This item is the archived peer-reviewed author-version of:

Combination of and domiphen bromide is fungicidal against biofilms of resistant Candida spp.

Reference: Tits Jana, Cools Freya, De Cremer Kaat, De Brucker Katrijn, Berman Judith, Verbruggen Kristof, Gevaert Bert, Cos Paul, Cammue Bruno P.A., Thevissen Karin.- Combination of miconazole and domiphen bromide is fungicidal against biofilms of resistant Candida spp. Antimicrobial agents and chemotherapy - ISSN 0066-4804 - 64:10(2020), e01296-20 Full text (Publisher's DOI): https://doi.org/10.1128/AAC.01296-20 To cite this reference: https://hdl.handle.net/10067/1705020151162165141

Institutional repository IRUA 1 Combination of miconazole and domiphen bromide is fungicidal against biofilms of resistant

2 Candida spp.

3 Jana Titsa, Freya Coolsb, Kaat De Cremera, Katrijn De Bruckera, Judith Bermanc, Kristof

4 Verbruggend, Bert Gevaertd, Paul Cosb, Bruno P.A. Cammuea, Karin Thevissena#

5

6 aCentre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium

7 bLaboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp,

8 Belgium

9 cDepartment of Molecular Microbiology & Biotechnology, School of Molecular Cell Biology and

10 Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.

11 dPurna pharmaceuticals, Puurs, Belgium

12

13 Running Head: Fungicidal combination against resistant Candida spp.

14

15 #Address correspondence to Karin Thevissen, [email protected]

1

16 Abstract

17 The occurrence and recurrence of mucosal biofilm-related Candida , such as oral and

18 vulvovaginal candidiasis, is a serious clinical issue. Vaginal infections caused by Candida spp., for

19 example, affect 70‐75% of women at least once during their lives. Miconazole (MCZ) is the

20 preferred topical treatment against these fungal infections, yet it has only moderate antibiofilm

21 activity. Through screening of a drug repurposing library we identified the quaternary ammonium

22 compound domiphen bromide (DB) as a MCZ potentiator against Candida biofilms. DB displayed

23 synergistic C. albicans antibiofilm activity with MCZ, reducing viable biofilm cells 1000-fold. In

24 addition, the MCZ-DB combination also resulted in significant killing of biofilm cells of azole-

25 resistant C. albicans, C. glabrata, and C. auris isolates. In vivo, the MCZ-DB combination had

26 significantly improved activity in a vulvovaginal candidiasis rat model as compared to single

27 compound treatments. Data from an artificial evolution experiment indicated that resistance

28 development against the combination was not occurring, highlighting the potential of MCZ-DB

29 combination therapy to treat Candida biofilm-related infections.

30

31 Introduction

32 Various fungal species have the capacity to form biofilms, which are structured microbial

33 communities, surrounded by a self-produced extracellular polymer matrix and attached to biotic

34 or abiotic surfaces (1–3). Fungal biofilms are characterized by an increased tolerance to

35 commonly used antimycotics and are therefore difficult to eradicate (1, 3–6). The number of

36 people suffering from biofilm-related fungal infections is increasing, mainly due to rising numbers

2

37 of immunocompromised individuals and increased use of indwelling medical devices, like

38 implants and catheters, on which biofilms can form (7–11).

39 Biofilm-related fungal infections can occur in the oral cavity, the respiratory -and urinary tract, on

40 reproductive organs, in wounds or on medical devices, and are dominated by the genus Candida,

41 of which Candida albicans is the most prevalent (12–17). However, the number of fungal

42 infections caused by non-albicans Candida species is rising with Candida glabrata infections being

43 first in frequency (18–24). C. glabrata is characterized by an innate resistance to azoles, which is

44 the preferred topical treatment against mucosal biofilm-related fungal infections. Hence, C.

45 glabrata mucosal biofilm-related infections are difficult to treat (24–26). Moreover, other

46 emerging Candida spp. present new challenges for antifungal drug development. Candida auris,

47 for instance, is known for its high levels of resistance to multiple antifungal drug classes and its

48 very efficient human-to-human transmissibility (27–31) .

49 Despite the increasing occurrence of fungal biofilm-related infections and their huge impact on

50 the healthcare system, only few novel antifungals with potent antibiofilm activity have been

51 developed during the last decades (12, 32–38). In addition to searches for novel types of

52 antimycotics a promising strategy to develop effective antibiofilm compounds is to increase the

53 activity of conventional antimycotics against fungal biofilms by combining them with a so-called

54 potentiator. This strategy may result in a fungicidal action of the combination against biofilms

55 (39–41). Potentiators can for example induce increased uptake of the antimycotic in the biofilm

56 cells or inhibit biofilm-specific tolerance pathways (42–47).

3

57 The preferred antimycotics for topical treatment against mucosal biofilm-related fungal

58 infections are azoles. They include imidazoles (e.g. miconazole, MCZ) and triazoles (e.g.

59 fluconazole), and interfere with the biosynthesis of ergosterol by inhibiting the enzyme lanosterol

60 14‐alpha‐demethylase. Since ergosterol is a major constituent of the fungal membrane, its

61 depletion results in growth inhibition (48, 49). Additionally, MCZ causes an accumulation of

62 reactive oxygen species in planktonic fungal cultures and distinguishes itself from most other

63 fungistatic azoles by its fungicidal action (50–54). MCZ is a preferred topical treatment of mucosal

64 Candida infections, as intravenously administered MCZ is linked with liver toxicity and phlebitis

65 (55). MCZ at its normal therapeutic level is characterized by fungicidal activity against planktonic

66 Candida cultures, but only at very high doses (5 mM), it is fungicidal against Candida biofilm cells

67 (51, 56). Such high doses can only be achieved in antifungal lock therapy but not therapeutically

68 (57, 58). Hence, increasing miconazole’s fungicidal antibiofilm activity by combining it with a

69 potentiator is highly relevant. This study aimed at identifying compounds that increase the

70 fungicidal activity of MCZ against Candida biofilm cells.

71 We therefore screened 1,311 compounds of a drug repurposing library in combination with a sub-

72 inhibitory concentration of MCZ against mature C. albicans biofilms. The quaternary ammonium

73 compound domiphen bromide (DB) was selected as most promising MCZ potentiator and the

74 MCZ-DB combination was further characterized with regard to in vitro and in vivo activity.

75 Furthermore, as resistance to azoles is occurring (25, 59–62), resistance development against

76 MCZ-DB was studied by experimental evolution of C. glabrata cultures.

77

4

78 Results

79 Screening for MCZ potentiators against C. albicans biofilms. Analogous to our previous study

80 (39), we screened another 1,311 off-patent drugs and bioactive agents (Pharmakon repositioning

81 library) to identify compounds that can enhance the antibiofilm activity of miconazole (MCZ)

82 against mature C. albicans biofilms, after which we assessed potential fungicidal activity of

83 synergistic antibiofilm MCZ-based combinations.

84 In this way, we identified five compounds that resulted in reduced viability of MCZ-treated C.

85 albicans biofilm cells and had primary applications other than as antimycotics (Table 1).

86 Checkerboard analyses and FICI determination confirmed that, out of these five potential hits,

87 only staurosporine, domiphen bromide (DB) and NSC-317926 act synergistically with MCZ to

88 reduce metabolic activity of C. albicans biofilms. These synergistic combinations were

89 characterized by FICI values of 0.49, 0.42 and 0.16, respectively. To determine whether these

90 synergistic MCZ-based combination treatments are fungicidal against C. albicans biofilm cells,

91 survival of biofilm cells after 24 h of drug exposure was assessed by determining CFUs. Maximal

92 concentrations of MCZ and the potentiators that did not result in significant killing of biofilm cells

93 upon single compound treatment were used in combination. These concentrations were

94 determined based on dose-response curves for fungicidal activity of the single compounds (as

95 assessed by CFU determination) and may vary slightly depending on the experimental setup, the

96 tested compounds and the organism in question. When potentiator concentrations are used that

97 are too low, potentiation will not occur, whereas the use of high, active potentiator

98 concentrations leaves little room for improvement, thereby complicating the analyses. DB was

99 most effective in potentiating MCZ toward fungicidal activity against biofilms, resulting in up to

5

100 1000-fold reduction in viable biofilm cells (Fig. 1 and Fig. S1) relative to MCZ alone, and was

101 selected for further analysis.

102 Combination treatment of imidazoles and DB is fungicidal against C. albicans biofilms. To

103 investigate the compound specificity of this combination, the fungicidal activity of combinations

104 consisting of other azoles or other quaternary ammonium compounds against Candida biofilms

105 was investigated. Maximal concentrations of azoles and quaternary ammonium compounds that

106 did not result in significant killing of biofilm cells upon single compound treatment were used in

107 combination.

108 First, we assessed the fungicidal activity of combinations of MCZ with other quaternary

109 ammonium compounds (Fig. 1). A significant reduction in biofilm CFUs was observed upon

110 treatment of the biofilms with MCZ in combination with either DB or dequalinium chloride (DQ),

111 but not in combination with tetraethylammonium bromide (TB), benzethonium chloride (BTC) or

112 benzalkonium chloride (BKC). Treatment of biofilms with MCZ, combined with DQ or DB, resulted

113 in an approximately 100-fold and 1000-fold reduction, respectively, as compared to single

114 compound treatment. DB was selected as the most promising hit because of its most pronounced

115 MCZ potentiating effect.

116 We next tested whether DB also potentiates other imidazoles (e.g. ketoconazole and

117 ) and triazoles (e.g. fluconazole and itraconazole) against C. albicans biofilms.

