Journal of Medical Microbiology Papers in Press. Published February 13, 2015 as doi:10.1099/jmm.0.000041

Journal of Medical Microbiology Miltefosine is active against Sporothrix brasiliensis isolates with in vitro low susceptibility to amphotericin B or itraconazole. --Manuscript Draft-Manuscript Number:

JMM-D-14-00341

Full Title:

Miltefosine is active against Sporothrix brasiliensis isolates with in vitro low susceptibility to amphotericin B or itraconazole.

Short Title:

Miltefosine is active against Sporothrix brasiliensis isolates with low susceptibility standart antifungals.

Article Type:

Standard

Section/Category:

Antimicrobial agents and chemotherapy

Keywords:

Sporothrix brasiliensis, sporotrichosis, antifungal, miltefosine, yeasts alterations.

Corresponding Author:

Sonia Rozental Universidade Federal do Rio de Janeiro Rio de Janeiro, Rio de Janeiro BRAZIL

First Author:

Luana Pereira Borba-Santos, MSc

Order of Authors:

Luana Pereira Borba-Santos, MSc Thalita Gagini Kelly Ishida Wanderley de Souza, PhD Sonia Rozental

Manuscript Region of Origin:

BRAZIL

Abstract:

Sporotrichosis is a common mycosis caused by dimorphic fungi from the Sporothrix schenckii complex. In recent years, sporotrichosis incidence rates have increased in the Brazilian state of Rio de Janeiro, where Sporothrix brasiliensis is the species more frequently isolated from patients. The standard antifungals itraconazole and amphotericin B are recommended as first-line therapy for cutaneous/lymphocutaneous and disseminated sporotrichosis, respectively, although decreased sensitivity to these drugs in vitro was reported for clinical isolates of S. brasiliensis. Here, we evaluated the activity of the phospholipid analogue miltefosine - already in clinical use against leishmaniasis - towards the pathogenic yeast form of S. brasiliensis isolates with low sensitivity to itraconazole or amphotericin B in vitro. Miltefosine had fungicidal activity, with minimum inhibitory concentration (MIC) values between 1-2 µg.mL-1. Miltefosine exposure led to loss of plasma membrane integrity, and transmission electron microscopy (TEM) analysis revealed decrease in cytoplasmic electron density, alterations in the thickness of cell wall layers, and accumulation of an electron dense material in the cell wall. Flow cytometry analysis using an anti-melanin antibody revealed an increase in cell wall melanin in yeasts treated with miltefosine, when compared with control cells. The cytotoxicity of miltefosine were comparable to those of amphotericin B, but miltefosine showed higher selectivity index towards the fungus. Our results suggest that miltefosine could be an effective alternative for the treatment of S. brasiliensis sporotrichosis, when standard treatment fails. Nevertheless, in vivo studies are required to confirm the antifungal potential of miltefosine for the treatment of sporotrichosis.

Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation

Manuscript Including References (Word document) Click here to download Manuscript Including References (Word document): FML_052_BorbaSantosRozental_2014_Revised version fin

1

Title: Miltefosine is active against Sporothrix brasiliensis isolates with in vitro low

2

susceptibility to amphotericin B or itraconazole.

3 4

Running title: Miltefosine activity against Sporothrix brasiliensis

5 6

Category: Antimicrobial Agents & Chemotherapy.

7 8

Authors: Luana Pereira Borba-Santos1, Thalita Gagini1, Kelly Ishida2, Wanderley de

9

Souza3,4, Sonia Rozental1#.

10 11 12

1

13

2

14 15

3

16 17

4

Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil.

Instituto Nacional de Metrologia, Qualidade e Tecnologia, Inmetro, Duque de Caxias, Brasil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil

18 19 20

#Corresponding author:

21

Mailing address: Laboratorio de Biologia Celular de Fungos, Instituto de Biofísica

22

Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil

23

Avenida Carlos Chagas Filho 373, Centro de Ciências da Saúde, Bloco G, Sala C0-026,

24

21.941-902, Rio de Janeiro/RJ, Brazil.Telefone number: +55-21-39386592

25

Fax number: +55-21-3938-8193

26

E-mail: [email protected]

27

28

Abstract

29

Sporotrichosis is a common mycosis caused by dimorphic fungi from the Sporothrix

30

schenckii complex. In recent years, sporotrichosis incidence rates have increased in the

31

Brazilian state of Rio de Janeiro, where Sporothrix brasiliensis is the species more

32

frequently isolated from patients. The standard antifungals itraconazole and

33

amphotericin B are recommended as first-line therapy for cutaneous/lymphocutaneous

34

and disseminated sporotrichosis, respectively, although decreased sensitivity to these

35

drugs in vitro was reported for clinical isolates of S. brasiliensis. Here, we evaluated the

36

activity of the phospholipid analogue miltefosine – already in clinical use against

37

leishmaniasis - towards the pathogenic yeast form of S. brasiliensis isolates with low

38

sensitivity to itraconazole or amphotericin B in vitro. Miltefosine had fungicidal

39

activity, with minimum inhibitory concentration (MIC) values between 1-2 µg.mL-1.

