Abstract
Primary amebic meningoencephalitis, caused by brain infection with a free-living ameba Naegleria fowleri, leads to an extensive inflammation of brain and death within 3–7 days after symptoms begin. Treatment of primary amebic meningoencephalitis relies on amphotericin B in combination with other drugs, but use of amphotericin B is associated with severe adverse effects. Despite a fatality rate of over 97%, economic incentive to invest in development of antiamebic drugs by the pharmaceutical industry is lacking. Development of safe and rapidly acting drugs remains a critical unmet need to avert future deaths. Since FDA-approved anti-inflammatory and anti-arthritic drug auranofin is a known inhibitor of selenoprotein synthesis and thioredoxin reductase and the genome of N. fowleri encodes genes for both selenocysteine biosynthesis and thioredoxin reductases, we tested the effect of auranofin against N. fowleri strains of different genotypes from USA, Europe and Australia. Auranofin was equipotent against all tested strains with an EC50 of 1–2 μM. Our growth inhibition study at different time points demonstrated that auranofin is fast-acting and ~90% growth inhibition was achieved within 16 hours of drug exposure. A short exposure of N. fowleri to auranofin led to the accumulation of intracellular reactive oxygen species. This is consistent with auranofin’s role in inhibiting antioxidant pathways. Further, combination of auranofin and amphotericin B led to 95% of growth inhibition with 2- to 9-fold dose reduction for amphotericin B and 3- to 20-fold dose reduction for auranofin. Auranofin has the potential to be repurposed for the treatment of primary amebic meningoencephalitis.
Keywords: free-living ameba, Naegleria, drug, primary amebic meningoencephalitis, auranofin, amphotericin B
Graphical Abstract
INTRODUCTION
Naegleria, commonly found in water resources such as swimming pools having inadequate levels of chlorine, lakes and rivers, feed mostly on bacteria, but can also act as an opportunistic pathogen causing infections of the central nervous system (CNS). Naegleria fowleri causes severe primary amebic meningoencephalitis (PAM) and occurs disproportionately among children less than 13 years old 1 with recent recreational fresh water exposure. Only 4 people out of 145 known infected individuals in the United States from 1962 to 2019 have survived 2. N. fowleri has been listed by the National Institute of Allergy and Infectious Diseases (NIAID) as a category B priority biodefense pathogen due to their low infectious dose and potential for dissemination through compromised water supplies in the United States. Through contaminated water, N. fowleri enters nostrils and then migrates via the olfactory nerves, through the cribriform plate into olfactory bulb of the brain. The time from initial exposure to onset of illness is usually 5–7 days but may be as short as 24 hours. Initial symptoms include sudden onset of bifrontal or bitemporal headaches, high fever, nuchal rigidity, anorexia, vomiting, irritability and restlessness. Other symptoms such as photophobia, neurological abnormalities, including altered mental status, lethargy, dizziness, ataxia, cranial nerve palsy, hallucinations, delirium, coma may occur late in the clinical course, leading to death in 3 to 7 days 3.
N. fowleri infection is not a notifiable or reportable disease in the US. Thus, it is possible that the infection is underreported because several states differ in their capacity to identify, investigate or report cases. For example, among all encephalitis deaths in the US between 1989 and 1998, 86.2% were due to unknown causes 4. It is possible that lack of investigation and non-notification of PAM cases may be the reason for some of these encephalitis deaths with unknown causes. PAM kills over 97% of infected people and the high mortality rate and lack of effective therapy make PAM a particularly tragic infection for many families. This high mortality is attributed to (i) delayed diagnosis, (ii) lack of safe and effective anti-N. fowleri drugs, and (iii) difficulty of delivering drugs to the brain.
The optimum treatment for PAM has not been established. To date, amphotericin B has been used, but it is not FDA-approved for this indication and no more than a dozen people worldwide have been successfully treated with amphotericin B, either alone or in combination with other drugs 5. Recently, an investigational drug, miltefosine, has shown some promise in combination with other drugs and induced hypothermia and management of elevated intracranial pressure based on the principles of traumatic brain injury 6, 7. However, one patient, who received miltefosine did not survive the infection. Thus, the discovery of new drugs to treat this deadly disease is a critical unmet need to prevent future deaths of children and young adults.
Metal-based compounds have been tested against different neglected tropical diseases. For example, silver polypyridylcomplexes were found active against Leishmania Mexicana 8 and platinum and palladium complexes showed potent activity against Trypanosoma cruzi 9, 10. Palladium complexes exhibited better activity than the current drug metronidazole when tested against parasitic Entamoeba histolytica 11. Gold complexes have been used for several years against rheumatoid arthritis 12 and gold-containing compounds were developed as lead compounds against Trypanosoma, Leishmania and Plasmodium 13,15. Earlier we showed the amebicidal activity of a seleno-organic compound ebselen against N. fowleri 16. We identified thioredoxin reductase in N. fowleri genome sequences. Mitochondrial thioredoxin reductase of N. fowleri contains selenocysteine 17, which enhances inhibitory effect of metal-containing drugs by facilitating metal release 18. Since auranofin, an FDA-approved anti-arthritic drug is a known inhibitor of selenoprotein synthesis 19, we tested the activity of auranofin against different genotypes of N. fowleri. We then determined the killing effect of auranofin at different time points, tested the effect of auranofin on reactive oxygen species, and finally explored the effect of auranofin in combination with the current standard of care amphotericin B.