118 Imidazoles and triazoles are two subclasses of azoles, characterized by 2 -and 3 nitrogen atoms in

119 the azole ring, respectively (63). Significant reductions in viable biofilm CFUs, relative to single

120 treatments or DMSO were only observed for DB combinations with the other imidazoles tested

6

121 (Fig. 1), but not with triazoles. These data indicate that the potentiating activity of DB toward

122 fungicidal antibiofilm activity is imidazole-specific.

123 Combination treatment of MCZ and DB is fungicidal against biofilms of azole-resistant Candida

124 spp. We aimed to gain insights into the combination’s antibiofilm activity spectrum as biofilm-

125 associated fungal infections and resistance occurrence are important problems. Therefore, we

126 further focused our research toward testing of the MCZ-DB combination against biofilms of

127 resistant Candida isolates, namely fluconazole-resistant C. albicans isolates, the intrinsically

128 azole-resistant C. glabrata and C. auris, reported to develop resistance to multiple antifungal drug

129 classes (25–27, 30). We used fluconazole-resistant isolates, characterized by fluconazole and

130 posaconazole MIC values of at least 64 mg/L and 8 mg/L, respectively. For each pathogen,

131 maximal concentrations of DB and MCZ that did not result in significant killing of biofilm cells

132 upon single compound treatment were selected and used in combination. The resulting

133 optimized MCZ-DB combinations were fungicidal against biofilms of all tested azole-resistant

134 Candida spp and resulted in significant CFU reductions as compared to single compound and

135 DMSO treatment (Fig. 2). Moreover, treatment of C. glabrata BG2 biofilms with a higher, active

136 DB concentration combined with MCZ resulted in a more than 100-fold reduction in viable biofilm

137 cells as compared to the DMSO only treatment (Fig. S2).

138 Combination treatment of MCZ and DB is fungicidal against planktonic cultures of C. albicans

139 and C. glabrata. To assess whether DB’s potentiating activity is biofilm-specific, planktonic

140 stationary phase cultures of the most prevalent Candida spp. C. albicans SC5314 and intrinsically

141 azole-resistant C. glabrata BG2 at a density and growth stage, similar to biofilms, were treated

142 with a concentration series of DB and MCZ alone or a combination of both, followed by CFU

7

143 determination. Planktonic cell cultures in stationary phase share some characteristics with

144 biofilms as they have a lower metabolic activity as compared to exponential cultures and are

145 difficult to kill. Hence, stationary Candida cultures with a similar cell density to 48 h old biofilms

146 were used. For each pathogen, maximal concentrations of DB and MCZ that did not result in

147 significant killing of the cells upon single compound treatment were selected and used in

148 combination. The resulting optimized MCZ-DB combinations were fungicidal against planktonic

149 stationary phase cultures of both C. albicans SC5314 and C. glabrata BG2 and resulted in

150 significant CFU reductions as compared to single compound and DMSO treatments (Fig. 3). These

151 data indicate that DB’s potentiating effect on MCZ activity is not biofilm-specific.

152 Combination treatment of MCZ and DB does not result in resistance development. To determine

153 whether resistance against the MCZ-DB combination arises, an artificial evolution experiment was

154 performed. To this end, 3 independent cultures of C. glabrata BG2 (lineages) were consecutively

155 exposed to either MCZ alone (MCZ only-evolved cultures) or to MCZ combined with DB (MCZ-DB

156 evolved cultures). C. glabrata was used, since the growth inhibitory effect of the combination

157 treatment on planktonic C. glabrata cultures was more pronounced than on planktonic C.

158 albicans. The effect of the treatments on the different lineages was assessed after every evolution

159 cycle and doses for subsequent treatment of the evolved cultures were adapted if necessary,

160 thereby maintaining the selection pressure between the treatments and over time. During the

161 whole evolution experiment, we used a treatment that resulted in 35% of yeast growth inhibition

162 (IC35 according to (64)), representing a mild selection pressure that enabled direct inoculation for

163 consecutive rounds of evolution.

8

164 After 61 cycles of evolution (representing more than 400 generations), sensitivity of the MCZ-DB

165 evolved cultures was compared to sensitivity of the starter cultures (cycle 0) toward the MCZ-DB

166 combination (Fig. 4). MCZ-DB treatment of starter cultures and MCZ-DB evolved cultures resulted

167 in a significant CFU reduction as compared to single compound treatments and DMSO only-

168 treatment. Furthermore, the MCZ-DB combination was at least as fungicidal on starter cultures

169 (cycle 0) as on the MCZ-DB evolved cultures (cycle 61). Note that for lineage 1, MCZ-DB evolved

170 cultures were significantly more sensitive to the MCZ-DB combination than the starter cultures.

171 Hence, resistance against the MCZ-DB combination was not observed after more than 60

172 consecutive experimental evolution cycles.

173 To check whether the mild selection pressure was high enough to induce experimental evolution

174 after 61 cycles, we assessed sensitivity of MCZ only-evolved cultures, MCZ-DB evolved cultures

175 and starter cultures to MCZ alone by determining MCZ inhibition halo’s. We observed that the

176 diameter of MCZ growth inhibition halo’s was reduced for all evolved cultures as compared to

177 those of starter cultures, pointing to MCZ resistance development in all cultures (Fig. S3). It could

178 therefore be concluded that the applied mild selection pressure was high enough to induce

179 experimental evolution in our setup. In conclusion, resistance against the MCZ-DB combination is

180 not occurring over time in our setup.

181 DB eliminates tolerance rather than altering the MIC of MCZ in both C. glabrata and C. albicans.

182 Azole tolerance is the appearance of cells that grow slowly at supraMIC concentrations of the

183 drug. For fluconazole in C. albicans, tolerance can be visualized with disk diffusion assays, where

184 susceptibility is measured as the size of the zone of inhibition (ZOI) and tolerance is quantified as

185 the fraction of growth (FoG) within the zone of inhibition (65). Furthermore, a number of adjuvant

9

186 drugs were able to eliminate fluconazole tolerance when added throughout the agar plate with

187 fluconazole in the drug disk (66). Here, we used a modified plating assay, in which MCZ was

188 spotted at the center of plates which did or did not include 2 µM DB (Fig. S4). The size of the ZOI

189 did not change considerably, indicating that the minimal inhibitory concentration to MCZ was not

190 affected by the presence of DB. Rather, addition of DB to the plates cleared most of the tolerance

191 (partial growth within the ZOI) for both C. glabrata and C. albicans isolates. This implies that DB,

192 like inhibitors of Hsp90, calcineurin, PKC and the TOR pathway (66), inhibits tolerance rather than

193 resistance and that its effect on biofilms is likely an effect on drug tolerance as well.

194 DB enhances MCZ efficacy in vivo. To assess the potentiating effect of DB in vivo, a rat vaginitis

195 model was used. This model is based on with C. albicans strain B2630 as infection by

196 this strain results in high fungal burdens (67). This is in contrast to infection with C. albicans strain

197 SC5314 or C. glabrata strain BG2, which both are rapidly cleared by the animals (data not shown).

198 As a therapeutic dosage of 2% MCZ reduced the infection burden in this rat model already with

199 approx. 75% (Fig. S5), an additional potentiating effect by DB would not be notable. Hence, we

200 further used a formulation containing a suboptimal MCZ dose (1%). It is not unusual to use

201 suboptimal doses when studying antifungal combination treatments in vivo. For example, Lafleur

202 and colleagues (2013) showed that a suboptimal dose of fluconazole (0.005%) can be potentiated

203 by AC-17 in an in vivo guinea pig model of cutaneous candidiasis (68). Another study

204 demonstrated a synergistic effect of verapamil and a suboptimal itraconazole dose in vivo,

205 resulting in a reduced fungal lung burden in mice infected by Aspergillus fumigatus (69). To test

206 the MCZ-DB combination in vivo, a formulation containing 1% MCZ nitrate and 0.144 % DB was

207 used. The ratio of these concentrations was chosen based on the in vitro data, pointing to a ratio

10

208 of 1:3-1:6 for optimal effects. As cytotoxicity of MCZ, assessed against the cell line MRC-5, was

209 only slightly affected by the presence of DB using this ratio (Table S1), we proceeded toward the

210 in vivo model. The infection burden over the test period is summarized using the “area under the

211 curve (AUC)” measurement (Fig. 5).

212 First, the efficacy of DB alone was evaluated. This treatment did not significantly change the AUC

213 compared to the vehicle-treated control. Secondly, the efficacy of treatment with MCZ or the

214 combination was assessed. While MCZ alone significantly reduced the AUC from 97.20 (95% CI,

215 91.25 to 103.2) Log(CFU)*h to 36.90 (95% CI, 17.88 to 55.92) Log(CFU)*h compared to the control

216 treatment (p < 0.001, linear mixed model), the combination with DB significantly further reduced

217 the AUC to 11.97 (95% CI, 0.3506 to 23.58) Log(CFU)*h (p < 0.001, linear mixed model).

218 Importantly, the combination proved to be statistically more efficient compared to treatment

219 with MCZ alone (p = 0.0217, linear mixed model).

220 A qualitative histological examination of vaginal tissue after H&E staining revealed changes in the

221 epithelial layer in all samples. Acanthosis (epidermal thickening) and hyperkeratosis were

222 observed in all samples, including in non-infected controls. These cornification effects were a

223 consequence of the administered hormone treatment. Inflammation grades were assigned based

224 on density and depth of inflammatory infiltration in the lamina propria (Fig. 6A-C). In the infected

225 control group, 75% (3/4) of the samples showed inflammation grades II or III in the epithelium. In

226 the MCZ treatment group, 70% (7/10) of all samples showed an inflammation grade II, while in

227 the combination therapy group, inflammation grades II or III were present in only 56% (5/9) of

228 the samples. PAS-D stained sections were then evaluated for presence of Candida (Fig. 6D). While

229 in the non-treated infected controls only 25% (1/4) of all samples stained positive for yeasts, after

11

230 MCZ treatment 50% (5/10) of all samples showed signs of invasive Candida. For the combination

231 therapy, only 11% (1/9) showed signs of Candida infiltration.