40

Miltefosine exposure led to loss of plasma membrane integrity, and transmission

41

electron microscopy (TEM) analysis revealed decrease in cytoplasmic electron density,

42

alterations in the thickness of cell wall layers, and accumulation of an electron dense

43

material in the cell wall. Flow cytometry analysis using an anti-melanin antibody

44

revealed an increase in cell wall melanin in yeasts treated with miltefosine, when

45

compared with control cells. The cytotoxicity of miltefosine were comparable to those

46

of amphotericin B, but miltefosine showed higher selectivity index towards the fungus.

47

Our results suggest that miltefosine could be an effective alternative for the treatment of

48

S. brasiliensis sporotrichosis, when standard treatment fails. Nevertheless, in vivo

49

studies are required to confirm the antifungal potential of miltefosine for the treatment

50

of sporotrichosis.

51 52

Key words: Sporothrix brasiliensis, sporotrichosis, antifungal, miltefosine, yeasts

53

alterations.

54 55 56 57 58

59

Introduction

60

Sporotrichosis is the most frequent subcutaneous mycosis in Latin America and has

61

high incidence rates in Brazil, especially in the Rio de Janeiro state, where zoonotic

62

transmission prevails (Barros et al., 2010). This disease is caused by dimorphic fungal

63

species from the Sporothrix schenckii complex (Marimon et al., 2007 and 2008),

64

including Sporothrix brasiliensis, the most virulent species (Arrigala-Moncrieff et al.,

65

2009; Fernandes et al., 2013) and the most prevalent in the clinical cases from the Rio

66

de Janeiro state (Oliveira et al., 2011; Rodrigues et al., 2013; Borba-Santos et al.,

67

2014). In zoonotic transmission, sporotrichosis is mainly acquired by scratches, bites or

68

direct contact with lesion secretion from contaminated animals, where yeasts are the

69

infective forms (Rodrigues et al., 2013b).

70

The ‘gold standard’ treatment for cutaneous and lymphocutaneous forms of

71

sporotrichosis is itraconazole, while amphotericin B is the first-line drug of choice for

72

disseminated forms of the disease (Kauffman et al., 2007). However, recent reports

73

demonstrated that these drugs are less potent against S. brasiliensis isolates, which have

74

high minimum inhibitory concentration (MIC) values in vitro for these antifungals

75

(Ottonelli et al., 2013; Rodrigues et al., 2014; Borba-Santos et al., 2014).

76

Miltefosine is a phospholipid analogue originally developed as an antitumor drug, but

77

later shown to have potent antiparasitic activity, which led to its licensing for the

78

treatment of cutaneous and visceral leishmaniasis in India and Colombia (Dorlo et al.,

79

2012). Miltefosine induces changes in the lipid composition of membranes (among

80

other cellular effects), leading to a disruption of membrane structure that affects

81

intracellular signaling processes essential for survival and cell growth (Jimenez-Lopes

82

et al., 2010). Recently, Brilhante and co-workers (2014) showed that miltefosine is

83

active against the filamentous (i.e., saprophytic) form of S. schenckii complex species,

84

including S. brasiliensis. However, these authors did not determine whether miltefosine

85

is active against the pathogenic (yeast) form of S. schenckii complex species, and did

86

not examine the intracellular effects of miltefosine in these fungi.

87

Given the clinical and epidemiological importance of S. brasiliensis, and the limited

88

repertoire of therapeutic options available to treat sporotrichosis by this highly virulent

89

species, the aim of our study was to evaluate the in vitro activity of miltefosine against

90

the pathogenic yeast form of clinical isolates of S. brasiliensis that have low

91

susceptibility to amphotericin B or itraconazole (Borba-Santos et al., 2014). In addition,

92

we analyzed the cytotoxicity of miltefosine, and evaluated the alterations induced by

93

exposure of yeast cells to this drug (by electron microscopy and flow cytometry), to

94

clarify the mechanism of action of miltefosine in S. brasiliensis.

95

Methods

96

Microorganisms

97

In this study, we used 13 clinical isolates of S. brasiliensis (Ss 34, Ss 37, Ss 56, Ss 68,

98

B91, B182, B628, B735, B848, B972, B1008, B1086, HE06) that displayed low

99

susceptibility to amphotericin B or itraconazole during in vitro testing (Borba-Santos et

100

al., 2014). The genome strain ATCC MYA 4823 was used as a reference (Teixeira et

101

al., 2014). The isolates were maintained in the filamentous (mycelial) form in potato

102

dextrose agar (PDA; Difco, Detroit, USA) at 4°C until their use in experiments. The

103

yeast phase was obtained from filamentous-form cultures by two successive 7-day

104

passages in brain heart infusion (BHI, Difco, Detroit, USA) broth supplemented with

105

2% glucose, at 36ºC, and with orbital agitation (Borba-Santos et al., 2014).

106

Drugs

107

Miltefosine (Cayman Chemical Company, MI, USA) was diluted in distilled water to

108

obtain stock solutions of 2,000 µg mL-1 and maintained at -20°C. Amphotericin B and

109

itraconazole (Sigma Chemical Co. St Louis, MO, USA) were used as reference

110

antifungal and kept as 1,600 µg mL-1 in DMSO stock solutions, and stored at -20°C.