RESULTS AND DISCUSSION
In vitro activity of auranofin, amphotericin B and miltefosine against different genotypes of N. fowleri
Auranofin is an FDA-approved drug and has been in clinical use to treat rheumatoid arthritis since 1985 20. In a short communication published earlier 21, authors tested the activity of auranofin on two US strains HB-1 and Lee, which belonged to the same genotype I and only one strain (HB-1) was a human isolate 22. Since drug susceptibility varies considerably from strains to strains of different genotypes of N. fowleri 23, it is imperative to test the effect of drugs against strains of different genotypes. We tested auranofin against five human strains of various genotypes originated from different geographic regions and compared its activity with that of standards of care, amphotericin B and miltefosine. Auranofin was equally potent against European KUL, Australian CDC:V1005, US genotype I Davis, US genotype II CAMP and US genotype III TY strains with EC50 ranging between 1–2 μM. Although auranofin is less potent than amphotericin B, amphotericin B is a highly toxic drug. On the other hand, auranofin is about 15- to 60-fold more potent than CDC-recommended miltefosine (Table 1).
Table 1.
EC50 values of auranofin against different strains of N. fowleri
Drugs | Strains | EC50 (μM) | ||
---|---|---|---|---|
Mean | 95% Lower CL | 95% Upper CL | ||
Auranofin | European KUL | 2 ± 0.01 | 1.9 | 2.1 |
Australian CDC:V1005 | 2.2 ± 0.03 | 1. 9 | 2. 6 | |
US Davis (Genotype I) | 1.6 ± 0.03 | 1.4 | 1.9 | |
US CAMP (Genotype II) | 1.2 ± 0.02 | 1.1 | 1.4 | |
US TY (Genotype III) | 1.8 ± 0.01 | 1.7 | 1.9 | |
Amphotericin B | European KUL | 0.09 ± 0.03 | 0.08 | 0.1 |
Australian CDC:V1005 | 0.2 ± 0.05 | 0.1 | 0.2 | |
US Davis (Genotype I) | 0.06 ± 0.05 | 0.05 | 0.08 | |
US CAMP (Genotype II) | 0.06 ± 0.04 | 0.05 | 0.08 | |
US TY (Genotype III) | 0.06 ± 0.02 | 0.05 | 0.06 | |
Miltefosine | European KUL | 54.5 ± 0.01 | 51.4 | 57.8 |
Australian CDC:V1005 | 15.9 ± 0.09 | 10.3 | 24.7 | |
US Davis (Genotype I) | 58.9 ± 0.07 | 41.3 | 83.9 | |
US CAMP (Genotype II) US TY (Genotype III) |
21.8 ± 0.02 37.7 ± 0.02 |
19.9 33.1 |
23.8 43 |
CL = confidence limit
Auranofin earlier showed efficacy against other protozoans including the diarrheagenic parasites E. histolytica 24, Giardia lamblia 25 and Cryptosporidium parvum 26, the sexually-transmitted parasite Trichomonas vaginalis 27, and intracellular parasites Leishmania donovani 28, 29, Toxoplasma gondii 30 and T. cruzi 31. Recently, it was found to be effective against a free-living CNS-invasive ameba, Balamuthia mandrillaris 32. Our study contributes in establishing auranofin as a broad-spectrum antiparasitic agent that is effective against multiple genotypes of N. fowleri.
Since auranofin is an FDA-approved drug, which is currently used for the treatment of rheumatoid arthritis, its toxicity against multiple human cell lines is well documented. The toxicity of auranofin differs depending on the mammalian cell types used. For example, it showed a CC50 of 8.2 μM when tested against primary human foreskin fibroblasts 30. Auranofin demonstrated a CC50 of 4 μM against RAW264.7 macrophages 33. The toxicity of auranofin to human keratinocyte was found to have a CC50 of 9.4 μM 34. All these data show that auranofin has a selectivity indices ranging from 2 to 9, depending on the strain of N. fowleri and mammalian cell types examined. When tested against more relevant cell line for brain infection, auranofin did not show toxicity against human astrocytes at concentrations up to 5 μM 35. Moreover, auranofin crosses the blood-brain barrier 35, which is a prerequisite for the antimicrobial drugs targeting brain infections. A 2 mg/kg dose administered in mice for 7 days, resulted in the brain concentration of about 5 μM 35, which is 2.5–5× of the EC50 of auranofin against European, Australian and three genotypes of US strains of N. fowleri. Recent studies with amphotericin B conjugated with nanoparticles not only enhanced growth inhibition of N. fowleri but also increased the delivery of the drug to brain 36, 37. The employment of this strategy of loading auranofin into the nanoparticles may further improve the antiamebic effect and blood-brain barrier permeability of auranofin for the treatment of PAM.
Since N. fowleri infection causes extensive inflammation in the brain, the anti-inflammatory property of auranofin 20, 24 may also play a beneficial role by reducing the inflammation and normalizing intracranial pressure.
Effect of auranofin on growth inhibition at different time points
Since PAM has a rapid clinical course, it is important to identify a drug that is fast-acting. To establish how fast auranofin kills N. fowleri, we measured dose-response (EC50) of auranofin at 2 h, 16 h, 24 h and 48 h of exposure. Growth inhibition curves generated at different time points (Figure 1) demonstrated ~90% growth inhibition at 12.5 μM of auranofin as early as 16 h post-exposure (EC50 of 6.3 μM) and 97% inhibition at the same concentration at 24 h post-exposure (EC50 of 3.1 μM).
Figure 1.
Growth inhibition curves of auranofin at different time points against N. fowleri.
