vitro (10, 12). Rifampin Although rifampin has been used in all of PAM survivor cases in the United States and Mexico (all three cases in the US and one survivor in Mexico), its efficacy remains questionable (29, 30). The primary issue involves whether sufficient CNS penetration of rifampin at standard therapeutic dosing occurs. Multiple reports have demonstrated favorable CNS concentration of rifampin in the CNS as measured by drug concentration in CSF (31, 32). However, one report by Minderman et al. investigated compartmental concentrations of rifampin in the CNS and found significant variations (33). Concentrations in the cerebral extracellular space and within normal brain tissue were measured at 0.32 ± 0.11 mcg/ml and 0.29 ± 0.15 mcg/ml, respectively. These concentrations would exceed the required MIC for most susceptible bacteria, but may not be sufficient for eradicating N. fowleri. An initial report by Thong et al. in 1977 showed that the natural product, rifamycin, delayed the growth of N. fowleri by 30-35% at concentrations of 10 mcg/ml over a 3 day incubation period. However, rifamycin had lost its ability to inhibit N. fowleri growth by the sixth day of incubation (34). Growth inhibition (>80%) was sustained for the 6 day period only when higher concentrations of rifampin, a semi-synthetic analogue of rifamycin, (100 mcg/ml) were used. A later report by Endarza failed to demonstrate an MIC for rifampin against N. fowleri and showed an Inhibitory Concentration (IC 50 ) >32 mcg/ml, which was the maximum 9

211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 concentration evaluated in the study (35). Based on this data, there is no evidence supporting the use of rifampin at standard doses for the treatment of PAM. The secondary issue regarding the use of rifampin in the treatment of N. fowleri is the high potential for drug-drug interactions in combination therapy. Rifampin is a well-known inducer of the CYP2 and CYP3 family of monooxygenase enzymes; specifically, CYP2C9, CYP2C19, and CYP3A4 (36, 37). The greatest likelihood for interaction with rifampin lies with the 14α-demethylase inhibitors, also known as the azole fungistatics. In a majority of the treatment cases, miconazole was initially used before giving way to fluconazole use in more recent cases. 14α-demethylase is a CYP-450 isoform and obvious interactions between rifampin and fluconazole have been reported (38, 39). Co-administration of the two drugs leads to significant changes in the pharmacokinetics of fluconazole: >20% decrease in area under the concentration time curve (AUC) and as much as 50% decrease in critically ill patients; at least a 30% increase in clearance rate; and a 28% shorter half-life. Because synergy has been demonstrated between 14α-demethylase inhibitors and amphotericin B against N. fowleri, it would appear that the addition of rifampin to the combination may be of little benefit and actually work against the maximum therapeutic effect of the other agents (40). Survivor Cases To date, there have been only seven survivors world-wide, of which four survivors were in North America, including three in the United States and one in Mexico. The first case of N. fowleri survival in North America was in the US in 1978 (published in 1982), which involved a nine-year old girl who had been swimming in Deep Creek Hot Springs in the San Bernardino National Forest on two separate occasions. She was treated intravenously and intrathecally using both conventional amphotericin B and miconazole in addition to oral rifampin, intravenous dexamethasone, and oral phenytoin (41). In 2004, one survivor was reported in Mexico (30). This survivor was a 10-year old male child who developed N. fowleri infection one week after swimming in an irrigation canal. The patient was successfully treated 10

235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 using intravenous amphotericin B for 14 days in combination with rifampin and fluconazole for one month. The patient was discharged from the hospital on day 23 of therapy when the brain computed tomography showed no evidence of infection. The two most recent US cases both occurred in 2013 (29). The first case involved a 12-year old girl who was diagnosed with N. fowleri infection 7 days after visiting a water park near Little Rock, Arkansas and 2 days after the onset of symptoms (42). The patient was started on therapy on the same day she presented to the emergency department using amphotericin B (intravenously and intrathecally), miltefosine, fluconazole, rifampin, dexamethasone, and azithromycin. Additionally, her treatment included induced hypothermia to help decrease brain swelling. The patient made a full recovery following treatment (13, 42). The second case in 2013 involved an eight-year old male in the US who was treated with a combination of intrathecal and intravenous amphotericin B, rifampin, fluconazole, dexamethasone, azithromycin and miltefosine (29). The patient survived the infection but suffered from brain damage secondary to the infection. The infection had been ongoing several days prior to seeking medical attention and medically induced hypothermia was not used as in the previous case (13, 29). Prevention Steps which can be taken by individuals who participate in water related sports in warmer climates include avoidance of exposure to fresh water bodies such as lakes, rivers, and ponds especially during the summer months when the water temperature is warmer. Both chlorinated and salt-water significantly decrease the risk of N. fowleri infection due to the inability of N. fowleri to survive in such environments. If fresh water activities cannot be avoided, it is recommended that individuals avoid jumping into the body of water, splashing, or submerging their heads under the water as to avoid N. fowleri from entering the nasal passages. If such activities cannot be avoided, individuals should use nose clips to decrease the chance of contaminated water from entering the nose. Some advocate rinsing the nose and nasal passages with clean water after swimming in fresh bodies of water, however, the 11