232 Discussion

233 Fungal infections can currently be treated by 3 main classes of antimycotics, being azoles,

234 echinocandins and polyenes. However, fungal biofilms are characterized by an increased

235 tolerance to conventional antimycotics, only a few of which (echinocandins and liposomal

236 formulations of ) can be used to treat biofilm-related infections (1, 3–6, 70–72).

237 Biofilms provide several different pathways that protect against the action of miconazole (MCZ),

238 a preferred treatment against mucosal Candida infections. We previously discovered that

239 treatment of C. albicans biofilms with MCZ induces genes classically considered important for

240 antifungal drug resistance: those involved in ergosterol biosynthesis and those encoding drug

241 efflux pumps (2016). Moreover, inhibitors of the electron transport chain act synergistically with

242 MCZ against C. albicans biofilms, but not against planktonic Candida cultures or in oxygen-

243 deprived conditions, suggesting a biofilm-specific oxygen-dependent tolerance mechanism (73).

244 This is consistent with the idea that antifungal tolerance is generally due to changes in stress

245 response pathways (reviewed in (74)).

246 In this study, we sought new combination treatments against biofilm-related Candida infections.

247 The number of patients suffering from such infections is growing. Vaginal infections caused by

248 Candida spp. for example already affect 70‐75% of women at least once during their lives and five

249 to eight percent of these patients subsequently develop recurrent vulvovaginal candidiasis (RVVC)

250 (7–11, 75). Azoles are the preferred treatment for mucosal Candida infections, yet failure to clear

251 the infections treated with azoles is increasingly problematic (25, 59–62). Most azoles are

12

252 fungistatic and not fungicidal, and hence, do not directly result in complete elimination of the

253 Candida cells. In most cases, clearance of (azole‐inhibited) Candida cells is performed by the

254 immune system (51, 76, 77).

255 Biofilm cells phenotypically exhibit an up to 1000-fold higher azole-resistance than planktonic

256 cultures and are less accessible to immune cells, and this contributes to the high recurrence rates

257 associated with fungal biofilm-related infections following antifungal treatment (3, 6, 78–80).

258 Biofilms acquire the ability to grow at higher drug concentrations only when they are in the

259 biofilm state, with growth rates that often differ from those of planktonic cells. This suggests that

260 biofilm drug responses are more akin to antifungal tolerance, which involves phenotypic

261 responses that can differ between isogenic cells as well as between different growth conditions

262 (74). Consistent with this idea, MCZ + DB inhibits MCZ tolerance and does not alter the resistance

263 level (MIC) of the strain. Importantly, the addition of DB to MCZ allows biofilm clearing, due to

264 the shift from the static effect of MCZ vs the cidal effect of the drug combination. Similar effects

265 on tolerance to fluconazole were seen with several different adjuvant drugs, albeit without

266 requiring the biofilm state (65, 66).

267 Thus, novel antibiofilm strategies that can increase the ability to clear fungal infections are

268 needed. In this study, we used an approach in which a potentiator enhances the activity of a

269 conventional antimycotic. Potentiation is often preferred over single compound treatment, since

270 it provides a broader drug activity spectrum, faster antifungal action, lowered dosing of toxic

271 drugs and a reduced risk for the emergence of fungal resistance (81). By screening a drug

272 repurposing library, composed of repurposed compounds that are characterized by a safe toxicity

273 profile and known dosing regimens, we aimed to provide a cost-effective and rapid solution to

13

274 the paucity of available antifungal drugs. Such compounds do not require new clinical trials and

275 the costs of possible drug reformulation are much lower than the cost of developing an entirely

276 new drug (82, 83). In this context, 2‐adamantanamine, a derivative of amantadine (an anti‐

277 influenza A drug also used to treat some of the symptoms of Parkinson’s disease), was previously

278 found to potentiate azole antifungals against C. albicans biofilms (68). Other miconazole

279 potentiators against C. albicans biofilms have been discovered, more specifically antimalarial

280 artemisinins like artesunate and artemether, pyrvinium pamoate (an anthelmintic drug) or

281 hexachlorophene (a disinfectant) (39). However, these combinations appeared to be fungistatic

282 and not fungicidal.

283 In this study, domiphen bromide (DB) was selected as the most promising MCZ potentiator. The

284 MCZ-DB combination is fungicidal and is able to eradicate C. albicans biofilms by killing the cells

285 that compose them, potentially reducing the chance on recurrence. DB potentiates the activity of

286 MCZ and other imidazoles against C. albicans, but not of the tested triazoles, suggesting an

287 imidazole-specific potentiation and the possibility to develop multiple combination treatments

288 using the same compound. Furthermore, MCZ-DB is effective against biofilms of C. glabrata, the

289 second-most common cause of Candida infections after C. albicans, which is difficult to treat due

290 to its inherent azole resistance (18–26). Moreover, MCZ-DB is fungicidal against fluconazole-

291 resistant C. albicans isolates as well as against C. auris, an emerging pathogen characterized by

292 intrinsic resistance against fluconazole and also reported to develop resistance to the other

293 classes of antifungals (27, 30).

294 DB is a quaternary ammonium compound, that acts as a cationic surfactant. Such compounds are

295 commonly used as surface disinfectants (84, 85). For several decades, DB has been used at

14

296 concentrations up to 0.1% in oral care products for daily use, like mouthwashes and toothpastes,

297 and is considered safe for mucosal applications (86). Furthermore, a double-blind placebo

298 controlled study has investigated tolerability and efficacy of DB in 31 patients suffering from acute

299 infectious dental diseases and showed that 0.5 mg/ 4-6 h of DB resulted in significantly less pain

300 and inflammation after 2 days. Apparently, treatment with DB led to a better prognosis, a good

301 response and fewer days of illness (87). Previous studies showed that DB has minimal cytotoxic

302 effects, which was confirmed by our cytotoxicity data (88).

303 We further demonstrated the DB-potentiating effect of MCZ in vivo. Indeed, using a rat

304 vulvovaginal candidiasis model, we showed superior activity of a combination treatment

305 consisting of MCZ and DB as compared to single compound treatment in curing the fungal

306 infection in vivo. Furthermore, histological data showed a similar grade of inflammation in animals

307 treated with MCZ-DB compared to MCZ alone. Importantly, histological data suggest a lower

308 grade of fungal infiltration in vaginal tissue after treatment with combination therapy.

309 Different quaternary ammonium compounds negatively affect cell membranes (89–91).

310 Understanding the mode of action of antifungal compounds or combinations is important, since

311 it may lead to a faster discovery of new antifungal therapies with a similar mode of action or to

312 the optimization of existing ones (92–94). Vacuolar sequestration of azoles is a recently

313 uncovered strategy of azole antifungal resistance, conserved in nonpathogenic and pathogenic

314 yeasts, that is proposed to reduce the effective azole concentration in the cells (95–97). Using

315 confocal microscopy and BCECF,AM staining, we found that DB’s action possibly increases the

316 permeability of the vacuolar membrane due to its surfactant properties, thereby releasing

317 sequestered azoles. However, additional research is required to confirm this hypothesis.

15

318 Finally, as shown for bacterial biofilms by Sass and colleagues, resistance to a single compound

319 and to combination treatment might already occur after 15 cycles of evolution (98). Here, our

320 evolution experiment, which was maintained for more than 60 consecutive testing cycles,

321 indicates the absence of resistance development in cultures that are grown in the presence of the

322 MCZ-DB combination; sensitivity toward the combination was unaffected after more than 400

323 generations under sublethal treatment with the combination. Thus, the MCZ-DB combination has

324 the potential to be developed into a next generation fungicidal and biofilm-active combination

325 therapy.

326

327 Materials and Methods

328 Strains and chemicals. C. albicans strain SC5314 (99), C. glabrata strain BG2 (100), fluconazole-

329 resistant C. albicans isolates Tansir_082 and Tansir_121B (clinical isolates, Prof Katrien Lagrou, UZ

330 Leuven, Belgium), a C. auris isolate (101) and C. albicans B2630 (67) were grown on YPD (1% yeast

331 extract, 2% bacteriological peptone (LabM, UK) and 2% glucose (Sigma-Aldrich, USA)) agar plates

332 at 30°C. MRC-5 cells (67) were used for cytotoxicity testing. Stock solutions of miconazole (MCZ)

333 (Sigma-Aldrich), ketoconazole (TCI Europe, Belgium), clotrimazole (Sigma-Aldrich), itraconazole

334 (TCI) and fluconazole (MP Biomedicals, France) were prepared in dimethyl sulfoxide (DMSO)

335 (VWR International, Belgium). RPMI 1640 medium (pH 7.0) with L-glutamine and without sodium

336 bicarbonate was purchased from Sigma-Aldrich and buffered with MOPS (Sigma-Aldrich). The

337 Pharmakon 1600 repositioning library (Microsource discovery systems, USA) was supplied by the

338 Centre of Drug Design and Discovery (Dr. Patrick Chaltin, KU Leuven, Belgium) and domiphen

16

339 bromide and benzalkonium chloride were purchased from Chemische Fabrik Berg GmbH.

340 Benzethonium chloride and tetraethylammonium bromide were purchased from TCI Europe and

341 dequalinium chloride was purchased from Sigma-Aldrich USA.