111

Minimum inhibitory concentration (MIC)

112

MIC values for the treatment of S. brasiliensis yeasts with miltefosine were determined

113

using both microdilution methods adapted from the M27-A3 document from the

114

Clinical and Laboratory Standards Institute (CLSI, 2008), as described in Borba-Santos

115

et al. (2014). Briefly, yeast cells (0.5-2.5 x 103 colony forming units or CFU.mL-1) were

116

treated with 0.01-16 µg.mL-1 miltefosine, in RPMI 1640 medium (Sigma Chemical Co.)

117

supplemented with 2% glucose and buffered with 0.165 M 3-(N-morpholine) propane

118

sulfonic acid (MOPS), pH 7.2. Samples were incubated at 35°C for 5 days and MIC was

119

defined as the lowest concentration that prevents visible fungal growth in an inverted

120

optical microscope (Axiovert 100, Zeiss, Germany).

121

Minimal fungicidal concentration (MFC)

122

To determine MFC values, 10-µL aliquots were collected from MIC samples at the end

123

of the 5-day incubation period, and were plated in drug-free PDA. Plates were incubated

124

at 35°C for 7 days, and the lowest concentration of miltefosine that failed to yield

125

fungal growth was defined as the MFC.

126

Drug interactions

127

Interactions between miltefosine and itraconazole were assayed for the reference strain

128

(ATCC MYA 4823) and also for two clinical isolates (Ss 37 and B737) with high MIC

129

values (≥ 4 µg.mL-1) for itraconazole, using the checkerboard microdilution method

130

(Eliopoulos and Moellering, 1991). Concentrations ranging from 0.03-16 µg.mL-1 for

131

itraconazole and 0.25-16 µg.mL-1 for miltefosine were tested. MICs were determined

132

for drugs alone and for their combinations, and the fractional inhibitory concentration

133

index (FICI) was used to classify drug interactions. FICI values were determined using

134

the equation: FICI = (MICa in combination/MICa tested alone) + (MICb in

135

combination/MICb tested alone), for drugs itraconazole (a) and miltefosine (b) tested

136

against the same fungal strain (Eliopoulos and Moellering, 1991). Interactions were

137

considered synergistic if FICI ≤ 0.5, absent if FICI > 0.5 and ≤ 4, and antagonistic if

138

FICI > 4 (Odds, 2003).

139

Time kill assays.

140

For time kill assays, S. brasiliensis ATCC MYA 4823 yeasts (103 CFU.mL-1 starting

141

inoculum) were exposed to different concentrations of miltefosine (0, MIC/2, MIC,

142

MICx2, MICx4, MICx8) in RPMI 1640 medium supplemented with 2% glucose and

143

buffered with 0.165 M MOPS (pH, 7.2), for 24, 48, 72, 96 and 168 hours, at 35ºC.

144

Treated cultures were diluted and 50 µL of diluted cultures were plated on PDA

145

medium and incubated at 35ºC for 7 days before CFU counting. Fungicidal activity was

146

defined as a reduction of ≥ 99.9% in the number of CFU relative to that found in the

147

starting inoculum; otherwise, the activity was considered fungistatic (Klepser et al.,

148

1998).

149

Membrane integrity assay

150

S. brasiliensis yeast cells (ATCC MYA 4823) were treated with sub-inhibitory

151

concentrations (1/2 MIC) of miltefosine, amphotericin B or itraconazole for 24 h at

152

35ºC. Untreated and treated yeasts were washed in PBS and cells were incubated with

153

10 µM of SYTOX® Blue dead cell stain (Molecular Probes®, Eugene, OR, USA) for

154

20 min at room temperature, and in a dark chamber. After incubation, cells were washed

155

in PBS and the fluorescence intensity was measured at 480 nm (excitation at 440 nm)

156

using a Spectra-MAX 340 tunable microplate reader (Molecular Devices Ltd, CA,

157

USA).

158

Transmission electron microscopy

159

S. brasiliensis ATCC MYA 4823 yeast cells were treated with a sub-inhibitory

160

concentration (1/2 MIC) of miltefosine for 24 h at 35ºC. Untreated and treated cells

161

were washed in PBS, fixed in 2.5% glutaraldehyde and 4% formaldehyde in 0.1 M

162

cacodylate buffer, (for 24 h at 4°C), and post-fixed in 1% osmium tetroxide in 0.1 M

163

cacodylate buffer containing 1.25 % potassium ferrocyanide and 5mM CaCl2, for 2 h at

164

4°C. Then, cells were dehydrated in ethanol and embedded in Spurr resin. Ultrathin

165

sections were stained in uranyl acetate and lead citrate, and observed in a JEOL 1200

166

EX electron microscope. Cell wall thickness values – CW* (thickness of the innermost

167

cell wall layer) and ML (thickness of the outer microfibrilar layer of cell wall) - were

168

measured in 20 cells for each sample, using the Image J software (NHI, USA).

169

Flow cytometry analysis.

170

Untreated and miltefosine-treated S. brasiliensis ATCC MYA 4823 yeast cells (treated

171

with sub-inhibitory concentration (1/2 MIC) for 24 h at 35ºC) were washed in PBS,

172

fixed in 4% of formaldehyde in PBS, incubated with 1 % BSA for 30 min and then

173

labeled with anti-melanin antibodies (0.1 mg/mL, for 30 min at room temperature)

174

purified from sera obtained from chromoblastomycosis patients (Alviano et al., 2004).

