Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About AAC
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • AAC Podcast
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Antimicrobial Agents and Chemotherapy
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About AAC
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • AAC Podcast
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Experimental Therapeutics

Efficacy of Oral E1210, a New Broad-Spectrum Antifungal with a Novel Mechanism of Action, in Murine Models of Candidiasis, Aspergillosis, and Fusariosis

Katsura Hata, Takaaki Horii, Mamiko Miyazaki, Nao-aki Watanabe, Miyuki Okubo, Jiro Sonoda, Kazutaka Nakamoto, Keigo Tanaka, Syuji Shirotori, Norio Murai, Satoshi Inoue, Masayuki Matsukura, Shinya Abe, Kentaro Yoshimatsu, Makoto Asada
Katsura Hata
1Next Generation Systems CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: k-hata@hhc.eisai.co.jp
Takaaki Horii
1Next Generation Systems CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mamiko Miyazaki
1Next Generation Systems CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nao-aki Watanabe
1Next Generation Systems CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Miyuki Okubo
2Biopharmaceutical Assessments CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jiro Sonoda
2Biopharmaceutical Assessments CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kazutaka Nakamoto
3Oncology PCU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Keigo Tanaka
1Next Generation Systems CFU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Syuji Shirotori
3Oncology PCU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Norio Murai
3Oncology PCU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Satoshi Inoue
3Oncology PCU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Masayuki Matsukura
3Oncology PCU, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shinya Abe
4Pharmaceutical Science & Technology CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tsukuba, Ibaraki 300-2635, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kentaro Yoshimatsu
5Eisai Product Creation Systems, Eisai Co., Ltd., Bunkyo-ku, Tokyo 112-8088, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Makoto Asada
5Eisai Product Creation Systems, Eisai Co., Ltd., Bunkyo-ku, Tokyo 112-8088, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/AAC.00366-11
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

E1210 is a first-in-class, broad-spectrum antifungal with a novel mechanism of action—inhibition of fungal glycosylphosphatidylinositol biosynthesis. In this study, the efficacies of E1210 and reference antifungals were evaluated in murine models of oropharyngeal and disseminated candidiasis, pulmonary aspergillosis, and disseminated fusariosis. Oral E1210 demonstrated dose-dependent efficacy in infections caused by Candida species, Aspergillus spp., and Fusarium solani. In the treatment of oropharyngeal candidiasis, E1210 and fluconazole each caused a significantly greater reduction in the number of oral CFU than the control treatment (P < 0.05). In the disseminated candidiasis model, mice treated with E1210, fluconazole, caspofungin, or liposomal amphotericin B showed significantly higher survival rates than the control mice (P < 0.05). E1210 was also highly effective in treating disseminated candidiasis caused by azole-resistant Candida albicans or Candida tropicalis. A 24-h delay in treatment onset minimally affected the efficacy outcome of E1210 in the treatment of disseminated candidiasis. In the Aspergillus flavus pulmonary aspergillosis model, mice treated with E1210, voriconazole, or caspofungin showed significantly higher survival rates than the control mice (P < 0.05). E1210 was also effective in the treatment of Aspergillus fumigatus pulmonary aspergillosis. In contrast to many antifungals, E1210 was also effective against disseminated fusariosis caused by F. solani. In conclusion, E1210 demonstrated consistent efficacy in murine models of oropharyngeal and disseminated candidiasis, pulmonary aspergillosis, and disseminated fusariosis. These data suggest that further studies to determine E1210's potential for the treatment of disseminated fungal infections are indicated.

INTRODUCTION

The expanding population of immunocompromised patients receiving immunosuppressive or anticancer therapy has resulted in an increased incidence of opportunistic mycoses. Invasive fungal infections have become increasingly common among such immunocompromised and immunosuppressed patients, including solid-organ and hematopoietic stem cell transplant recipients and individuals on immunosuppressive drug regimens (3, 17, 19, 30, 32, 45). In response, the previously established guidelines for the treatment of invasive fungal infections have been recently updated (31, 43). There is still, however, a high rate of morbidity and mortality associated with invasive fungal infections (3, 17, 20, 45), because the currently available antifungal drugs, such as polyenes, azoles, and echinocandins, are limited in terms of their antifungal spectrum, side effects, and mode of action (7). In addition, there has been an increase in resistance to commonly used antifungal compounds, especially azoles, and an epidemiological shift toward more drug-resistant strains (19, 22, 33, 34, 35, 42). Thus, there is a critical need for new antifungal compounds with a novel mechanism of action that have a broad spectrum of activity and fewer side effects, although the development of such new antifungals is just starting to be reported (18, 23, 26).