342 Antibiofilm screening assay. The antibiofilm screening assay was performed as described

343 previously (39). Briefly, 24-h-old biofilms were treated with 10 µM of MCZ in combination with

344 25 µM of a compound from the Pharmakon 1600 library (2 mM stock solution in DMSO) in RPMI

345 (1,1% DMSO background). After 24 h at 37°C, biofilms were washed, quantified with Cell-Titre

346 Blue (CTB; Promega, USA) and percentage of metabolically active biofilm cells was expressed

347 relative to the control treatment (1,1% DMSO). A confirmation experiment for compounds,

348 resulting in less than 60% residual metabolic activity of C. albicans biofilms in combination with

349 10 µM MCZ compared to the control, was performed twice and compound-only controls were

350 included.

351 Biofilm checkerboard assay. To determine synergistic interactions between antifungal agents

352 and identified potentiators, a checkerboard assay was used, as previously described (39). Briefly,

353 a combination of antifungal compound (e.g. MCZ) and potentiator, two-fold diluted across rows

354 and columns of a microplate respectively, was added to C. albicans biofilms. After a 24 h

355 treatment at 37°C, biofilms were quantified with the CTB method. FICI (fractional inhibitory

356 concentration index) calculations were made to determine synergism (102). The FICI was

357 calculated by the formula FICI=[C(BEC-2A)/ BEC-2A]+[C(BEC-2B)/BEC-2B], in which C(BEC-2A) and

358 C(BEC-2B) are the BEC-2 values of the antifungal drugs in combination, and BEC-2A and BEC-2B are

359 the BEC-2 values of antifungal drugs A and B alone. The interaction was defined as synergistic for

360 a value of FICI≤0.5, indifferent for 0.5

17

361 Fungicidal activity assay. To assess whether a treatment is fungicidal against (i) biofilms or (ii)

362 planktonic cultures, a fungicidal activity assay was performed. (i) Biofilms of C. albicans,

363 fluconazole-resistant C. albicans strains, C. auris and C. glabrata were grown in RPMI and treated

364 with the azole (e.g. MCZ), the quaternary ammonium compound (e.g. DB) or a combination. 24 h

365 or 48 h (C. glabrata) after treatment, CFU determination was performed. To this end, biofilms

366 were washed with PBS, thoroughly scraped off from the bottom of the plate and dissolved in 100

367 µl triton (1%). Serial dilutions were plated on YPD agar plates, followed by an incubation period

368 of 24 h at 37°C and subsequent colony counting. (ii) Planktonic stationary cultures of C. albicans

369 SC5314 or C. glabrata BG2 at OD600nm=1 in RPMI were treated with MCZ, DB or a combination of

370 both. 2,5 h after treatment, shaking at 37°C, CFU determination was performed. To this end,

371 cultures were washed with and dissolved in PBS and serial dilutions were plated on YPD agar

372 plates, followed by an incubation period of 24 h at 37°C and subsequent colony counting.

373 Evolution experiment. The evolution experiment was performed in a volume of 500 µL RPMI

374 medium in a Masterblock® (Greiner bio-one, Germany). During the evolution experiment, we

375 used a treatment that resulted in 35% of growth inhibition on the cultures, representative for a

376 mild selection pressure. 3 independent cultures of C. glabrata BG2 (lineages) were exposed to 61

377 cycles of evolution. One cycle consisted of multiple steps. First, C. glabrata BG2 cells were grown

378 in RPMI medium in the presence of a concentration series of MCZ, either alone or combined with

379 a fixed concentration of 3.125 µM DB. 3.125 µM was the maximal DB concentration that did not

380 inhibit growth of C. glabrata BG2 cultures itself. Subsequently, optical density at 490 nm was

381 measured to determine the IC35 of MCZ. Finally, the culture grown in the IC35 of MCZ (OD600 of

382 approx. 1.0) was 100-fold diluted in RPMI and used to inoculate a subsequent evolution cycle.

18

383 Each cycle consisted of approx. 7 generations of C. glabrata, resulting in more than 400

384 generations over the entire experiment.

385 Halo assay and modified plating assay. A halo assay was performed to check whether the

386 selection pressure, applied in the evolution experiment, was high enough to induce experimental

387 evolution. To this end, overnight cultures of C. glabrata BG2 evolved and starter cultures were

388 diluted to an OD600nm=0.1 in YPD agar. 5 µL of a MCZ concentration series, dissolved in DMSO,

389 was spotted on the resulting YPD agar plates. After an incubation period of 24 h at 37°C, inhibitory

390 halos of MCZ were measured and pictures were taken.

391 To study the effect of DB on tolerance and MIC of MCZ, a modified plating assay was performed.

392 Here, C. albicans SC5314 and C. glabrata BG2 overnight cultures were diluted to an OD600nm=0.1

393 in YPD agar containing 2 µM DB (final DMSO concentration of 0.1%). 5 µL of 50 µM MCZ, dissolved

394 in DMSO, was spotted on the agar plates, followed by an incubation period of 24 h at 37°C.

395 Cytotoxicity assay. MRC-5 cells were grown into polystyrene 96-well plates at an initial

396 concentration of 1.5 * 105 cells/mL cells per well and incubated at 37°C and 5% CO2. In each well,

397 190 µL of cell suspension was added together with 10 µL of watery compound dilutions. Cell

398 growth was compared to untreated control wells (100% cell growth) and medium-control wells

399 (0% cell growth). After 3 days of incubation, cell viability was assessed using 10 µg/mL resazurin.

400 After an additional incubation of 4 hours, fluorescence was measured at λemission = 590 nm, λexcitation

401 = 550 nm using a spectrophotometer (Promega Discover) as described earlier. A compound is

402 classified non-toxic when the IC50 is greater than 20 µM. Tamoxifen was used as a positive

403 control.

19

404 Formulation for vaginitis rat model. A vaginal cream was developed for the application of MCZ,

405 DB or MCZ-DB. For all applications the viscosity of the corresponding cream was identical. As DB

406 affects viscosity of the cream, the formulation was slightly adapted as described below. First, the

407 placebo creams (Table 2) were prepared by heating the cetostearyl alcohol with the mineral oil,

408 the lauroyl polyoxyl-6 glycerides and the PEG-and glycol stearate to 70°C. Once the water had a

409 temperature of 70°C, it was mixed with the oil phase and cooled down under stirring. Once the

410 temperature was below 25 °C, water was added until final weight. After preparing the placebo

411 creams, MCZ nitrate and/or DB were added to obtain the final formulations. In this way the

412 following samples were prepared: 1% w/w MCZ nitrate cream, 0.144% w/w DB cream

413 (corresponding to 1/6 of the molar concentration of 1% w/w MCZ nitrate) and 1% w/w MCZ

414 nitrate with 0.144% w/w DB cream. After production, 0.2 mL of the different formulations was

415 filled into a 1 mL syringe (VWR) ready for application. Creams were prepared 3 days before

416 application and stored at 5°C until application.

417 Vaginitis rat model. All animal experiments were approved by the Ethical Commission of the

418 University of Antwerp (UA-ECD 2018-80) and carried out in strict accordance to all mandatory

419 guidelines (European Union directive 2010/63/EU on the protection of animals used for scientific

420 purposes and the Declaration of Helsinki). Female Wistar rats (± 180 g; Janvier Labs) were

421 ovariectomized 2 weeks prior to infection (67). To induce artificial oestrus, 17β-oestradiol 3-

422 benzoate (Sigma-Aldrich) and progesterone (Sigma-Aldrich) were administered 3 days prior to the

423 infection. Hormone treatment was repeated 2 and 7 days post-infection to maintain oestrus. At

424 the day of infection, 100 µL of an 1 x 108 CFU/mL C. albicans B2630 suspension was applied

425 vaginally. Vaginal swabs were taken at several days p.i. to assess the infection burden.

20

426 Simultaneously, oestrus was monitored by Giemsa staining of a fixated vaginal smear. Rats were

427 treated twice per day starting 24 hours after infection for 6 consecutive days. For the evaluation

428 of treatment with DB alone, 8 animals in total per group were used in 2 independent experiments.

429 Swabs were taken at 4, 9 and 14 days p.i.. To evaluate treatment with MCZ or the combination, 3

430 independent repeats with a total of 24 animals per treatment group were used. Vaginal swabs

431 were taken at 4, 7, 11 and 14 days p.i.. In all experiments, 4 vehicle-treated rats were used as

432 control animals after initial validation of the experimental setup of the controls. Group averages

433 of intravaginal burdens were used to plot graphs, and the area under the infection curve (AUC)

434 was calculated for each animal as a measure for infection burden.

435 Vaginal tissue collection and processing. Vaginal tissue was collected 14 days p.i.. Full-tissue

436 thickness samples were fixed in phosphate-buffered 4% paraformaldehyde for 4 hours and stored

437 in PBS at 4°C before processing. Tissue was subsequently dehydrated in graded ethanol solutions

438 and embedded in paraffin. Samples were cut into 4.5 µm sections and stained using routine

439 hematoxylin and eosin (H&E) method and periodic acid-Schiff-diastase (PAS-D) method to

440 observe tissue damage and candidial infiltration. Samples were observed by an independent

441 histopathologist (Histogenex, Belgium) and severity of inflammation was scored as grade I, II or

442 III based on density and depth of inflammatory infiltrations in the lamina propria as seen in the

443 H&E staining (Table 3).

444 Data analysis. GraphPad Prism 6 was used to analyze all in vitro data. Significant differences in

445 CFUs were determined on the mean log values with P < 0.05 considered as statistically significant.

446 Significant CFU reductions were determined by means of a two-way ANOVA, followed by the

447 appropriate multiple comparisons test. For the in vivo experiments, the area under curve (AUC)

21

448 was calculated for each individual rat based upon the natural logarithm of the burden plus 1, to

449 provide a summary value of the cumulative burden over all time points. Since batch effects cannot

450 be excluded, a linear mixed model was used, with AUC as dependent variable, treatment as fixed

451 effect and batch as random effect. This latter term corrects for the non-independence between

452 observations within the same batch. A posthoc analysis with Tukey correction for multiple testing

453 was used to assess pairwise differences between groups. Analysis of the in vivo data was carried

454 out using R, version 3.5.1 (R Core team (2018). R: A language and environment for statistical

455 computing. R Foundation for Statistical Computing, Vienna, Austria. URL https://www.R-

456 project.org/).