The early pharmacokinetic studies of auranofin from long-term therapy after oral administration detected about 25% of the administered dose in plasma and the peak concentrations reached within 1–2 hour 38. Recent phase I clinical trial in healthy individuals, who received 6 mg of auranofin orally daily for 7 days and were followed for 126 days, also showed that auranofin is safe and well-tolerated 39. Once-daily dosing for 7 days led to a plasma gold concentration of 0.312 μg/ml or 1.58 μM 39, which is equal to the in vitro EC50 of auranofin for N. fowleri. Mean terminal half-life of auranofin is approximately 35 days 39. The FDA has approved clinical trials of auranofin at up to 21 mg/day for the treatment of relapsed chronic lymphocytic leukemia (CLL) after a daily total dose of 12 mg was found well-tolerated for at least 28 days (Clinical Trial registration no. NCT01419691). Population PK modeling and Monte Carlo simulation showed that the plasma gold concentration nearly doubled after 14 days of treatment with 9 mg/day of auranofin treatment 39. The safety profile at higher dose and 28 days of treatment for the CLL suggests that higher plasma concentration of auranofin is achievable in the treatment of PAM. Relatively shorter treatment regimens required for the PAM patients than arthritis should minimize possible adverse events reported for auranofin 39.
Effect of auranofin on intracellular reactive oxygen species
Since auranofin is an inhibitor of selenoprotein synthesis 19 and thioredoxin reductase function 15, 18, 28, 40, 41, which is involved in protecting cells from damage caused by oxidative stress, we hypothesized that auranofin could exert potent activity against N. fowleri by targeting redox enzymes, thus inhibiting antioxidant pathways that maintain the intracellular redox homeostasis.
Since accumulation of intracellular reactive oxygen species is also associated with apoptosis 42, 43, there was a possibility that the treatment of cells with a concentration of drug that leads to cell death might mask the effect of auranofin as an inhibitor of the antioxidant pathways. Therefore, we undertook a short exposure of N. fowleri trophozoites with auranofin alone at a concentration that does not lead to cell death within specific time period. We compared the presence of intracellular reactive oxygen species between DMSO-treated control N. fowleri trophozoites and those treated with auranofin. After treatment with 3 μM auranofin alone or auranofin plus 300 μM hydrogen peroxide, H2O2, for 18 hours, we detected the presence of reactive oxygen species (Figure 2) with fluorescence generated by the oxidation of dichlorodihydrofluorescein added to the medium. This validates auranofin’s role in inhibiting antioxidant pathways in N. fowleri.
Figure 2.
Accumulation of intracellular reactive oxygen species in N. fowleri is enhanced by exposure to auranofin. Fluorescence imaging of N. fowleri detected reactive oxygen species within trophozoites following treatment with 3 μM of auranofin (ANF) for 18 hours or auranofin plus H2O2. Control trophozoites were treated with 0.5% DMSO alone and 0.5% DMSO plus 300 μM H2O2. Magnification, ×40.
Auranofin selectively inhibits dithiol function of the antioxidant enzymes such as thioredoxin reductase, thioredoxin-glutathione reductase, trypanothione reductase and glutathione peroxidase 15, 18, 28, 40, 41, 44. The genome of N. fowleri encodes thioredoxin reductases and glutathione peroxidase. Whether the increased potency of auranofin against N. fowleri is due to more inhibition of N. fowleri thioredoxin reductase or glutathione peroxidase than human thioredoxin reductase or glutathione peroxidase, requires further investigation.
Combination of auranofin and amphotericin B
A successful treatment of PAM requires combination therapy. All treatment regimens that resulted in patient survival contained amphotericin B. Amphotericin B was used in combination with other drugs like rifampin, fluconazole, sulfadiazine, miconazole, sulfisoxazole, ketoconazole, dexamethasone, ornidazole and chloramphenicol to increase the success of the treatment 45–52. However, among these drugs only miconazole was reported to have an additive or synergistic effect with amphotericin B 50. Tetracycline and minocycline were also found to have a synergistic effect when combined with amphotericin B 53. Azithromycin showed synergistic effect with amphotericin B against N. fowleri, both in vitro and in a mouse model of PAM 54. Yet no successful treatment with these combinations has been reported.
Pairing amphotericin B with a synergistic drug may reduce the dose required to achieve a maximum effect with minimum nephrotoxicity associated with amphotericin B. We determined the inhibitory effects of auranofin and amphotericin B at fixed concentration ratios. The dose-effect relationships between two drugs were assessed by classical isobolograms built to calculate Chou-Talalay combination indices (CI) and dose-reduction indices (DRI) as shown in Table 2, using CompuSyn software. The calculated parameters indicated synergy at different drug ratios. Thus, 95% growth inhibition with 2- to 9-fold dose reduction for amphotericin B and 3- to 20-fold dose reduction for auranofin was achieved.
Table 2.
Summary of synergism assay with amphotericin B and auranofin, shown for 95% growth inhibition of N. fowleri trophozoites
Amphotericin B : Auranofin ratio | % Growth Inhibition | Combination Index (CI) | Dose Reduction Index (DRI) | Dose required to achieve 95% inhibition (μM) | ||
---|---|---|---|---|---|---|
Amphotericin B | Auranofin | Amphotericin B | Auranofin | |||
1:16 | 95 | 0.5 ± 0.3 | 9.3 ± 3.2 | 2.8 ± 1.6 | 0.3 ± 0.1 | 5.2 ± 1.1 |
1:8 | 95 | 0.5 ± 0.1 | 5.2 ± 0.5 | 3.1 ± 1 | 0.6 ± 0.1 | 4.5 ± 0.5 |
1:4 | 95 | 0.7 ± 0.2 | 2.8 ± 0.4 | 3.3 ± 1 | 1.0 ± 0.1 | 4.1 ± 0.1 |
1:2 | 95 | 0.5 ± 0.1 | 3.0 ± 0.4 | 7.2 ± 2.5 | 0.9 ± 0.1 | 1.9 ± 0.2 |
1:1 | 95 | 0.5 ± 0.1 | 2.6 ± 0.1 | 12.2 ± 3.2 | 1.1 ± 0.1 | 1.1 ± 0.1 |
2:1 | 95 | 0.6 ± 0.1 | 2.0 ± 0.4 | 19.5 ± 7.4 | 1.4 ± 0.2 | 0.7 ± 0.1 |
The use of amphotericin B is mainly limited due to its nephrotoxicity. When tested against three different epithelial kidney cell lines (Vero, MDCK-PTR9 and GMK), the CC50 of amphotericin B was about 5.4 μM 55, 56. It showed a CC50 of 28.7 μM against human embryonic kidney cells (HEK293) 57. Auranofin also did not show any cytotoxic effect on HEK293 when tested up to 5 μM concentration 58. Considering the doses of amphotericin B and auranofin required to show synergism are much lower (Table 2) than the concentrations that demonstrate toxicity on kidney cells, we believe that the concentrations used in the combinations to achieve 95% growth inhibition of N. fowleri will not have any significant toxic effect on kidney cells. Since PAM is a brain infection and a combination of auranofin and amphotericin B may exert effect on brain cells, we considered the toxicity of auranofin and amphotericin B on human astrocytes, a type of brain cell. Both auranofin and amphotericin B did not exhibit any toxicity on human astrocytes at concentrations up to 5 μM 35 and 10.8 μM 59, respectively. Given that the combinations of auranofin and amphotericin B use lower concentrations to show synergistic activity against N. fowleri (Table 2) than the concentrations that demonstrate toxicity on astrocytes, it is unlikely that the doses used in the combination study will have toxic effect on human astrocytes. Although all the ratios that showed synergistic activity could be useful in the combination of auranofin and amphotericin B against N. fowleri, 1:1 ratio might be considered the best based on the use of low concentrations of both the drugs.