259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 effectiveness of this method is hypothetical and unknown at this time. If water is going to be used for sinus rinsing, the CDC recommends either commercially available distilled or purified bottled water. In the absence of the aforementioned options, the CDC recommends treating water for sinus rinsing by either boiling or filtering the water using a filter with 1 micron or smaller pores. Acknowledgments: We would like to thank Drs. Marciano-Cabral and Yoder for their support and insight in reviewing this manuscript prior to submission. References: 1. De Jonckheere JF. 2011. Origin and evolution of the worldwide distributed pathogenic amoeboflagellate Naegleria fowleri. Infect Genet Evol 11:1520-1528. 2. Jarolim KL, McCosh JK, Howard MJ, John DT. 2000. A light microscopy study of the migration of Naegleria fowleri from the nasal submucosa to the central nervous system during the early stage of primary amebic meningoencephalitis in mice. J Parasitol 86:50-55. 3. Marciano-Cabral F, Cabral GA. 2007. The immune response to Naegleria fowleri amebae and pathogenesis of infection. FEMS Immunol Med Microbiol 51:243-259. 4. John DT, Cole TB, Jr., Bruner RA. 1985. Amebostomes of Naegleria fowleri. J Protozool 32:12-19. 5. Visvesvara GS, Moura H, Schuster FL. 2007. Pathogenic and opportunistic free-living amoebae: Acanthamoeba spp., Balamuthia mandrillaris, Naegleria fowleri, and Sappinia diploidea. FEMS Immunol Med Microbiol 50:1-26. 6. Craun GF. 1972. Microbiology--waterborne outbreaks. J Water Pollut Control Fed 44:1175-1182. 7. Martínez AJ. 1985. Free-living amebas: natural history, prevention, diagnosis, pathology, and treatment of disease. CRC Press. 8. Centers for Disease C. 05/28/08 2008. Primary Amebic Meningioencephalitis Arizona, Florida, and Texas 2007. Accessed 07/23. 9. Hebbar S, Bairy I, Bhaskaranand N, Upadhyaya S, Sarma MS, Shetty AK. 2005. Fatal case of Naegleria fowleri meningo-encephalitis in an infant: case report. Ann Trop Paediatr 25:223-226. 10. Kim JH, Lee YJ, Sohn HJ, Song KJ, Kwon D, Kwon MH, Im KI, Shin HJ. 2008. Therapeutic effect of rokitamycin in vitro and on experimental meningoencephalitis due to Naegleria fowleri. Int J Antimicrob Agents 32:411-417. 11. Goswick SM, Brenner GM. 2003. Activities of azithromycin and amphotericin B against Naegleria fowleri in vitro and in a mouse model of primary amebic meningoencephalitis. Antimicrob Agents Chemother 47:524-528. 12. Kim JH, Jung SY, Lee YJ, Song KJ, Kwon D, Kim K, Park S, Im KI, Shin HJ. 2008. Effect of therapeutic chemical agents in vitro and on experimental meningoencephalitis due to Naegleria fowleri. Antimicrob Agents Chemother 52:4010-4016. 13. Centers for Disease C. 5/28/2014 2014. N. Fowleri Treatment Accessed 11/25. 14. Barnett ND, Kaplan AM, Hopkin RJ, Saubolle MA, Rudinsky MF. 1996. Primary amoebic meningoencephalitis with Naegleria fowleri: clinical review. Pediatr Neurol 15:230-234. 12