457 Acknowledgements

458 We want to thank Prof Katrien Lagrou, UZ Leuven, Belgium for providing the C. auris isolate and

459 the fluconazole-resistant C. albicans isolates. KT acknowledges receipt of an IOF (Industrial

460 Research Fund) mandate of the KU Leuven. Research was supported by VLAIO (grant

461 HBC.2018.0420) to BC, KT, PC, KV. The funders had no role in study design, data collection and

462 interpretation, or the decision to submit the work for publication.

463 References

464 1. Donlan RM. 2002. Biofilms : Microbial Life on Surfaces. Emerg Infect Dis 8:881–890.

465 2. Hall-stoodley L, Costerton JW, Stoodley P. 2004. Bacterial biofilms: from the natural

466 environment to infectious diseases. Nat Rev Microbiol 2:95–108.

467 3. Ramage G, Rajendran R, Sherry L, Williams C. 2012. Fungal Biofilm Resistance. Int J

468 Microbiol 2012:528521.

22

469 4. Lewis K. 2001. Riddle of Biofilm Resistance. Antimicrob Agents Chemother 45:999–1007.

470 5. Hawser SP, Douglas LJ. 1995. Resistance of Candida albicans Biofilms to Antifungal Agents

471 In Vitro. Antimicrob Agents Chemother 39:2128–2131.

472 6. Delattin N, Cammue BP, Thevissen K. 2014. Reactive oxygen species-inducing antifungal

473 agents and their activity against fungal biofilms. Future Med Chem 6:77–90.

474 7. Cauda R. 2009. Candidaemia in Patients with an Inserted Medical Device. Drugs 69

475 Suppl.1:S33-38.

476 8. Lebeaux D, Fernández-hidalgo N, Chauhan A, Lee S, Ghigo J, Almirante B, Beloin C. 2014.

477 Management of infections related to totally implantable venous-access ports : challenges

478 and perspectives. Lancet Infect Dis 14:146–159.

479 9. Tumbarello M, Fiori B, Trecarichi EM, Posteraro P, Losito AR, De Luca A, Sanguinetti M,

480 Fadda G, Cauda R, Posteraro B. 2012. Risk Factors and Outcomes of Candidemia Caused

481 by Biofilm-Forming Isolates in a Tertiary Care Hospital. PLoS One 7:e33705.

482 10. Rautemaa R, Ramage G. 2011. Oral candidosis – Clinical challenges of a biofilm disease.

483 Crit Rev Microbiol 37:328–336.

484 11. Alves MJ, Barreira JCM, Carvalho I, Trinta L, Perreira L, Ferreira ICFR, Pintado M. 2014.

485 Propensity for biofilm formation by clinical isolates from urinary tract infections :

486 developing a multifactorial predictive model to improve antibiotherapy. J Med Microbiol

487 63:471–477.

488 12. Ramage G, Williams C. 2013. The Clinical Importance of Fungal Biofilms, Adv Appl

23

489 Microbiol 84:27-83

490 13. Corsello S, Spinillo A, Osnengo G, Penna C, Guaschino S, Beltrame A, Blasi N, Festa A.

491 2003. An epidemiological survey of vulvovaginal candidiasis in Italy. Eur J Obstet Gynecol

492 Reprod Biol 110:66–72.

493 14. Gökmanoğlu C, Kara N, Beldüz M, Kamburoğlu A, Tosun İ, Sadik E, Kara C. 2018.

494 Evaluation of Candida Albicans Biofilm Formation on Various Parts of Implant Material

495 Surfaces. Niger J Clin Pract 21:33–37.

496 15. Gajdács M, Dóczi I, Ábrók M, Lázár A, Burián K. 2019. Epidemiology of candiduria and

497 Candida urinary tract infections in inpatients and outpatients : results from a 10-year

498 retrospective survey. Cent European J Urol 72:209–214.

499 16. Shweihat Y, Perry Iii J, Shah D. 2015. Isolated Candida infection of the lung. Respir Med

500 Case Rep 16:18–19.

501 17. Nobile CJ, Johnson AD. 2015. Candida albicans Biofilms and Human Disease. Annu Rev

502 Microbiol 69:71–92.

503 18. Deorukhkar SC, Saini S, Mathew S. 2014. Non- albicans Candida Infection : An Emerging

504 Threat. Interdiscip Perspect Infect Dis 2014:615958.

505 19. Spinillo A, Capuzzo E, Gulminetti R, Marone P, Colonna L, Piazzi G. 1997. Prevalence of

506 and risk factors for fungal vaginitis caused by non-albicans species. Am J Obstet Gynecol

507 176:138–141.

508 20. Tortorano AM, Prigitano A, Lazzarini C, Passera M, Deiana ML, Cavinato S, De Luca C,

24

509 Grancini A, Lo Cascio G, Ossi C, Sala E, Montagna MT. 2013. A 1-year prospective survey

510 of candidemia in Italy and changing epidemiology over one decade. Infection 41:655–662.

511 21. Pfaller MA, Jones RN, Messer SA, Edmond MB, Wenzel RP, SCOPE Participant Group.

512 1998. National Surveillance of Nosocomial Blood Stream Infection Due to Species of

513 Candida Other than Candida albicans : Frequency of Occurrence and Antifungal

514 Susceptibility in the SCOPE Program. Diagn Microbiol Infect Dis 30:121–129.

515 22. Pfaller MA, Jones RN, Doern G V, Sader HS, Messer SA, Houston A, S C, Hollis RJ, SENTRY

516 Participant Group. 2000. Bloodstream Infections Due to Candida Species : SENTRY

517 Antimicrobial Surveillance Program in North America and Latin America , 1997-1998.

518 Antimicrob Agents Chemother 44:747–751.

519 23. Borg-von Zepelin M, Kunz L, Rüchel R, Reichard U, Weig M, Groß U. 2007. Epidemiology

520 and antifungal susceptibilities of Candida spp . to six antifungal agents : results from a

521 surveillance study on fungaemia in Germany from July 2004 to August 2005. J Antimicrob

522 Chemother 60:424–428.

523 24. Pfaller MA, Jones RN, Castanheira M. 2014. Regional data analysis of Candida non-

524 albicans strains collected in United States medical sites over a 6-year period, 2006 – 2011.

525 Mycoses 57:602–611.

526 25. Oxman DA, Chow JK, Frendl G, Hadley S, Hershkovitz S, Ireland P, McDermott LA, Tsai K,

527 Marty FM, Kontoyiannis DP, Golan Y. 2010. Candidaemia associated with decreased in

528 vitro fluconazole susceptibility: is Candida speciation predictive of the susceptibility

529 pattern? J Antimicrob Chemother 65:1460–1465.

25

530 26. White DJ, Habib AR, Vanthuyne A, Langford S, Symonds M. 2001. Combined topical

531 flucytosine and amphotericin B for refractory vaginal Candida glabrata infections. Sex

532 Transm Infect 77:212–213.

533 27. Lockhart SR, Etienne KA, Vallabhaneni S, Farooqi J, Chowdhary A, Govender NP, Colombo

534 AL, Calvo B, Cuomo CA, Desjardins CA, Berkow EL, Castanheira M, Magobo RE, Jabeen K,

535 Asghar RJ, Meis JF, Jackson B, Chiller T, Litvintseva AP. 2017. Simultaneous Emergence of

536 Multidrug-Resistant Candida auris on 3 Continents Confirmed by Whole-Genome

537 Sequencing and Epidemiological Analyses. Clin Infect Dis 64:134–140.

538 28. Biswal M, Rudramurthy SM, Jain N, Shamanth AS, Sharma D, Jain K, Yaddanapudi LN,

539 Chakrabarti A. 2017. Controlling a possible outbreak of Candida auris infection : lessons

540 learnt from multiple interventions. J Hosp Infect 97:363–370.

541 29. Piedrahita CT, Cadnum JL, Jencson AL, Shaikh AA, Ghannoum MA, Donskey CJ. 2017.

542 Environmental Surfaces in Healthcare Facilities are a Potential Source for Transmission of

543 Candida auris and Other Candida Species. Infect Control Hosp Epidemiol 38:1107–1109.

544 30. Welsh RM, Bentz ML, Shams A, Houston H, Lyons A, Rose LJ, Litvintseva AP. 2017.

545 Survival, Persistence, and Isolation of the Emerging Multidrug-Resistant Pathogenic Yeast

546 Candida auris on a Plastic Health Care Surface. J Clin Microbiol 55:2996–3005.

547 31. Tsay S, Welsh RM, Adams EH, Chow NA, Gade L, Berkow EL, Poirot E, Lutterloh E, Quinn

548 M, Chaturvedi S, Kerins J, Black SR, Kemble SK, Barrett PM, Barton K, Shannon DJ, Bradley

549 K, Lockhart SR, Litvintseva AP, Moulton-Meissner H, Shugart A, Kallen A, Vallabhaneni S,

550 Chiller TM, Jackson BR. 2017. Ongoing Transmission of Candida auris in Health Care

26

551 Facilities - United States, June 2016-May 2017. Morb Mortal Wkly Rep 66:514–515.

552 32. Brown GD, Denning DW, Gow NAR, Levitz SM, Netea MG, White TC. 2012. Hidden Killers :

553 Human Fungal Infections. Sci Transl Med 4:165rv13.

554 33. Harriott MM, Noverr MC. 2011. Importance of Candida – bacterial polymicrobial biofilms

555 in disease. Trends Microbiol 19:557–563.