175

Then, samples were incubated with FITC–labeled anti-human IgG (Molecular Probes®,

176

Eugene, OR, USA) for 30 min at room temperature, and analyzed in a BD AccuriTM C6

177

flow cytometer (10000 events/sample). Data were analyzed using the BD Accuri C6

178

Software. Negative control samples were not incubated with melanin antibodies.

179

Citotoxicity test

180

For cytotoxicity testing, aliquots of 100 µL of LLC-MK2 (Rhesus monkey kidney

181

epithelial cells) cell suspensions at 5 x 104 cells/mL were dispensed onto a 96-well

182

microtitre and incubated for 24 h. Then, monolayers were treated with different

183

concentrations of miltefosine or amphotericin B (1-100 µg.mL-1) for 48 h, at 37°C and

184

5% CO2. Cell viability was analyses by the tetrazolium (XTT) reduction assay and

185

selectivity indexes (SIs) were calculated using the formula: SI = CC50/MIC. CC50 values

186

correspond to the concentration that killed 50% of LLC-MK2 cells.

187

Statistical analysis

188

Statistical analyses were performed using the GraphPad Prism 5.0 software. Wilcoxon’s

189

tests were used to analyze differences between antifungal treatments and Mann

190

Whitney’s tests were used to analyze differences in cell wall thickness values. Linear

191

regression was used to analyze correlations between MIC values. One-way ANOVA

192

was used (with Dunnett’s post hoc test) to compare membrane integrity evaluation

193

results. A 5% significance level was adopted (p < 0.05).

194 195

Results

196

The yeast form of S. brasiliensis is sensitive to low concentrations (≤ 2 g.mL-1) of

197

miltefosine.

198

The sensitivity of the infective yeast form of S. brasiliensis to miltefosine in vitro was

199

established by determining MIC and MFC values, using reference protocols, and these

200

values were compared to those reported previously for the standard antifungals

201

amphotericin B and itraconazole (Table 1). S. brasiliensis clinical isolates displaying

202

high MIC values (MIC ≥ 4 µg.mL-1) for amphotericin B or itraconazole (Borba-Santos

203

et al., 2014) were sensitive to miltefosine (MIC ≤ 2 µg.mL-1), with MIC values between

204

1 and 2 µg.mL-1 for all isolates tested (Table 1). Miltefosine was more potent than

205

amphotericin B (p=0.005) and itraconazole (p=0.03) against all clinical S. brasiliensis

206

isolates tested, and the reference strain ATCC MYA 4823 (from feline sporotrichosis)

207

was also susceptible to miltefosine treatment. No significant correlation was found

208

between MIC values for amphotericin B, itraconazole and miltefosine (r < 0.4).

209

According to MFC values, miltefosine was also more potent against S. brasiliensis than

210

both amphotericin B and itraconazole (Table 1). Additionally, time kill assays

211

performed using the reference strain (ATCC MYA 4823) showed that miltefosine has

212

fungicidal activity towards S. brasiliensis, with 2 µg.mL-1 (MICx2) of this drug

213

resulting in complete culture growth elimination after as little as 24 hours of treatment

214

(Fig. 1). After prolonged treatment (168 hours, or 7 days) with miltefosine, 1 µg.mL-1

215

(MIC) and 0.5 µg.mL-1 (MIC/2) of this drug inhibited 100% and ~80% (1Log10) of

216

starting inoculum growth, respectively (Fig. 1).

217

Combinations of itraconazole with miltefosine showed no synergistic effect against

218

yeast cells from the isolates ATCC MYA 4823, Ss 37 and B735 (0.5˂ FICI ≤ 4).

219

Exposure to miltefosine disrupts yeast membrane integrity, affects membrane-to-cell

220

wall interactions, and alters cell wall structure, increasing melanin content in the cell

221

wall.

222

To identify early changes in fungal morphology after treatment with miltefosine, S.

223

brasiliensis yeast cells were treated with sub-inhibitory concentration of this drug (0.5

224

µg.mL-1) for 24 hours, and plasma membrane integrity and ultrastructural characteristics

225

were analyzed.

226

Miltefosine promoted loss of plasma membrane integrity in S. brasiliensis yeast cells, as

227

demonstrated by the increase in Sytox blue fluorescence intensity of cell populations

228

(p16 µg mL-1 range) confirm that

288

miltefosine is potent against the tested isolates, and time kill experiments indicated that

289

miltefosine has fungicidal activity against the yeast form of S. brasiliensis, in agreement

290

with that previously described for other fungal species, including Candida sp.,

291

Cryptococcus sp. and Aspergillus sp. (Widmer et al., 2006; Ravu et al., 2013), and also

292

for the mycelial form of S. schenckii complex species (Brilhante et al., 2014).

293

Synergistic effects of miltefosine-azoles combinations have been described for some

294

fungal species (Biswas et al., 2013); however, we did not detected any interactions

295

between itraconazole and miltefosine when treating S. brasiliensis yeast cells from the

296

reference strain ATCC MYA 4823, or from two clinical isolates with high MIC values

297

for itraconazole (Ss 37 and B735).