The drug combinations with highest synergy were analyzed microscopically for their effect on N. fowleri growth and morphology (Figure 3). The amphotericin B-auranofin pair, combined at concentrations of 0.6 μM and 4.5 μM, respectively, (CI=0.5), completely inhibited the trophozoite growth, had a detrimental effect on cell morphology and caused death of the majority of N. fowleri cells in 48 h of drug exposure; DMSO-treated control cells grew and appeared normal. These drug combination results give us an opportunity to reduce amphotericin concentration by an order of magnitude to achieve 95% of N. fowleri growth inhibition. This may allow minimizing the dose-limiting adverse effects of amphotericin B.
Figure 3.
Synergistic effect of drugs at low concentrations. The phase contrast microscope images show N. fowleri trophozoites treated for 48 hours with 0.5% DMSO, 0.6 μM amphotericin B, 4.5 μM of auranofin, and a combination of 0.6 μM of amphotericin B and 4.5 μM of auranofin. The auranofin-amphotericin B-treated N. fowleri cells visible in the microscope field are rounded, much smaller in size and not viable, whereas DMSO-, 0.6 μM amphotericin B- and 4.5 μM of auranofin-treated cells are irregularly shaped with visible cytoplasm. Magnification, ×20.
Future studies will involve confirmation of efficacy of auranofin, either alone or in combination with amphotericin B, in an animal model of PAM. Given that clinical trial for PAM is not possible, proving in vivo efficacy will allow repurposing of auranofin, either as a monotherapy or in combination with the standard of care amphotericin B, for the treatment of PAM.
METHODS
Maintenance of N. fowleri
Trophozoites of pathogenic N. fowleri European strain KUL (ATCC 30808) and different clinical strains were axenically cultured in Nelson’s medium supplemented with 10% FBS at 37°C 60. Clinical strains used in the study were Davis, CAMP, TY and CDC:V1005. These four clinical strains were acquired from Centers for Disease Control and Prevention (CDC), USA. US strains Davis, CAMP 61 and TY 62 belong to genotypes I, II and III, respectively. CDC:V1005 is an Australian strain. Experiments were conducted using trophozoites harvested at 48 hours when they were at the logarithmic phase of growth. Trophozoites were counted using a hemocytometer.
In vitro activity of auranofin, amphotericin B and miltefosine against different genotypes of N. fowleri
10 mM stock solutions of auranofin (Enzo Life Sciences) and amphotericin B (GoldBio) were prepared in 100% DMSO and stored at −20°C. 40 mM stock solution of miltefosine (Sigma) was prepared in sterile water. Sixteen concentrations of auranofin and amphotericin B and eight concentrations of miltefosine were tested against KUL, CAMP, Davis, TY and CDC:V1005 strains of N. fowleri. Briefly, 5 μL of 10 mM auranofin and amphotericin B was added in a 96-well clear bottom dilution plate. A two-fold serial dilution was then performed using 2.5 μL of compound and adding 2.5 μL of 100% DMSO, yielding a concentration range of 10 mM-0.0003 mM. From this dilution plate, 0.5 μL was transferred in triplicate into a 96-well screen plate followed by addition of 99.5 μL of trophozoites (10,000 amebae per well) into each well to yield a final 16-point concentration that ranged from 50 μM to 0.0015 μM in final 0.5% DMSO 16, 63. Similarly, a final 8-point concentration spanning 200 μM to 1.5 μM was achieved from a 40 mM stock of miltefosine. 0.5% DMSO was used as a vehicle control and 50 μM amphotericin B was used as a positive control. Assay plates were incubated for 48 hours at 37°C and after 48 hours, plates were kept at room temperature for 30 minutes. Effects of auranofin, amphotericin B and miltefosine on cell viability were measured by adding 25 μL of CellTiter-Glo Luminescent Cell Viability Assay (Promega) in each well of the 96-well plates. CellTiter-Glo first induced cell lysis when plates were kept on an orbital shaker at room temperature for 10 minutes. The resulting luminescent signal was then stabilized after equilibrating the plates at room temperature for 10 minutes. Released ATP bioluminescence of the trophozoites was measured at room temperature using an EnVision Multilabel Reader (PerkinElmer, Waltham, MA).