556 34. Donelli G, Vuotto C. 2014. Biofilm-based infections in long-term care facilities. Future

557 Microbiol 9:175–188.

558 35. Darouiche RO. 2004. Treatment of Infections Associated with Surgical Implants. N Engl J

559 Med 350:1422–1429.

560 36. Butler MS, Cooper MA. 2011. Antibiotics in the clinical pipeline in 2011. J Antibiot (Tokyo)

561 64:413–425.

562 37. Roemer T, Krysan DJ. 2014. Antifungal Drug Development : Challenges , Unmet Clinical

563 Needs , and New Approaches. Cold Spring Harb Perspect Med 4:a019703.

564 38. Ostrosky-zeichner L, Casadevall A, Galgiani JN, Odds FC, Rex JH. 2010. An insight into the

565 antifungal pipeline : selected new molecules and beyond. Nat Rev Drug Discov 9:719–

566 727.

567 39. De Cremer K, Lanckacker E, Cools TL, Bax M, De Brucker K, Cos P, Cammue BPA, Thevissen

568 K. 2015. Artemisinins , New Miconazole Potentiators Resulting in Increased Activity

569 against Candida albicans Biofilms. Antimicrob Agents Chemother 59:421–426.

570 40. Onyewu C, Blankenship JR, Poeta M Del, Heitman J. 2003. Ergosterol Biosynthesis

27

571 Inhibitors Become Fungicidal when Combined with Calcineurin Inhibitors against Candida

572 albicans , Candida glabrata , and Candida krusei. Antimicrob Agents Chemother 47:956–

573 964.

574 41. Marchetti O, Entenza JM, Sanglard D, Bille J, Glauser MP, Moreillon P. 2000. Fluconazole

575 plus Cyclosporine : a Fungicidal Combination Effective against Experimental Endocarditis

576 Due to Candida albicans. Antimicrob Agents Chemother 44:2932–2938.

577 42. Vallières C, Raulo R, Dickinson M, Avery S V. 2018. Novel Combinations of Agents

578 Targeting Translation That Synergistically Inhibit Fungal Pathogens. Front Microbiol

579 9:2355.

580 43. Eckert R, Brady KM, Greenberg EP, Qi F, Yarbrough DK, He J, Mchardy I, Anderson MH, Shi

581 W. 2006. Enhancement of Antimicrobial Activity against Pseudomonas aeruginosa by

582 Coadministration of G10KHc and Tobramycin. Antimicrob Agents Chemother 50:3833–

583 3838.

584 44. Brackman G, Cos P, Maes L, Nelis HJ, Coenye T. 2011. Quorum Sensing Inhibitors Increase

585 the Susceptibility of Bacterial Biofilms to Antibiotics In Vitro and In Vivo. Antimicrob

586 Agents Chemother 55:2655–2661.

587 45. Gawande P V, Leung KP, Madhyastha S. 2014. Antibiofilm and Antimicrobial Efficacy of

588 DispersinB-KSL-W Peptide-Based Wound Gel Against Chronic Wound Infection Associated

589 Bacteria. Curr Microbiol 68:635–641.

590 46. Maisetta G, Grassi L, Di Luca M, Bombardelli S, Medici C, Brancatisano FL, Esin S, Batoni G.

591 2016. Anti-biofilm properties of the antimicrobial peptide temporin 1Tb and its ability , in

28

592 combination with EDTA , to eradicate Staphylococcus epidermidis biofilms on silicone

593 catheters. Biofouling 32:787–800.

594 47. Martins M, Henriques M, Lopez-Ribot JL, Oliveira R. 2012. Addition of DNase Improves

595 the In Vitro Activity of Antifungal Drugs against Candida albicans Biofilms. Mycoses

596 55:80–85.

597 48. Hitchcock CA, Dickinson K, Brown SB, Evans EG V, Adams DJ. 1990. Interaction of azole

598 antifungal antibiotics with cytochrome P-450- dependent 14a-sterol demethylase purified

599 from Candida albicans. Biochem J 266:475–480.

600 49. Sud IJ, Feingold DS. 1981. Mechanisms of Action of the Antimycotic Imidazoles. J Invest

601 Dermatol 76:438–441.

602 50. Kobayashi D, Kondo K, Uehara N, Otokozawa S, Tsuji N, Yagihashi A, Watanabe N. 2002.

603 Endogenous Reactive Oxygen Species Is an Important Mediator of Miconazole Antifungal

604 Effect. Antimicrob Agents Chemother 46:3113–3117.

605 51. François IEJA, Cammue BPA, Borgers M, Ausma J, Dispersyn GD, Thevissen K. 2006.

606 Azoles : Mode of Antifungal Action and Resistance Development. Effect of Miconazole on

607 Endogenous Reactive Oxygen Species Production in Candida albicans. Antiinfect Agents

608 Med Chem 5:3–13.

609 52. Yan L, Li M, Cao Y, Gao P, Cao Y, Wang Y, Jiang Y. 2009. The alternative oxidase of Candida

610 albicans causes reduced fluconazole susceptibility. J Antimicrob Chemother 64:764–773.

611 53. Thevissen K, Ayscough KR, Aerts AM, Du W, De Brucker K, Meert EMK, Ausma J, Borgers

29

612 M, Cammue BPA, François IEJA. 2007. Miconazole induces changes in actin cytoskeleton

613 prior to reactive oxygen species induction in yeast. J Biol Chem 282:21592–21597.

614 54. Yan L, Zhang JD, Cao YB, Gao PH, Jiang YY. 2007. Proteomic analysis reveals a metabolism

615 shift in a laboratory fluconazole-resistant Candida albicans strain. J Proteome Res 6:2248–

616 2256.

617 55. Fischer TJ, Klein RB, Kershnar HE, Borut TC, Stiehm ER. 1977. Miconazole in the treatment

618 of chronic mucocutaneous candidiasis: A preliminary report. J Pediatr 91:815–819.

619 56. Vandenbosch D, Braeckmans K, Nelis HJ, Coenye T. 2010. Fungicidal activity of

620 miconazole against Candida spp. biofilms. J Antimicrob Chemother 65:694–700.

621 57. Schinabeck MK, Long LA, Hossain MA, Chandra J, Mukherjee PK, Mohamed S, Ghannoum

622 MA. 2004. Rabbit Model of Candida albicans Biofilm Infection: Liposomal Amphotericin B

623 Antifungal Lock Therapy. Antimicrob Agents Chemother 48:1727–1732.

624 58. Buckler BS, Sams RN, Goei VL, Krishnan KR, Bemis MJ, Parker DP, Murray DL. 2008.

625 Treatment of central venous catheter fungal infection using liposomal amphotericin-B

626 lock therapy. Pediatr Infect Dis J 27:762–764.

627 59. Marchaim D, Lemanek L, Bheemreddy S, Kaye KS, Sobel JD. 2012. Fluconazole-Resistant

628 Candida albicans Vulvaginitis. Obstet Gynecol 120:1407–1414.

629 60. Gallè F, Sanguinetti M, Colella G, Di Onofrio V, Torelli R, Rossano F, Liguori G. 2011. Oral

630 candidosis : characterization of a sample of recurrent infections and study of resistance

631 determinants. New Microbiol 34:379–389.

30

632 61. Thomaz DY, de Almeida Jr JN, Lima GME, de Oliveira Nunes M, Camargo CH, de Carvalho

633 Grenfell R, Benard G, Del Negro GMB. 2018. An Azole-Resistant Candida parapsilosis

634 Outbreak : Clonal Persistence in the Intensive Care Unit of a Brazilian Teaching Hospital.

635 Front Microbiol 9:2997.

636 62. Shyamala R, Parandekar PK. 2014. Identification and In vitro Azole resistance of Candida

637 species isolated from Oropharyngeal Candidiasis in Human Immunodeficiency Virus

638 infected patients. Int J Curr Microbiol Appl Sci 3:816–822.

639 63. Cleary JD, Taylor JW, Chapman SW. 1990. Imidazoles and triazoles in antifungal therapy.

640 DICP 24:148–152.

641 64. Van Dijck P, Sjollema J, Cammue BPA, Lagrou K, Berman J, D’Enfert C, Andes DR, Arendrup

642 MC, Brakhage AA, Calderone R, Cantón E, Coenye T, Cos P, Cowen LE, Edgerton M,

643 Espinel-Ingroff A, Filler SG, Ghannoum M, Gow NAR, Haas H, Jabra-Rizk MA, Johnson EM,

644 Lockhart SR, Lopez-Ribot JL, Maertens J, Munro CA, Nett JE, Nobile CJ, Pfaller MA,

645 Ramage G, Sanglard D, Sanguinetti M, Spriet I, Verweij PE, Warris A, Wauters J, Yeaman

646 MR, Zaat SAJ, Thevissen K. 2018. Methodologies for in vitro and in vivo evaluation of

647 efficacy of antifungal and antibiofilm agents and surface coatings against fungal biofilms.

648 Microb Cell 5:300–326.

649 65. Gerstein AC, Rosenberg A, Hecht I, Berman J. 2016. DiskImageR: Quantification of

650 resistance and tolerance to antimicrobial drugs using disk diffusion assays. Microbiology

651 162:1059–1068.