298

Sytox blue staining of yeasts treated with miltefosine indicates that this phospholipid

299

analogue promotes cell death due to pronounced disturbances in the plasma membrane,

300

among other effects described previously. Miltefosine may have several cellular targets,

301

due to its ubiquitous effects on cell membranes (Barrat et al., 2009). In Trypanossama

302

cruzi and Leishmania species, the drug alters the phospholipid and sterol contents of

303

cells (Barrat et al., 2009), promoting a reduction in the amounts of phospholipids,

304

ergosterol and fatty acids (Dorlo et al., 2012). Despite the antifungal activity of

305

miltefosine and its potential as a therapeutic alternative to treat fungal infections, the

306

mode of action of this drug in fungi is poorly understood. Zuo et al. (2011)

307

demonstrated that miltefosine is rapidly incorporated into Saccharomyces cerevisiae

308

yeast cells, where it crosses the mitochondrial inner membrane, penetrating the

309

mitochondria and leading to an apoptosis-like cell death. In S. brasiliensis, miltefosine

310

may act on the synthesis of membrane components, which may promote instability and

311

loss of membrane integrity, ultimately leading to yeast cell death.

312

We also showed by TEM that the exposure to miltefosine promoted considerable

313

changes in S. brasiliensis yeast cells, including: loss in electron density (Fig. 3C),

314

accumulation of electron dense material in the cell wall (arrow in Fig.3D) and

315

alterations in cell wall thickness (Fig. 3E). Alterations in cell wall components after

316

miltefosine exposure were also described in C. albicans yeasts (Villa et al., 2013).

317

However, the aggregation of electron dense material in the cell wall following

318

miltefosine treatment had not been observed previously for other fungal species. This

319

electron dense material visualized by TEM is morphologically similar to the melanin

320

granules described by Teixeira et al (2010). In addition, flow cytometry analysis using

321

anti-melanin antibodies showed that miltefosine-treated cells had increased melanin

322

content compared to untreated cells. Thus, our results suggest that miltefosine treatment

323

could be modulates melanin turnover (breakdown and/or production) in S. brasiliensis

324

yeast cells, leading to the accumulation of aggregates of this pigment in the cell wall.

325

Similarly, Teixeira et al (2010) also showed that the amount of melanin in the cell wall

326

of S. schenckii could be modulated by varying culture medium composition (Teixeira et

327

al., 2010).

328

Although miltefosine displayed relatively high cytotoxicity towards kidney cells, this is

329

comparable to that observed for amphotericin B, the only available treatment option for

330

severe forms of sporotrichosis. Moreover, in vivo tests with BALB/c mice infected with

331

L. amazonensis showed that miltefosine was toxic only at doses higher than 50 mg kg-1

332

day-1 (Godinho et al., 2012). Given the lower MIC values for miltefosine against S.

333

brasiliensis in vitro, this drug shows higher selective indexes than amphotericin B for

334

the isolates tested (Table 2). Thus, miltefosine is likely to have a wider ‘therapeutic

335

window’ than amphotericin B against S. brasiliensis sporotrichosis, which should allow

336

the use of lower effective doses of this drug, to limit toxicity. Another advantage of

337

miltefosine treatment is that this drug can be administrated orally, whereas amphotericin

338

B treatment is intravenous.

339

Miltefosine is already used to treat some of the clinical manifestations of leishmaniasis

340

(Dorlo et al., 2012), which should facilitate its approval for use in the treatment of other

341

infectious diseases. Interestingly, miltefosine was approved for the topical treatment of

342

metastasized mammary carcinoma in Germany (Paparazafiri et al., 2005), in 1992; thus,

343

topical administration of miltefosine - which might be particularly useful for

344

sporotrichosis treatment – may be also effective in patients. Although some resistance to

345

miltefosine has been described in patients infected with Leishmania donovani, the main

346

resistance mechanisms seems to be associated with a defect in translocation of the drug

347

to the cytoplasm (Perez-Victoria et al., 2003).

348

The dose of miltefosine used for the treatment of leishmaniasis in humans is 2.5 mg kg-1

349

day-1, for 28 days (Dorlo et al., 2012). The reported IC50 value for the treatment of

350

intracellular amastigotes of Leishmania amazonensis with miltefosine in vitro was 9 µM

351

(3.6 µg.mL-1) (Santa-Rita et al., 2004), which is higher than the MIC values of 2.5-5

352

µM (1-2 µg.mL-1) reported here for the treatment of S. brasiliensis isolates. Therefore,

353

our results indicate that lower miltefosine doses than those used against human

354

leishmaniasis could be effective in the treatment of sporotrichosis.

355

In conclusion, ours results show that miltefosine might represent an interesting

356

alternative for the treatment of sporotrichosis caused by S. brasiliensis, especially when

357

standard treatment fails. Nevertheless, further in vivo studies in experimentally infected

358

animals are required to confirm the efficacy of miltefosine as an antifungal agent for the

359

treatment of sporotrichosis.

360

Declaration of interest: The authors report no conflict of interests.

361

Acknowledgements

362

The authors thank Dr. Zoilo Pires de Camargo and Dr. Anderson Messias Rodrigues

363

from the Universidade Estadual de São Paulo (São Paulo, SP, Brazil), Dra. Leila Maria

364

Lopes-Bezerra from Universidade Estadual do Rio de Janeiro (Rio de Janeiro, RJ,

365

Brazil) and Dr. Marcio Nucci from the Universidade Federal do Rio de Janeiro (Rio de

366

Janeiro, RJ, Brazil) for kindly providing the S. brasiliensis isolates used in this study.