Data analysis and statistics
Growth inhibition percentage relative to maximum and minimum reference signal controls was calculated using the formula:
% inhibition = (1- [(experimental value − mean of maximum signal reference control)/(mean of minimum signal reference control − mean of maximum signal reference control)]) × 100
EC50 of auranofin, amphotericin B and miltefosine and 95% confidence intervals were determined using GraphPad Prism software 5.0.
Effect of auranofin on growth inhibition at different time points
To establish how fast auranofin kills N. fowleri, we measured the effect of auranofin on KUL strain at different time points. For this, KUL strain was incubated with different concentrations of auranofin, starting from 0.39 μM to 50 μM, in triplicate for 2 hours, 16 hours, 24 hours and 48 hours in 96-well microplates. Growth inhibition percentage and EC50 at different timepoints was determined by using the CellTiter-Glo Luminescent Cell Viability Assay following the same protocol as described above. Experiment was done in triplicate in three independent biological replicates.
Measurement of intracellular reactive oxygen species
To assess the effect of auranofin on antioxidant pathways of N. fowleri, 2′,7′-dichlorodihydrofluorescein diacetate (DCFDA) (Sigma) was used. DCFDA is a cell-permeable molecule which is oxidized by the reactive oxygen species inside the cell forming the fluorescent dichlorofluorescein (DCF) 64. Reactive oxygen species production assay was optimized for N. fowleri following earlier method 24. Briefly, 100 μL (20,000 amebae) of N. fowleri KUL trophozoites were preincubated in a transparent, flat bottom 96-well plate (Fisher Scientific) with 0.5% DMSO, 3 μM auranofin in duplicate for 18 h. In another set, compound-treated and 0.5% DMSO-treated trophozoites were incubated with 300 μM H2O2 for 2 h. After 2 h, media were removed from the wells, washed and replaced with prewarmed Nelson’s medium containing 0.4 mM of DCFDA. The plate was incubated at 37°C for 30 min in the dark. Each well was washed again and pictures of the fluorescent cells were taken with an Axio Vert.A1 inverted phase microscope (Zeiss) and Zen lite software (Zeiss).
Combination of auranofin and amphotericin B
To determine the effect of combination of auranofin and amphotericin B against N. fowleri, eight-point two-fold dilutions of auranofin and amphotericin B were made from 1.25 mM amphotericin B and 5 mM auranofin. 0.25 μL of each serially diluted drug was transferred into a solid bottom tissue culture 96-well plate (E&K Scientific) in a matrix-way. 99.5 μL of N. fowleri trophozoites (10,000 amebae) were added to yield a final volume of 100 μL per well reaching different ratios (1:1, 1:2, 1:4, 1:8, 1:16, 2:1, 4:1, 8:1) in the combination matrix formed by the mixtures of amphotericin B from 3.125 μM to 0.024 μM in rows and auranofin from 12.5 μM to 0.095 μM in columns. Identical concentrations of amphotericin B and auranofin were separately achieved in two columns to test the effect of each drug independently. 0.5% DMSO was used as a negative control and 50 μM amphotericin B as a positive control. All the experiments were performed in triplicate. The assay plates were incubated for 48 h at 37°C. The plates were equilibrated to room temperature and the CellTiter-Glo® luminescent cell viability assay was used to quantify the growth inhibition percentage of each combination as well as the individual drugs. The growth inhibition percentage values were calculated and the effect of combination was analyzed using CompuSyn software following the Chou-Talalay method 65, 66. The combination index (CI) values indicating either the additive (CI=1) or the antagonistic (CI>1) or the synergistic (CI<1) effect 67 were calculated from the CompuSyn software.
ACKNOWLEDGMENT
We thank Dr. Ibne Karim M. Ali and Shantanu Roy of CDC for providing the Australian and US strains of N. fowleri. We also thank Zhaoyu Huang for her help with the testing of amphotericin B and miltefosine against a few strains of N. fowleri.
Funding Sources
This work was supported by the grants 1KL2TR001444, R21AI141210, R21AI133394, R21AI146460 from NIH to A.D.
ABBREVIATIONS
- CNS
central nervous system
- PAM
primary amebic meningoencephalitis
REFERENCES
- 1.Yoder JS; Eddy BA; Visvesvara GS; Capewell L; Beach MJ, The epidemiology of primary amoebic meningoencephalitis in the USA, 1962–2008. Epidemiol Infect 2010, 138 (7), 968–75. [DOI] [PubMed] [Google Scholar]
- 2.Capewell LG; Harris AM; Yoder JS; Cope JR; Eddy BA; Roy SL; Visvesvara GS; Fox LM; Beach MJ, Diagnosis, Clinical Course, and Treatment of Primary Amoebic Meningoencephalitis in the United States, 1937–2013. J Pediatric Infect Dis Soc 2015, 4 (4), e68–75. [DOI] [PubMed] [Google Scholar]
- 3.Visvesvara GS; Moura H; Schuster FL, Pathogenic and opportunistic free-living amoebae: Acanthamoeba spp., Balamuthia mandrillaris, Naegleria fowleri, and Sappinia diploidea. FEMS Immunol Med Microbiol 2007, 50 (1), 1–26. [DOI] [PubMed] [Google Scholar]