With this in mind, we have directed our research toward the development of new promising antifungals with a novel mechanism of action. We have discovered a key compound, 1-(4-butylbenzyl) isoquinoline (BIQ) that inhibits the surface expression of glycosylphosphatidylinositol (GPI)-anchored proteins in Saccharomyces cerevisiae, resulting in inhibition of fungal growth, and then identified the GWT1 (GPI-anchored wall protein transfer 1) gene, the target molecule of BIQ, which encodes a new acyltransferase involved in an early step in the GPI biosynthetic pathway of fungi (39, 40). We have performed exploratory syntheses of many different compounds designed to enhance the antifungal activity of BIQ and finally discovered a candidate compound likely to be effective as a new antifungal (27, 38).

E1210, 3-(3-{4-[(pyridin-2-yloxy)methyl]benzyl}isoxazol-5-yl)pyridin-2-amine (Fig. 1), is a first-in-class, new antifungal compound that was discovered by the Tsukuba Research Laboratories of Eisai Co., Ltd. (Ibaraki, Japan). It has potent broad-spectrum antifungal activity with a novel mechanism of action, i.e., inhibition of fungal GPI biosynthesis, and favorable properties as a drug candidate (13, 24, 25, 29, 44). In the present study, the efficacies of oral E1210 and reference antifungal drugs, such as fluconazole (36), voriconazole (2), caspofungin (1), and liposomal amphotericin B (8), were evaluated in murine models of candidiasis, aspergillosis, and fusariosis. In addition, the initial pharmacokinetic and in vivo toxicological profiles of E1210 were also demonstrated.

Fig. 1.
  • Open in new tab
  • Download powerpoint
Fig. 1.

Chemical structure of E1210.

(This work was presented in part at the 50th Interscience Conference on Antimicrobial Agents and Chemotherapy, abstracts F1-842 [12a] and F1-844 [29], Boston, MA, 12 to 15 September 2010.)

MATERIALS AND METHODS

Antifungals.E1210 was synthesized at Eisai Co. Ltd., Tokyo, Japan. The negative logarithm of the dissociation constant (pKa) of E1210 was determined by capillary electrophoresis. The pKa values for the conjugate acid of E1210 were 3 and 5.1 (I [ionic strength] = 0.05). The hydrophobicity (LC18; apparent log P determined by high-performance liquid chromatography [HPLC]) of E1210 at neutral pH was 3.48. Fluconazole and voriconazole were extracted at Eisai Co. Ltd. from commercial products obtained from Pfizer, Inc. (Tokyo, Japan). Caspofungin, amphotericin B, and liposomal amphotericin B were commercially obtained from Merck & Co., Inc. (Whitehouse Station, NJ), Bristol-Myers KK (Tokyo, Japan), and Dainippon Sumitomo Pharma Co., Ltd. (Osaka, Japan), respectively. All drugs were dissolved individually in dimethyl sulfoxide (DMSO) and then diluted with culture medium at required concentrations for in vitro studies. For in vivo efficacy studies in mice, E1210 was dissolved in 250 mmol/liter HCl at a concentration of 25 mg/ml and then diluted with vehicle to the required concentrations. For in vivo toxicological studies in rats, E1210 was dissolved in 400 mmol/liter HCl at a concentration of 100 mg/ml and then diluted with 400 mmol/liter HCl to the required concentrations. Voriconazole was dissolved in 1 mol/liter HCl to a concentration of 20 mg/ml and then diluted with vehicle to the required concentrations. The dosing formulations of E1210 and voriconazole were prepared and stored in a −40°C freezer until use. Other drugs were prepared on the day of use according to the manufacturer's specifications.

Organisms.In total, the following six fungal strains were used for these studies: Candida albicans IFM49971, Candida albicans IFM49738, Candida tropicalis E83037, Aspergillus flavus IFM50915, Aspergillus fumigatus IFM51126, and Fusarium solani IFM50956. These strains were provided by Chiba University (Chiba, Japan) and Gifu University (Gifu, Japan) and were stored as glycerin stock at −80°C.

Animals.Specific-pathogen-free female ICR mice (age, 5 weeks; weight, approximately 25 g; Charles River Japan Inc., Kanagawa, Japan), specific-pathogen-free female DBA/2N mice (age, 8 weeks; weight, approximately 18 g; Charles River Japan Inc., Kanagawa, Japan), or specific-pathogen-free male and female Sprague-Dawley rats (age, 8 weeks; weight, approximately 190 to 270 g; Charles River Japan Inc., Kanagawa, Japan) were used for these experiments. They were housed in cages of 5 to 10 animals per group and had access to food and water ad libitum. All procedures were performed in an animal facility accredited by the Center for Accreditation of Laboratory Animal Care and Use by the Japan Health Sciences Foundation. All protocols were approved by the Institutional Animal Care and Use Committee and carried out according to Eisai animal experimentation regulations.