367

This study was supported by the Brazilian funding agencies Fundação Carlos Chagas

368

Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Coordenação de

369

Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Conselho Nacional de

370

Desenvolvimento Científico e Tecnológico (CNPq).

371 372

References

373

Alfeltra, J. & Vermeij, P.E. (2003). Antifungal activity of non antifungal drugs. Eur J

374

Clin Microbiol Infect Dis 22, 397-407.

375

Alviano, D.S., Franzen, A.J., Travassos, L.R., Holandino, C., Eizemberg, R.,

376

Alviano, C.S., Rodrigues, M.L. (2004). Melanin from Fonsecaea pedrosoi induces

377

production of human antifungal antibodies and enhances the antimicrobial efficacy of

378

phagocytes. Infect Immun 72, 229-237.

379

Arrigalla-Moncrieff, I., Capilla, J., Mayayo, E., Marimon, R., Mariné, M., Gené,

380

J., Cano, J. & Guarro, J. (2009). Different virulence levels of the species of

381

Sporothrix in murine model. Clin Microbiol Infect 15, 651-655.

382

Barrat, G., Saint-Pierre-Chazalet, M., Loiseau & P.M. (2009). Cellular transport and

383

lipid interactions of miltefosine. Curr Drug Meta 10, 247-255.

384

Barros, M.B.L., Schubach, T.P., Coll J.O., Gremião, I.D., Wanke, B. & Schubach,

385

A. (2010). Sporotrichosis: development and challenges of an epidemic. Rev Panam

386

Salud Publica 27, 455-460 [in Portuguese].

387

Biswas, C., Sorrel., T.C., Djordjevic, J.T., Zou, X., Jolliffe, K.A. & Chen, S.C.

388

(2013). In vitro of miltefosine as a single agent and in combination with voriconazole or

389

posaconazole against uncommon filamentous fungal pathogens. J Antimicrob

390

Chemother, 1-5.

391

Borba-Santos, L.P., Rodrigues, A.M., Gagini, T.B., Fernandes, G.F., Castro, R., de

392

Camargo, Z.P., Nucci, M., Lopes-Bezerra, L.M., Ishida, K. & Rozental, S. (2014).

393

Susceptibility of Sporothrix brasiliensis isolates to amphotericin B, azoles and

394

terbinafine. Medical Mycology published online November 13, 1-11.

395

Brilhante, R.S.N., Malaquias, A.D.M., Caetano, E.P., Castelo-Branco, D.S.C.M., de

396

Lima, R.A.C., Marques, F.J.F., Silva, N.F., de Alencar, L.P., Monteiro, A.J., de

397

Camargo, Z.P., Bandeira, T.J.P.G., Rodrigues, A.M., Cordeiro, R.A., Moreira,

398

J.L.B., Sidrim, J.J.C. & Rocha, M.F.G. (2014). In vitro inhibitory effect of

399

miltefosine against strains of Histoplasma capsulatum var. capsulatum and Sporothrix

400

spp. Med Mycol 52, 320-325.

401

Clinical and Laboratory Standards Institute (2008). Reference Method for Broth

402

Dilution Antifungal Susceptibility Testing of Yeasts, approved standard, M27-A3, 3nd ed.

403

Vilanova: Clinical and Laboratory Standards Institute.

404

Dorlo, T.P.C., Balasegaram, M., Beijnen, J.H. & Vries, P.J. (2012). Miltefosine: a

405

review of its pharmacology and therapeutic efficacy in the treatment of leishmaniasis. J

406

Antimicrob Chemother 67, 2567-2597.

407

Eliopoulos GM, Moellering RC. Antimicrobial combinations. In: Lorian V, editor.

408

Antibiotics in laboratory medicine. 3 ed. Baltimore, MD:Williams&Wilkins Co., 1991:

409

432–92.

410

Fernandes, G. F., dos Santos, P. O., Rodrigues, A. M., Sasaki, A. A., Burger, E., de

411

Camargo, Z. P. (2013). Characterization of virulent profile, protein secretion and

412

immunogenicity of diferente Sporothrix schenckii sensu stricto isolates compared with

413

S. globosa and S. brasiliensis species. Virulence 4, 1-9.

414

Godinho, J.L., Simas-Rodrigues, C., Silva, R., Urmenyi, T.P., De Souza, W. &

415

Rodrigues, J.C. (2012). Efficacy of miltefosine treatment in Leishmania amazonensis-

416

infected BALB/c mice. Int J Antimicrob Agents 39, 326-331.

417

Ishida, K., Mello, J.C.P., Cortez, D.A.G., Dias Filho, B.P., Ueda-Nakamura, T.,

418

Nakamura, C.V. (2006). Influence of Strypnhodendron adstringens tannins on growth

419

and virulence factors of Candida albicans. J Antimicrob Chemother 58, 942–949.

420

Jimenez-Lopes, J.M., Rios-Marco, P., Marco, C., Segovia, J.L. & Carrasco, M.P.

421

(2010). Alterations in the homeostasis of phospholipids and cholesterol by antitumor

422

alkylphospholipis. Lipids Health Dis 9, 9-33.

423

Kauffman, C. A., Bustamante, B., Chapman, S.W., Pappas, P.G. & Infectious

424

Diseases Society of America. (2007). Clinical practice guideline for the management of

425

sporotrichosis: 2007 update by Infectious Diseases Society of America. Clin Infect Dis

426

45, 1255-1265.