- 4.Khetsuriani N; Holman RC; Lamonte-Fowlkes AC; Selik RM; Anderson LJ, Trends in encephalitis-associated deaths in the United States. Epidemiol Infect 2007, 135 (4), 583–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Visvesvara GS, Amebic meningoencephalitides and keratitis: challenges in diagnosis and treatment. Curr Opin Infect Dis 2010, 23 (6), 590–4. [DOI] [PubMed] [Google Scholar]
- 6.Centers for Disease, C.; Prevention, Investigational drug available directly from CDC for the treatment of infections with free-living amebae. MMWR Morb Mortal Wkly Rep 2013, 62 (33), 666. [PMC free article] [PubMed] [Google Scholar]
- 7.Linam WM; Ahmed M; Cope JR; Chu C; Visvesvara GS; da Silva AJ; Qvarnstrom Y; Green J, Successful treatment of an adolescent with Naegleria fowleri primary amebic meningoencephalitis. Pediatrics 2015, 135 (3), e744–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Navarro M; Cisneros-Fajardo EJ; Marchan E, New silver polypyridyl complexes: synthesis, characterization and biological activity on Leishmania mexicana. Arzneimittelforschung 2006, 56 (8), 600–4. [DOI] [PubMed] [Google Scholar]
- 9.Otero L; Vieites M; Boiani L; Denicola A; Rigol C; Opazo L; Olea-Azar C; Maya JD; Morello A; Krauth-Siegel RL; Piro OE; Castellano E; Gonzalez M; Gambino D; Cerecetto H, Novel antitrypanosomal agents based on palladium nitrofurylthiosemicarbazone complexes: DNA and redox metabolism as potential therapeutic targets. J Med Chem 2006, 49 (11), 3322–31. [DOI] [PubMed] [Google Scholar]
- 10.Vieites M; Otero L; Santos D; Toloza J; Figueroa R; Norambuena E; Olea-Azar C; Aguirre G; Cerecetto H; Gonzalez M; Morello A; Maya JD; Garat B; Gambino D, Platinum(II) metal complexes as potential anti-Trypanosoma cruzi agents. J Inorg Biochem 2008, 102 (5–6), 1033–43. [DOI] [PubMed] [Google Scholar]
- 11.Maurya MR; Uprety B; Avecilla F; Tariq S; Azam A, Palladium(II) complexes of OS donor N-(di(butyl/phenyl)carbamothioyl)benzamide and their antiamoebic activity. Eur J Med Chem 2015, 98, 54–60. [DOI] [PubMed] [Google Scholar]
- 12.Colotti G; Fiorillo A; Ilari A, Metal- and metalloid-containing drugs for the treatment of trypanosomatid diseases. Front Biosci (Landmark Ed) 2018, 23, 954–966. [DOI] [PubMed] [Google Scholar]
- 13.Colotti G; Ilari A; Fiorillo A; Baiocco P; Cinellu MA; Maiore L; Scaletti F; Gabbiani C; Messori L, Metal-based compounds as prospective antileishmanial agents: inhibition of trypanothione reductase by selected gold complexes. ChemMedChem 2013, 8 (10), 1634–7. [DOI] [PubMed] [Google Scholar]
- 14.Fricker SP, Cysteine proteases as targets for metal-based drugs. Metallomics 2010, 2 (6), 366–77. [DOI] [PubMed] [Google Scholar]
- 15.Sannella AR; Casini A; Gabbiani C; Messori L; Bilia AR; Vincieri FF; Majori G; Severini C, New uses for old drugs. Auranofin, a clinically established antiarthritic metallodrug, exhibits potent antimalarial effects in vitro: Mechanistic and pharmacological implications. FEBS Lett 2008, 582 (6), 844–7. [DOI] [PubMed] [Google Scholar]
- 16.Debnath A; Nelson AT; Silva-Olivares A; Shibayama M; Siegel D; McKerrow JH, In Vitro Efficacy of Ebselen and BAY 11–7082 Against Naegleria fowleri. Front Microbiol 2018, 9, 414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.da Silva MT; Caldas VE; Costa FC; Silvestre DA; Thiemann OH, Selenocysteine biosynthesis and insertion machinery in Naegleria gruberi. Mol Biochem Parasitol 2013, 188 (2), 87–90. [DOI] [PubMed] [Google Scholar]
- 18.Angelucci F; Sayed AA; Williams DL; Boumis G; Brunori M; Dimastrogiovanni D; Miele AE; Pauly F; Bellelli A, Inhibition of Schistosoma mansoni thioredoxin-glutathione reductase by auranofin: structural and kinetic aspects. J Biol Chem 2009, 284 (42), 28977–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Manhas R; Gowri VS; Madhubala R, Leishmania donovani Encodes a Functional Selenocysteinyl-tRNA Synthase. J Biol Chem 2016, 291 (3), 1203–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Roder C; Thomson MJ, Auranofin: repurposing an old drug for a golden new age. Drugs R D 2015, 15 (1), 13–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Peroutka-Bigus N; Bellaire BH, Antiparasitic Activity of Auranofin against Pathogenic Naegleria fowleri. J Eukaryot Microbiol 2019, 66 (4), 684–688. [DOI] [PubMed] [Google Scholar]
- 22.Pélandakis M; Kaundun SS; De Jonckheere JF; Pernin P, DNA diversity among the free-living amoeba, Naegleria fowleri, detected by the random amplified polymorphic DNA method. FEMS Microbiol Lett 1997, 151 (1), 31–39. [Google Scholar]