652 66. Rosenberg A, Ene I V., Bibi M, Zakin S, Segal ES, Ziv N, Dahan AM, Colombo AL, Bennett

31

653 RJ, Berman J. 2018. Antifungal tolerance is a subpopulation effect distinct from resistance

654 and is associated with persistent candidemia. Nat Commun 9:2470.

655 67. de Wit K, Paulussen C, Matheeussen A, Rossem K Van, Cos P, Maes L. 2010. In Vitro

656 Profiling of Pramiconazole and In Vivo Evaluation in Microsporum canis Dermatitis and

657 Candida albicans Vaginitis Laboratory Models. Antimicrob Agents Chemother 54:4927–

658 4929.

659 68. Lafleur MD, Sun L, Lister I, Keating J, Nantel A, Long L, Ghannoum M, North J, Lee RE,

660 Coleman K, Dahl T, Lewis K. 2013. Potentiation of Azole Antifungals by 2-

661 Adamantanamine. Antimicrob Agents Chemother 57:3585–3592.

662 69. Zeng Q, Zhang Z, Chen P, Long N, Lu L, Sang H. 2019. In vitro and in vivo efficacy of a

663 synergistic combination of itraconazole and verapamil against Aspergillus fumigatus.

664 Front Microbiol 10:1266.

665 70. Fiori B, Posteraro B, Torelli R, Tumbarello M, Perlin DS, Fadda G, Sanguinetti M. 2011. In

666 Vitro Activities of Anidulafungin and Other Antifungal Agents against Biofilms Formed by

667 Clinical Isolates of Different Candida and Aspergillus Species. Antimicrob Agents

668 Chemother 55:3031–3035.

669 71. Uppuluri P, Srinivasan A, Ramasubramanian A, Lopez-ribot JL. 2011. Effects of Fluconazole

670 , Amphotericin B , and Caspofungin on Candida albicans Biofilms under Conditions of Flow

671 and on Biofilm Dispersion. Antimicrob Agents Chemother 55:3591–3593.

672 72. Kuhn DM, George T, Chandra J, Mukherjee PK, Ghannoum MA. 2002. Antifungal

673 Susceptibility of Candida Biofilms: Unique Efficacy of Amphotericin B Lipid Formulations

32

674 and Echinocandins. Antimicrob Agents Chemother 46:1773–1780.

675 73. De Cremer K, De Brucker K, Staes I, Peeters A, Van Den Driessche F, Coenye T, Cammue

676 BPA, Thevissen K. 2016. Stimulation of superoxide production increases fungicidal action

677 of miconazole against Candida albicans biofilms. Sci Rep 6:27463.

678 74. Berman J, Krysan DJ. Drug resistance and tolerance in fungi. Nat Rev Microbiol 18:319-

679 331.

680 75. Sobel JD. 2007. Vulvovaginal candidosis. Lancet 369:1961–1971.

681 76. Manavathu EK, Cutright JL, Chandrasekar PH. 1998. Organism-Dependent Fungicidal

682 Activities of Azoles. Antimicrob Agents Chemother 42:3018–3021.

683 77. Ha KC, White TC. 1999. Effects of Azole Antifungal Drugs on the Transition from Yeast

684 Cells to Hyphae in Susceptible and Resistant Isolates of the Pathogenic Yeast Candida

685 albicans. Antimicrob Agents Chemother 43:763–768.

686 78. Lamfon H, Porter SR, Mccullough M, Pratten J. 2004. Susceptibility of Candida albicans

687 biofilms grown in a constant depth film fermentor to , fluconazole and

688 miconazole : a longitudinal study. J Antimicrob Chemother 53:383–385.

689 79. Al-fattani MA, Douglas LJ. 2006. Biofilm matrix of Candida albicans and Candida

690 tropicalis : chemical composition and role in drug resistance. J Med Microbiol 55:999–

691 1008.

692 80. Watnick P, Kolter R. 2000. Biofilm , City of Microbes. J Bacteriol 182:2675–2679.

693 81. Bink A, Pellens K, Cammue BPA, Thevissen K. 2011. Anti-Biofilm Strategies : How to

33

694 Eradicate Candida Biofilms ? The Open Mycology Journal 5:29–38.

695 82. Sekhon BS. 2013. Repositioning drugs and biologics : Retargeting old / existing drugs for

696 potential new therapeutic applications. J Pharm Educ Res 4:1–15.

697 83. Azevedo MM, Teixeira-Santos R, Pilva AP, Cruz L, Ricardo E, Pina-Vaz C, Rodrigues AG.

698 2015. The effect of antibacterial and non-antibacterial compounds alone or associated

699 with antifugals upon fungi. Front Microbiol 6:669.

700 84. Simoes M, Pereira MO, Vieira MJ. 2005. Action of a cationic surfactant on the activity and

701 removal of bacterial biofilms formed under different flow regimes. Water Res 39:478–

702 486.

703 85. Gerba CP. 2015. Quaternary Ammonium Biocides : Efficacy in Application. Appl Environ

704 Microbiol 81:464–469.

705 86. Federal Register. 1994. Oral Health Care Drug Products for Over-the-Counter Human Use;

706 Tentative Final Monograph for Oral Drug Products; Proposed Rule. Department

707 of Health and Human Services. Food and Drug Administration, Part II. Fed. Regist.

708 59(27):6096–6097 (codified at 21 CFR Part 356).

709 87. Scaglione F, Coduri M, Nebuloni R, Fraschini F. 1983. Activity and Tolerability of Domifen

710 Bromide in Patients Affected by Acute Infectious Dental Diseases: A Double-Blind

711 Placebo-Controlled Trial. Int J Clin Pharmacol Res 3:357–361.

712 88. Hayes MP, Soto-Velasquez M, Fowler CA, Watts VJ, Roman DL. 2018. Identification of

713 FDA-Approved Small Molecules Capable of Disrupting the Calmodulin–Adenylyl Cyclase 8

34

714 Interaction through Direct Binding to Calmodulin. ACS Chem Neurosci 9:346–357.

715 89. Hamilton WA. 1968. The Mechanism of the Bacteriostatic Action of

716 Tetrachlorosalicylanilide : a Membrane-active Antibacterial Compound. J Gen Microbiol

717 50:441–458.

718 90. Ferreira C, Pereira AM, Pereira MC, Melo LF, Simoes M. 2011. Physiological changes

719 induced by the quaternary ammonium compound benzyldimethyldodecylammonium

720 chloride on Pseudomonas fluorescens. J Antimicrob Chemother 66:1036–1043.

721 91. Ioannou CJ, Hanlon GW, Denyer SP. 2007. Action of Disinfectant Quaternary Ammonium

722 Compounds against Staphylococcus aureus. Antimicrob Agents Chemother 51:296–306.

723 92. Scorzoni L, de Paula e Silva AC, Marcos CM, Assato PA, de Melo WC, de Oliveira HC,

724 Costa-Orlandi CB, Mendes-giannini MJS, Fusco-almeida AM. 2017. Antifungal Therapy :

725 New Advances in the Understanding and Treatment of Mycosis. Front Microbiol 8:36.

726 93. Pippi B, Lopes W, Reginatto P, Silva FÉK, Joaquim AR, Alves RJ, Silveira GP, Vainstein MH,

727 Andrade SF, Fuentefria AM. 2019. New insights into the mechanism of antifungal action

728 of 8-hydroxyquinolines. Saudi Pharm J 27:41–48.

729 94. 2010. Mechanism matters. Nat Med 16:347.

730 95. Khandelwal NK, Wasi M, Nair R, Gupta M, Kumar M, Mondal AK, Gaur NA, Prasad R. 2019.

731 Vacuolar Sequestration of Azoles , a Novel Strategy of Azole Antifungal Resistance

732 Conserved across Pathogenic and Nonpathogenic Yeast. Antimicrob Agents Chemother

733 63:e01347-18.

35

734 96. Luna-Tapia A, Tournu H, Peters TL, Palmer GE. 2016. Endosomal trafficking defects can

735 induce calcium-dependent azole tolerance in Candida albicans. Antimicrob Agents

736 Chemother 60:7170–7177.

737 97. Luna-Tapia A, Kerns ME, Eberle KE, Jursic BS, Palmer GE. 2015. Trafficking through the

738 late endosome significantly impacts Candida albicans tolerance of the azole antifungals.

739 Antimicrob Agents Chemother 59:2410–2420.

740 98. Sass A, Slachmuylders L, Van Acker H, Vandenbussche I, Ostyn L, Bové M, Crabbé A,

741 Chairelli L, Buroni S, Van Nieuwerburgh F, Abatih E, Coenye T. 2019. Various Evolutionary

742 Trajectories Lead to Loss of the Tobramycin-Potentiating Activity of the Quorum-Sensing.

743 Antimicrob Agents Chemother 63:e02092-18.

744 99. Fonzi WA, Irwin MY. 1993. Isogenic Strain Construction and Gene Mapping in Candida

745 albicans. Genetics 134:717–728.

746 100. Kaur R, Ma B, Cormack BP. 2007. A family of glycosylphosphatidylinositol-linked aspartyl

747 proteases is required for virulence of Candida glabrata. Proc Natl Acad Sci U S A

748 104:7628–7633.