427

Klepser, M. E., Ernst, E.J., Lewis, R.E., Ernst, M.E. & Pfaller, M.A. (1998).

428

Influence of Test Conditions on Antifungal Time-Kill Curve Results: Proposal for

429

Standartized Methods. Antimicrob Agents Chemother 42, 1207-1212.

430

Marimon, R., Cano, J., Gené, J., Sutton, D. A., Kawaki, M. & Guarro, J. (2007).

431

Sporothrix brasiliensis, S. globosa, and S. mexicana, three new Sporothrix species of

432

clinical interest. J Clin Microbiol 45, 3198-3206.

433

Marimon, R., Gené, J., Cano, J. & Guarro, J. (2008). Sporothrix luriei: a rare fungus

434

from clinical origin. Med Mycol 46, 621-625.

435

Odds, F.C. (2003). Synergy, antagonism, and what the chequerboard puts between

436

them. J Antimicrob Chemother 52, 1.

437

Oliveira, M. M. E., Almeida-Paes, R., Muniz, M. M., Gutierrez-Galhardo, M. C. &

438

Zancope-Oliveira, R. M. (2011). Phenotypic and molecular identification of

439

Sporothrix isolates from an epidemic area of sporotrichosis in Brazil. Mycopathologia

440

172, 257-267.

441

Ottonelli Stopiglia, C. D., Magagnin, C. M., Castrillón, M. R., Mendes, S. D.,

442

Heidrich, D., Valente, P. & Scroferneker, M. L. (2013). Antifungal susceptibilities

443

and identification of species of the Sporothrix schenckii complex isolated in Brazil. Med

444

Mycol, 52, 56-64.

445

Papazafiri, P., Avlonitis, N., Angelous, N., Calogeropoulou, T., Koufaki, M.,

446

Scoulica, E. & Fragiadaki, I. (2005). Structure-activity relationships of antineoplastic

447

ring-substituted ether phopholipid derivatives. Cancer Chemother Pharmacol 56, 261-

448

270.

449

Pérez-Victoria,

450

donovani resistance to miltefosine involves a defective inward translocation of the drug.

451

Antimicrob. Agents Chemother 47, 2397-2403.

452

Ravu, R.R., Chen, Y.L., Jacob, M.R., Pan, X., Agarwal, A.K., Khan, S.I., Heitman,

453

J., Clark, A.M. & Li, X.C. (2013). Synthesis and antifungal activities of miltefosine

454

analogs. Bioorg Med Chem Lett 23, 4828-4831.

455

Rodrigues, A.M., de Hoog, G.S., Pires, D., Brilhante, R.S., Sidrim, J.J., Gadelha,

456

M.F., Colombo, A.L., de Camargo, Z.P. (2014). Genetic diversity and antifungal

457

susceptibility profiles in causative agents of sporotrichosis. BMC Infect Dis 14, 219.

458

Rodrigues, A.M., de Hoog, S., de Camargo, Z.P. (2013a). Emergence of

459

pathogenicity in the Sporothrix schenckii complex. Med Mycol 51, 405-412.

460

Rodrigues, A. M., Teixeira, M. M., de Hoog, G. S., Schubach, T. M. P., Pereira,

461

S.A., Fernandes, G., F., Bezerra, L. M. L., Felipe, M. S. & de Camargo, Z. P.

462

(2013b). Phylogenetic analysis reveals a high prevalence of Sporothrix brasiliensis in

463

feline sporotrichosis outbreaks. PLoS Negl Trop Dis 7, e2281.

F.

J.,

Castanys, S.

& Gamarro,

F.

(2003).

Leishmania

464

Santa-Rita, R.M., Henrique-Pons, A., Barbosa, H.S. & De Castro, S.L. (2004).

465

Effect of the lysophospholipid analogues edelfosine, imofosine and miltefosine against

466

Leishmania amazonensis. J Antimicrob Chemother 54, 704-710.

467

Teixeira, M.M., de Almeida, L.G.P., Kubitschek-Barreira, P., Alves, F.L.,

468

Kioshima, E.S., Abadio, A.K.R., Fernandes, L., Derengowski, L.S., Ferreira, K.S.,

469

Souza, R.C., Ruiz, J.C., de Andrade, N.C., Paes, H.C., Nicola, A.M., Albuquerque,

470

P., Gerber, A.L., Martins, V.P., Peconick, L.D.F., Neto, A.V., Chaucanez, C.B.,

471

Silva, P.A., Cunha, O.L., de Oliveira, F.F.M., dos Santos, T.C., Barros, A.L.N.,

472

Soares, M.A., de Oliveira, L.M., Marini, M.M., Villalobos-Duno, H., Cunha,

473

M.M.L., de Hoog, S., da Silveira, J.F., Henrissat, B., Niño-Vega, G.A., Cisalpino,

474

P.S., Mora-Montes, H.M., Almeida, S.R., Stajich, J.E., Lopes-Bezerram L.M.,

475

Vasconcelos, A.T.R., Felipe, M.S.S. (2014). Comparative genomics of the major

476

fungal agents of human and animal Sporotrichosis: Sporothrix schenckii and Sporothrix

477

brasiliensis. BMC Genomics 15, 1-22.