- 23.Duma RJ; Finley R, In vitro susceptibility of pathogenic Naegleria and Acanthamoeba speicies to a variety of therapeutic agents. Antimicrob Agents Chemother 1976, 10 (2), 370–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Debnath A; Parsonage D; Andrade RM; He C; Cobo ER; Hirata K; Chen S; Garcia-Rivera G; Orozco E; Martinez MB; Gunatilleke SS; Barrios AM; Arkin MR; Poole LB; McKerrow JH; Reed SL, A high-throughput drug screen for Entamoeba histolytica identifies a new lead and target. Nat Med 2012, 18 (6), 956–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tejman-Yarden N; Miyamoto Y; Leitsch D; Santini J; Debnath A; Gut J; McKerrow JH; Reed SL; Eckmann L, A reprofiled drug, auranofin, is effective against metronidazole-resistant Giardia lamblia. Antimicrob Agents Chemother 2013, 57 (5), 2029–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Debnath A; Ndao M; Reed SL, Reprofiled drug targets ancient protozoans: drug discovery for parasitic diarrheal diseases. Gut Microbes 2013, 4 (1), 66–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hopper M; Yun JF; Zhou B; Le C; Kehoe K; Le R; Hill R; Jongeward G; Debnath A; Zhang L; Miyamoto Y; Eckmann L; Land KM; Wrischnik LA, Auranofin inactivates Trichomonas vaginalis thioredoxin reductase and is effective against trichomonads in vitro and in vivo. Int J Antimicrob Agents 2016, 48 (6), 690–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ilari A; Baiocco P; Messori L; Fiorillo A; Boffi A; Gramiccia M; Di Muccio T; Colotti G, A gold-containing drug against parasitic polyamine metabolism: the X-ray structure of trypanothione reductase from Leishmania infantum in complex with auranofin reveals a dual mechanism of enzyme inhibition. Amino Acids 2012, 42 (2–3), 803–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sharlow ER; Leimgruber S; Murray S; Lira A; Sciotti RJ; Hickman M; Hudson T; Leed S; Caridha D; Barrios AM; Close D; Grogl M; Lazo JS, Auranofin is an apoptosis-simulating agent with in vitro and in vivo anti-leishmanial activity. ACS Chem Biol 2014, 9 (3), 663–72. [DOI] [PubMed] [Google Scholar]
- 30.Andrade RM; Chaparro JD; Capparelli E; Reed SL, Auranofin is highly efficacious against Toxoplasma gondii in vitro and in an in vivo experimental model of acute toxoplasmosis. PLoS Negl Trop Dis 2014, 8 (7), e2973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.da Silva MT; Silva-Jardim I; Portapilla GB; de Lima GM; Costa FC; Anibal Fde F; Thiemann OH, In vivo and in vitro auranofin activity against Trypanosoma cruzi: Possible new uses for an old drug. Exp Parasitol 2016, 166, 189–93. [DOI] [PubMed] [Google Scholar]
- 32.Kangussu-Marcolino MM; Ehrenkaufer GM; Chen E; Debnath A; Singh U, Identification of plicamycin, TG02, panobinostat, lestaurtinib, and GDC-0084 as promising compounds for the treatment of central nervous system infections caused by the free-living amebae Naegleria, Acanthamoeba and Balamuthia. Int J Parasitol Drugs Drug Resist 2019, 11, 80–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.James LR; Xu ZQ; Sluyter R; Hawksworth EL; Kelso C; Lai B; Paterson DJ; de Jonge MD; Dixon NE; Beck JL; Ralph SF; Dillon CT, An investigation into the interactions of gold nanoparticles and anti-arthritic drugs with macrophages, and their reactivity towards thioredoxin reductase. J Inorg Biochem 2015, 142, 28–38. [DOI] [PubMed] [Google Scholar]
- 34.Thangamani S; Mohammad H; Abushahba MF; Sobreira TJ; Seleem MN, Repurposing auranofin for the treatment of cutaneous staphylococcal infections. Int J Antimicrob Agents 2016, 47 (3), 195–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Madeira JM; Renschler CJ; Mueller B; Hashioka S; Gibson DL; Klegeris A, Novel protective properties of auranofin: inhibition of human astrocyte cytotoxic secretions and direct neuroprotection. Life Sci 2013, 92 (22), 1072–80. [DOI] [PubMed] [Google Scholar]
- 36.Lemke A; Kiderlen AF; Petri B; Kayser O, Delivery of amphotericin B nanosuspensions to the brain and determination of activity against Balamuthia mandrillaris amebas. Nanomedicine 2010, 6 (4), 597–603. [DOI] [PubMed] [Google Scholar]
- 37.Rajendran K; Anwar A; Khan NA; Siddiqui R, Brain-Eating Amoebae: Silver Nanoparticle Conjugation Enhanced Efficacy of Anti-Amoebic Drugs against Naegleria fowleri. ACS Chem Neurosci 2017, 8 (12), 2626–2630. [DOI] [PubMed] [Google Scholar]
- 38.Kean WF; Hart L; Buchanan WW, Auranofin. Br J Rheumatol 1997, 36 (5), 560–72. [DOI] [PubMed] [Google Scholar]
- 39.Capparelli EV; Bricker-Ford R; Rogers MJ; McKerrow JH; Reed SL, Phase I Clinical Trial Results of Auranofin, a Novel Antiparasitic Agent. Antimicrob Agents Chemother 2017, 61 (1), e01947–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Caroli A; Simeoni S; Lepore R; Tramontano A; Via A, Investigation of a potential mechanism for the inhibition of SmTGR by Auranofin and its implications for Plasmodium falciparum inhibition. Biochem Biophys Res Commun 2012, 417 (1), 576–81. [DOI] [PubMed] [Google Scholar]
- 41.Prast-Nielsen S; Huang HH; Williams DL, Thioredoxin glutathione reductase: its role in redox biology and potential as a target for drugs against neglected diseases. Biochim Biophys Acta 2011, 1810 (12), 1262–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Brodska B; Holoubek A, Generation of reactive oxygen species during apoptosis induced by DNA-damaging agents and/or histone deacetylase inhibitors. Oxid Med Cell Longev 2011, 2011, 253529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Thannickal VJ; Fanburg BL, Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol 2000, 279 (6), L1005–28. [DOI] [PubMed] [Google Scholar]
- 44.Radenkovic F; Holland O; Vanderlelie JJ; Perkins AV, Selective inhibition of endogenous antioxidants with Auranofin causes mitochondrial oxidative stress which can be countered by selenium supplementation. Biochem Pharmacol 2017, 146, 42–52. [DOI] [PubMed] [Google Scholar]