749 101. Dewaele K, Frans J, Smismans A, Ho E, Tollens T, Lagrou K. 2018. First case of Candida

750 auris infection in Belgium in a surgical patient from Kuwait. Acta Clin Belg doi:

751 10.1080/17843286.2018.1555114

752 102. Odds FC. 2003. Synergy , antagonism , and what the chequerboard puts between them. J

753 Antimicrob Chemother 52:1.

36

754 103. Walsky RL, Obach RS, Hyland R, Kang P, Zhou S, West M, Geoghegan KF, Helal CJ, Walker

755 GS, Goosen TC, Zientek MA. 2012. Selective Mechanism-Based Inactivation of CYP3A4 by

756 CYP3cide ( PF-04981517 ) and Its Utility as an In Vitro Tool for Delineating the Relative

757 Roles of CYP3A4 versus CYP3A5 in the Metabolism of Drugs. Drug Metab Dispos 40:1686–

758 1697.

759 104. Gao P, Yang Y, Xiao C, Liu Y, Gan M, Guan Y, Hao X, Meng J, Zhou S, Chen X, Cui J. 2012.

760 Identification and validation of a novel lead compound targeting 4-diphosphocytidyl-2-C-

761 methylerythritol synthetase ( IspD ) of mycobacteria. Eur J Pharmacol 694:45–52.

762 105. Jennings MC, Minbiole KPC, Wuest WM. 2015. Quaternary Ammonium Compounds: An

763 Antimicrobial Mainstay and Platform for Innovation to Address Bacterial Resistance. ACS

764 Infect Dis 1:288–303.

765 106. Wu J, Shih H, Vigont V, Hrdlicka L, Diggins L, Singh C, Mahoney M, Chesworth R, Shapiro

766 G, Zimina O, Chen X, Wu Q, Glushankova L, Ahlijanian M, Koenig G, Mozhayeva GN,

767 Kaznacheyeva E, Bezprozvanny I. 2011. Neuronal store-operated calcium entry pathway

768 as a novel therapeutic target for Huntington ’ s disease treatment. Chem Biol 18:777–

769 793.

770 107. Yu X, Vazquez A, Levine AJ, Carpizo DR. 2012. Allele Specific p53 Mutant Reactivation.

771 Cancer Cell 21:614–625.

772 108. Tamaoki T, Nomoto H, Takahashi I, Kato Y, Morimoto M, Tomita F. 1986. Staurosporine, a

773 potent inhibitor of phospholipi/Ca++ dependent protein kinase. Biochem Biophys Res

774 Commun 135:397–402.

37

775

776 TABLE 1 Hits from MCZ potentiator screena

Compound Application Mode of action PF-04981517 In vitro research tool CYP3A4-inhibitor (103) Domiphen Antiseptic Cell wall interactions; cell death (85, 104, bromide 105) QNZ (EVP4593) Possible therapeutic Inhibitor of NF-κB activation (106) agent to treat Huntington’s disease NSC-319726 Anticancer p53-mutant reactivator, metal ion chelator, redox modulator (107) Staurosporine Antibiotic, antifungal, Protein kinase inhibitor (108) antihypertensive, anticancer 777 aCompounds resulting in reduced viability of MCZ-treated C. albicans biofilm cells are presented

778 along with their primary application and mode of action.

779

38

780 TABLE 2 Quantitative formulation of the vaginal placebo creams

Formulation of the vaginal placebo creams Chemical name Commercial name Function Supplier PEG-6, PEG-32 and glycol stearate Tefose 63 O/W Gattefossé emulsifier lauroyl polyoxyl-6 glycerides labrifil M 2130 CS Oil phase Gattefossé cetostearyl alcohol Lanette-D Stifner BASF

Mineral oil Finavestan A 360 Oil-phase Total lubrifiants Water / Aqueous phase 781

782

39

783 TABLE 3 Scoring system for severity of inflammation based on H&E staining

Grade Histopathological image 784 I Minimal inflammatory reaction with few chronic inflammatory 785 cells, especially lymphocytes in the lamina propria II Moderate inflammatory reaction with increasing extent and more 786 organization, with chronic inflammatory cells, eosinophils and

vessels 787 III Severe inflammatory reaction with a dense organized granulomatous reaction with chronic inflammatory cells, 788 eosinophils, fibroblasts and vessels 789

790

40

791 FIG 1 Combining imidazole antifungals and specific quaternary ammonium compounds results

792 in fungicidal activity against C. albicans SC5314 biofilms. Biofilms of C. albicans SC5314 were

793 treated with combinations of imidazoles (either 150 µM miconazole (MCZ), 300 µM ketoconazole

794 (KTC) or 150 µM clotrimazole (CLT)) and a quaternary ammonium compound (QUAT). Maximal

795 concentrations of the quaternary ammonium compound and the imidazole antifungal that did

796 not result in significant killing of biofilm cells upon single compound treatment were selected and

797 used in combination. Following QUATs were used: 50 µM tetraethylammonium bromide (TB), 50

798 µM benzethonium chloride (BTC), 50 µM benzalkonium chloride (BKC), 12.5 µM dequalinium

799 chloride (DQ), or 37,5 µM domiphen bromide (DB). DMSO background was 1%. After 24 h of

800 treatment, the number of CFUs was determined. Mean log CFU values are shown for at least 5

801 biological replicates. Statistical analysis was performed to assign significant differences upon

802 combination treatment as compared to single compound and DMSO only treatment (‘control

803 treatments’). A 2-way ANOVA and either Sidak’s multiple comparisons test (MCZ) or Tukey’s

804 multiple comparisons test (KTC, CLT) was applied and significant differences (p<0.05) relative to

805 the control treatments are shown in orange.

806 FIG 2 Combining MCZ and DB results in fungicidal activity against biofilms of fluconazole-

807 resistant clinical isolates C. albicans Tansir_082 and C. albicans Tansir_121B, and of intrinsically

808 azole-resistant C. glabrata BG2 and C. auris. For each pathogen, maximal concentrations of DB

809 and MCZ that did not result in significant killing of biofilm cells upon single compound treatment

810 were selected and used in combination. Biofilms of fluconazole-resistant C. albicans isolates or C.

811 auris were treated with 150 µM miconazole (MCZ) combined with either 12.5 µM domiphen

812 bromide (DB) or 37.5 µM DB, respectively. Biofilms of C. glabrata BG2 were treated with 500 µM

41

813 MCZ combined with 25 µM DB. DMSO background was 1%. After 24 h (or 48 h for C. glabrata

814 BG2) of treatment, the number of CFUs was determined. Mean log CFU values are shown for at

815 least 4 biological replicates. Statistical analysis was performed to assign significant differences

816 upon combination treatment as compared to single compound or DMSO only treatment (‘control

817 treatments’). A 2-way ANOVA and Tukey’s multiple comparisons test was applied and significant

818 differences (p<0.05) relative to the control treatments are shown in orange.

819 FIG 3 Combining MCZ and DB results in fungicidal action against planktonic stationary phase

820 cultures of C. albicans SC5314 and C. glabrata BG2. For each pathogen, maximal concentrations

821 of DB and MCZ that did not result in significant killing of planktonic cells upon single compound

822 treatment were selected and used in combination. Planktonic stationary cultures of C. albicans

823 SC5314 or C. glabrata BG2 were treated with 25 µM domiphen bromide (DB), combined with 62.5

824 µM or 250 µM miconazole (MCZ), respectively. DMSO background was 1%. After 24 h of

825 treatment, the number of CFUs was determined. Mean log CFU values are shown for 4 biological

826 replicates. Statistical analysis was performed to assign significant differences upon combination

827 treatment as compared to single compound or DMSO only treatment (‘control treatments’). A 2-

828 way ANOVA and Tukey’s multiple comparisons test was applied and significant differences

829 (p<0.01) relative to the control treatments are shown in orange.

830 FIG 4 Resistance against the MCZ-DB combination is not occurring in MCZ-DB evolved C.

831 glabrata BG2 cultures. Sensitivity of three independent C. glabrata BG2 stationary phase cultures

832 (lineages), either evolved for 61 evolution cycles or not (cycle 0, starter culture), to 25 µM

833 domiphen bromide (DB), 250 µM miconazole (MCZ) or the combination was assessed by CFU

834 determination after 24 h incubation. DMSO background was 1%. Mean log CFU values are shown

42

835 for at least 4 biological replicates. Within each evolution cycle, a 2-way ANOVA and Tukey’s

836 multiple comparisons test was performed to assign significant differences upon combination

837 treatment as compared to single compound or DMSO only treatment. Significant differences

838 (p<0.001) are shown in orange. Between evolution cycles, significant differences upon

839 corresponding treatments were assessed by a 2-way ANOVA and Sidak’s multiple comparisons

840 test. ***, p < 0.001; ****, p < 0.0001.

841 FIG 5 Efficacy of treatment with 1% MCZ nitrate or 1% MCZ nitrate with 0.144% DB combination

842 in a C. albicans B2630 vaginitis rat model. First, 2 independent repeats were used to evaluate

843 the effect of DB alone (8 rats in total/group). Subsequently, to evaluate the effect of treatment

844 with MCZ or the combination, 3 independent repeats were used with a total of 24 rats per group

845 for treatments and 12 rats for control. The AUC representing the cumulative infection burden

846 over 14 days is shown. Differences in AUC between 1% MCZ + 0.144% DB and all other treatment

847 groups were tested using a linear mixed model followed by a posthoc analysis with Tukey

848 correction for multiple testing. Significant differences as compared to the control are shown in

849 orange. *, p < 0.05.

850 FIG 6 Histopathology of inflammation and infection stained with H&E (A-C) and PAS-D (D). A:

851 Grade I score showing minimal inflammation with few chronic inflammatory cells; B: Grade II

852 score showing moderate inflammation with chronic inflammatory cells; C: Grade III score showing

853 a severe and dense organized chronic granulomatous reaction; D: Candidial infiltration was

854 observed after PAS-D staining as a magenta colour (black arrows). Depth of inflammatory

855 infiltration in the lamina propria is indicated with a double black arrow (A-C).

43