478

Teixeira, P.A., de Castro, R.A., Ferreira, F.R., Cunha, M.M., Torres, A.P., Penha,

479

C.V., Rozental., S., Lopes-Bezerra, L.M. (2010). L-DOPA accessibility in culture

480

medium increases melanin expression and virulence of Sporothrix schenckii yeast cells.

481

Med Mycol 48, 687-695.

482

Tong, Z., Widmer, F., Sorrel, T.C., Guse, Z., Jolliffe, K.A., Halliday, C., Lee, O.C.,

483

Kong, F., Wright, L.C. & Chen, S.C. (2007). In vitro activities of miltefosine and two

484

novel antifungal biscationic salts against a panel of 77 dermatophytes. Antimicrob

485

Agents Chemother 51, 2219-2222.

486

Urbina, J.A. (2006). Mechanisms of action of lysophospholipid analogues against

487

trypanossomatid parasites. Trans R Soc Trop Med Hyg 100, S9-S16.

488

Villa, T.V., Ishida, K., de Souza, W., Prousis, K., Calogeropoulou, T., Rozental, S.

489

(2013). Effect of alkylphospholipids on Candida albicans biofilm formation and

490

maturation. J Antimicrob Chemother 68, 113-125.

491

Widmer, F., Wright, L.C., Obando, D., Handke, R., Ganendren, R., Ellis, D.H. &

492

Sorrel, T.C. (2006). Hexadecylphosphocoline (Miltefosine) has broad-spectrum

493

fungicidal activity and is efficacious in a mouse model of Cryptococcosis. Antimicrob

494

Agents Chemother 50, 414-441.

495

Zuo, X., Djordjevic, J.T., Bijosono Oei, J., Desmarini, D., Schibeci, S.D., Jolliffe,

496

K.A., Sorrell, T.C. (2011). Miltefosine induces apoptosis-like cell death in yeast via

497

Cox9p in cytochrome c oxidase. Mol Pharmacol 80,476-85.

498 499 500 501 502 503 504 505 506

507

Table 1. Minimum inhibitory concentration (MIC) and minimum fungicidal

508

concentration (MFC) for the treatment of Sporothrix brasiliensis yeasts with

509

miltefosine, compared to reported values for amphotericin B and itraconazole [Borba-

510

Santos et al., 2014]. All values are in µg mL-1.

S. brasiliensis isolates MYA 4823 B91 B182 B628 B735 B848 B972 B1008 B1036 HE06 Ss 68 Ss 34 Ss 37 Ss 56 511 512 513 514 515 516 517 518 519 520 521 522 523

a

a

Miltefosine MIC 1 2 2 1 2 2 2 2 2 2 1 1 1 1

MFC 2 4 2 1 2 >16 2 2 2 8 1 2 16 1

Amphotericin Bb MIC 0.5 2 1 4 2 4 4 4 4 1 4 4 4 4

MFC 2 16 16 4 2 4 4 4 4 1 4 4 4 4

Itraconazoleb MIC 0.5 8 4 1 16 2 0.5 1 2 8 2 2 4 2

MFC >16 >16 4 16 >16 16 1 1 >16 >16 8 16 8 16

Reference (genome) strain, from feline sporotrichosis. All other strains are from human sporotrichosis. b MIC and MFC values for amphotericin B and itraconazole reported in Borba-Santos et al., 2014.

524

Table 2. Analyses of miltefosine and amphothericin B cytotoxicity towards the

525

epithelial cell line LLC-MK2 (CC50) and selectivity index (SI) for the treatment of

526

Sporothrix brasiliensis yeast cells. LLC-MK2 Antifungal agents

mean MICa -1

CC50 -1

(µg mL )

(µg mL )

Miltefosine

2

5

2.5

Amphotericin B

4

6.25

1.56

527 528

a

Mean MIC values for treatment of the yeast form of S. brasiliensis.

529

b

SI = CC50/MIC.

530

SIb

531

Legends

532

Figure 1. Time-kill plot showing the activity of miltefosine against the yeast form of

533

Sporothrix brasiliensis ATCC MYA 4823. Yeast cell suspensions (103 colony forming

534

units or CFU ml-1) were treated with different concentrations of miltefosine at 35oC,

535

diluted and plated on potato dextrose agar (PDA). Plates were incubated for 7 days at

536

35oC, and CFU numbers were determined by manual counting. Experiments were made

537

in duplicate.

538

Figure 2. Membrane integrity evaluation after treatment of Sporothrix brasiliensis

539

(ATCC MYA 4823) yeast cells with miltefosine, amphotericin B and itraconazole. Cells

540

were left untreated (control) or were treated with 0.5 µg.mL-1 miltefosine, or with 0.25

541

µg.mL-1 of amphotericin B or itraconazole, for 24 hours, before staining with the

542

exclusion dye Sytox blue (Molecular Probes). Treatment with miltefosine only induced

543

a statistically significant loss in plasma membrane integrity compared to untreated

544

controls (**p

Miltefosine is active against Sporothrix brasiliensis isolates with in vitro low susceptibility to amphotericin B or itraconazole.

Sporotrichosis is a common mycosis caused by dimorphic fungi from the Sporothrix schenckii complex. In recent years, sporotrichosis incidence rates ha...
2MB Sizes 0 Downloads 9 Views