- 45.Anderson K; Jamieson A, Primary amoebic meningoencephalitis. Lancet 1972, 1 (7756), 902–3. [DOI] [PubMed] [Google Scholar]
- 46.Apley J; Clarke SK; Roome AP; Sandry SA; Saygi G; Silk B; Warhurst DC, Primary amoebic meningoencephalitis in Britain. Br Med J 1970, 1 (5696), 596–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jain R; Prabhakar S; Modi M; Bhatia R; Sehgal R, Naegleria meningitis: a rare survival. Neurol India 2002, 50 (4), 470–2. [PubMed] [Google Scholar]
- 48.Loschiavo F; Ventura-Spagnolo T; Sessa E; Bramanti P, Acute primary meningoencephalitis from entamoeba Naegleria Fowleri. Report of a clinical case with a favourable outcome. Acta Neurol (Napoli) 1993, 15 (5), 333–40. [PubMed] [Google Scholar]
- 49.Poungvarin N; Jariya P, The fifth nonlethal case of primary amoebic meningoencephalitis. J Med Assoc Thai 1991, 74 (2), 112–5. [PubMed] [Google Scholar]
- 50.Seidel JS; Harmatz P; Visvesvara GS; Cohen A; Edwards J; Turner J, Successful treatment of primary amebic meningoencephalitis. N Engl J Med 1982, 306 (6), 346–8. [DOI] [PubMed] [Google Scholar]
- 51.Vargas-Zepeda J; Gomez-Alcala AV; Vasquez-Morales JA; Licea-Amaya L; De Jonckheere JF; Lares-Villa F, Successful treatment of Naegleria fowleri meningoencephalitis by using intravenous amphotericin B, fluconazole and rifampicin. Arch Med Res 2005, 36 (1), 83–6. [DOI] [PubMed] [Google Scholar]
- 52.Wang A; Kay R; Poon WS; Ng HK, Successful treatment of amoebic meningoencephalitis in a Chinese living in Hong Kong. Clin Neurol Neurosurg 1993, 95 (3), 249–52. [DOI] [PubMed] [Google Scholar]
- 53.Lee KK; Karr SL Jr.; Wong MM; Hoeprich PD, In vitro susceptibilities of Naegleria fowleri strain HB-1 to selected antimicrobial agents, singly and in combination. Antimicrob Agents Chemother 1979, 16 (2), 217–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Soltow SM; Brenner GM, Synergistic activities of azithromycin and amphotericin B against Naegleria fowleri in vitro and in a mouse model of primary amebic meningoencephalitis. Antimicrob Agents Chemother 2007, 51 (1), 23–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kagan S; Ickowicz D; Shmuel M; Altschuler Y; Sionov E; Pitusi M; Weiss A; Farber S; Domb AJ; Polacheck I, Toxicity mechanisms of amphotericin B and its neutralization by conjugation with arabinogalactan. Antimicrob Agents Chemother 2012, 56 (11), 5603–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Klimek K; Strubinska J; Czernel G; Ginalska G; Gagos M, In vitro evaluation of antifungal and cytotoxic activities as also the therapeutic safety of the oxidized form of amphotericin B. Chem Biol Interact 2016, 256, 47–54. [DOI] [PubMed] [Google Scholar]
- 57.Zia Q; Mohammad O; Rauf MA; Khan W; Zubair S, Biomimetically engineered Amphotericin B nano-aggregates circumvent toxicity constraints and treat systemic fungal infection in experimental animals. Sci Rep 2017, 7 (1), 11873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Marzo T; Cirri D; Gabbiani C; Gamberi T; Magherini F; Pratesi A; Guerri A; Biver T; Binacchi F; Stefanini M; Arcangeli A; Messori L, Auranofin, Et3PAuCl, and Et3PAuI Are Highly Cytotoxic on Colorectal Cancer Cells: A Chemical and Biological Study. ACS Med Chem Lett 2017, 8 (10), 997–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Motoyoshi-Yamashiro A; Tamura M; Moriyama M; Takano K; Kawabe K; Nakajima H; Katoh-Semba R; Furuichi T; Nakamura Y, Activation of cultured astrocytes by amphotericin B: stimulation of NO and cytokines production and changes in neurotrophic factors production. Neurochem Int 2013, 63 (2), 93–100. [DOI] [PubMed] [Google Scholar]
- 60.Debnath A; Calvet CM; Jennings G; Zhou W; Aksenov A; Luth MR; Abagyan R; Nes WD; McKerrow JH; Podust LM, CYP51 is an essential drug target for the treatment of primary amoebic meningoencephalitis (PAM). PLoS Negl Trop Dis 2017, 11 (12), e0006104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhou L; Sriram R; Visvesvara GS; Xiao L, Genetic variations in the internal transcribed spacer and mitochondrial small subunit rRNA gene of Naegleria spp. J Eukaryot Microbiol 2003, 50 Suppl, 522–6. [DOI] [PubMed] [Google Scholar]
- 62.Schuster FL; Rechthand E, In vitro effects of amphotericin B on growth and ultrastructure of the amoeboflagellates Naegleria gruberi and Naegleria fowleri. Antimicrob Agents Chemother 1975, 8 (5), 591–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Debnath A; Tunac JB; Galindo-Gomez S; Silva-Olivares A; Shibayama M; McKerrow JH, Corifungin, a new drug lead against Naegleria, identified from a high-throughput screen. Antimicrob Agents Chemother 2012, 56 (11), 5450–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang H; Joseph JA, Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med 1999, 27 (5–6), 612–6. [DOI] [PubMed] [Google Scholar]
- 65.Chou TC, Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 2006, 58 (3), 621–81. [DOI] [PubMed] [Google Scholar]
- 66.Chou TC; Talalay P, Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul 1984, 22, 27–55. [DOI] [PubMed] [Google Scholar]
- 67.Chou TC, Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 2010, 70 (2), 440–6. [DOI] [PubMed] [Google Scholar]