298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 15. Hannisch W, Hallagan LF. 1997. Primary amebic meningoencephalitis: a review of the clinical literature. Wilderness Environ Med 8:211-213. 16. Ferrante A. 1982. Comparative sensitivity of Naegleria fowleri to amphotericin B and amphotericin B methyl ester. Trans R Soc Trop Med Hyg 76:476-478. 17. Stevens AR, Shulman ST, Lansen TA, Cichon MJ, Willaert E. 1981. Primary amoebic meningoencephalitis: a report of two cases and antibiotic and immunologic studies. J Infect Dis 143:193-199. 18. Debnath A, Tunac JB, Galindo-Gomez S, Silva-Olivares A, Shibayama M, McKerrow JH. 2012. Corifungin, a new drug lead against Naegleria, identified from a high-throughput screen. Antimicrob Agents Chemother 56:5450-5457. 19. Dorlo TP, Balasegaram M, Beijnen JH, de Vries PJ. 2012. Miltefosine: a review of its pharmacology and therapeutic efficacy in the treatment of leishmaniasis. J Antimicrob Chemother 67:2576-2597. 20. Menez C, Buyse M, Farinotti R, Barratt G. 2007. Inward translocation of the phospholipid analogue miltefosine across Caco-2 cell membranes exhibits characteristics of a carriermediated process. Lipids 42:229-240. 21. Sindermann H, Engel J. 2006. Development of miltefosine as an oral treatment for leishmaniasis. Trans R Soc Trop Med Hyg 100 Suppl 1:S17-20. 22. Schuster FL, Guglielmo BJ, Visvesvara GS. 2006. In-vitro activity of miltefosine and voriconazole on clinical isolates of free-living amebas: Balamuthia mandrillaris, Acanthamoeba spp., and Naegleria fowleri. J Eukaryot Microbiol 53:121-126. 23. Centers for Disease C. 2013. Investigations Drug Available Directly from CDC for the treatment of infections with Free-Living Amebae. MMWR CDC Surveill Summ 62:1. 24. Fowler M, Carter RF. 1965. Acute pyogenic meningitis probably due to Acanthamoeba sp.: a preliminary report. Br Med J 2:740-742. 25. Wang A, Kay R, Poon WS, Ng HK. 1993. Successful treatment of amoebic meningoencephalitis in a Chinese living in Hong Kong. Clin Neurol Neurosurg 95:249-252. 26. Loschiavo F, Ventura-Spagnolo T, Sessa E, Bramanti P. 1993. Acute primary meningoencephalitis from entamoeba Naegleria Fowleri. Report of a clinical case with a favourable outcome. Acta Neurol (Napoli) 15:333-340. 27. Brown RL. 1991. Successful treatment of primary amebic meningoencephalitis. Arch Intern Med 151:1201-1202. 28. Jacobs S, Price Evans DA, Tariq M, Al Omar NF. 2003. Fluconazole improves survival in septic shock: a randomized double-blind prospective study. Crit Care Med 31:1938-1946. 29. Capewell LG, Harris, A.M., Yoder, J.S., Cope, J.R., Eddy, B.A., Roy, S.L., Visvesvara, G.S., Fox, L.M., Beach, M.J. 2014. Diagnosis, Clinical Course, and Treatment of Primary Amoebic Meningoencephalitis in the United States, 1937 2013. J Ped Infect Dis doi:10.1093/jpids/piu103. 30. Vargas-Zepeda J, Gomez-Alcala AV, Vasquez-Morales JA, Licea-Amaya L, De Jonckheere JF, Lares-Villa F. 2005. Successful treatment of Naegleria fowleri meningoencephalitis by using intravenous amphotericin B, fluconazole and rifampicin. Arch Med Res 36:83-86. 31. Sullins AK, Abdel-Rahman SM. 2013. Pharmacokinetics of antibacterial agents in the CSF of children and adolescents. Paediatr Drugs 15:93-117. 32. Nau R, Prange HW, Menck S, Kolenda H, Visser K, Seydel JK. 1992. Penetration of rifampicin into the cerebrospinal fluid of adults with uninflamed meninges. J Antimicrob Chemother 29:719-724. 33. Mindermann T, Zimmerli W, Gratzl O. 1998. Rifampin concentrations in various compartments of the human brain: a novel method for determining drug levels in the cerebral extracellular space. Antimicrob Agents Chemother 42:2626-2629. 13

346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 34. Thong YH, Rowan-Kelly B, Shepherd C, Ferrante A. 1977. Growth inhibition of Naegleria fowleri by tetracycline, rifamycin, and miconazole. Lancet 2:876. 35. Ondarza RN, Iturbe A, Hernandez E. 2006. In vitro antiproliferative effects of neuroleptics, antimycotics and antibiotics on the human pathogens Acanthamoeba polyphaga and Naegleria fowleri. Arch Med Res 37:723-729. 36. Mahatthanatrakul W, Nontaput T, Ridtitid W, Wongnawa M, Sunbhanich M. 2007. Rifampin, a cytochrome P450 3A inducer, decreases plasma concentrations of antipsychotic risperidone in healthy volunteers. J Clin Pharm Ther 32:161-167. 37. Ridtitid W, Wongnawa M, Mahatthanatrakul W, Punyo J, Sunbhanich M. 2002. Rifampin markedly decreases plasma concentrations of praziquantel in healthy volunteers. Clin Pharmacol Ther 72:505-513. 38. Nicolau DP, Crowe HM, Nightingale CH, Quintiliani R. 1995. Rifampin-fluconazole interaction in critically ill patients. Ann Pharmacother 29:994-996. 39. Panomvana Na Ayudhya D, Thanompuangseree N, Tansuphaswadikul S. 2004. Effect of rifampicin on the pharmacokinetics of fluconazole in patients with AIDS. Clin Pharmacokinet 43:725-732. 40. Lee KK, Karr SL, Jr., Wong MM, Hoeprich PD. 1979. In vitro susceptibilities of Naegleria fowleri strain HB-1 to selected antimicrobial agents, singly and in combination. Antimicrob Agents Chemother 16:217-220. 41. Seidel JS, Harmatz P, Visvesvara GS, Cohen A, Edwards J, Turner J. 1982. Successful treatment of primary amebic meningoencephalitis. N Engl J Med 306:346-348. 42. Linam WM, Ahmed M, Cope JR, Chu C, Visvesvara GS, da Silva AJ, Qvarnstrom Y, Green J. 2015. Successful treatment of an adolescent with Naegleria fowleri primary amebic meningoencephalitis. Pediatrics 135:e744-748. 14

383 384 385 386 Figure Legend Figure 1. Miltefosine Chemical Structure Figure 2. Metabolites of Miltefosine 15