In vitro susceptibility testing.The MICs of E1210 and the reference compounds were determined using the broth microdilution method detailed by the Clinical and Laboratory Standards Institute (CLSI) in documents M27-A3 (6) and M38-A2 (5). RPMI 1640 medium buffered to pH 7.0 with 0.165 M 3-(N-morpholino)-propanesulfonic acid (MOPS) was used. The results were expressed as the median MIC of each compound derived from three independent experiments.

The Candida spp. were subcultured in Sabouraud dextrose broth (SDB) at 35°C for 1 to 2 days. The Aspergillus spp. were subcultured onto potato dextrose agar (PDA) and then incubated at 35°C for 1 to 2 weeks. F. solani was subcultured onto PDA and incubated at 35°C for 2 to 3 days and then at 25°C for 4 to 5 days. The conidia were scraped from the PDA surface and suspended in sterile normal saline containing 0.05% Tween 80. The cell counts from the yeast cultures or conidial suspensions were determined with a hemocytometer by a modification of the CLSI method, and the cell suspensions were diluted with RPMI 1640 medium buffered to pH 7.0 with 0.165 M MOPS to obtain an inoculum size of 1.5 × 103 cells/ml for Candida spp. or 1.2 × 104 cells/ml for the filamentous fungi. The test organisms were cultured in medium containing E1210 (0.001 to 32 μg/ml), fluconazole (0.001 to 32 μg/ml), voriconazole (0.001 to 32 μg/ml), caspofungin (0.0005 to 16 μg/ml), amphotericin B (0.016 to 8 μg/ml), or 0.5% DMSO, and the growth inhibition induced by the test compounds was evaluated. For all the test compounds except caspofungin, the plates were incubated under the following conditions: at 35°C for 22 to 26 h for C. albicans and C. tropicalis and at 35°C for 46 to 50 h for the filamentous fungi. For caspofungin, the plates were incubated at 35°C for 22 to 26 h for all test strains.

For Candida spp., the reduction in growth was determined based on the changes in optical density of the medium at 660 nm (using a MTP-450 microplate reader; Corona Electric Co., Ltd., Ibaraki, Japan). The MICs of E1210, fluconazole, voriconazole, and caspofungin were defined as the lowest concentrations resulting in a prominent decrease in turbidity (that is, a 50% reduction in growth determined spectrophotometrically) relative to that in a control well by a modification of the CLSI method. The MICs of amphotericin B were defined as the lowest concentration resulting in complete growth inhibition determined visually.

For the filamentous fungi, the growth reductions were graded visually and expressed as a numerical score, ranging from 0 to 4, in accordance with the CLSI document for filamentous fungi (5). The MICs of amphotericin B and voriconazole were defined as the lowest concentration at which a score of 0 was observed, while those of E1210, fluconazole, and caspofungin were defined as the lowest concentration at which a score of 2 was observed. The MICs of caspofungin against the filamentous fungi corresponded to the minimal effective concentrations (MECs) defined in the CLSI guidelines (5).

Oropharyngeal candidiasis model.C. albicans was used to infect mice that were immunosuppressed with cortisone, and the number of C. albicans cells in the oral cavity of each mouse was measured following drug treatment (16, 37). ICR mice were immunosuppressed using 4 mg of subcutaneously administered cortisone acetate given 1 day before and 3 days after infection. The mice were also given 1 mg/ml tetracycline hydrochloride via their drinking water, starting on the day of cortisone administration and continuing throughout the experiment, in order to prevent bacterial infection. C. albicans IFM49971 was grown on Sabouraud dextrose agar (SDA) at 35°C for 2 days. The cells were suspended in sterile normal saline. The cells were counted with a hemocytometer and adjusted to the required density with sterile normal saline. The mice were then anesthetized with chlorpromazine hydrochloride (0.5 mg/mouse given subcutaneously). By use of a micropipette, aliquots (10 μl) of C. albicans IFM49971 suspension were inoculated into the oral cavities of the anesthetized mice. Then the challenge dose of 4 × 105 CFU of C. albicans (CFU)/mouse was given. This was followed with either E1210 orally administered twice daily (BID) or fluconazole orally administered once daily (QD) for three consecutive days starting 3 days after infection. The control group was given the equivalent volume of 5% glucose BID. The mice were anesthetized with chlorpromazine hydrochloride (0.5 mg/mouse subcutaneously) the day after the final dose of the study drug. Efficacy was assessed by determination of the number of C. albicans cells in the oral cavity of each mouse after study drug treatment. The oral cavity (that is, the cheek, tongue, and soft palate) was thoroughly swabbed using a fine-tipped cotton swab. After swabbing, the cotton end was placed into a test tube containing 1 ml sterile normal saline. The cells recovered were suspended in sterile normal saline by mixing them on a vortex mixer before being cultured, after serial 10-fold dilutions, on SDA plates supplemented with ampicillin (0.1 mg/ml). The SDA plates were incubated at 35°C overnight, and then the viable cells were counted as the number of CFU. The cell number was expressed in units of log10 CFU/swab. The lowest detectable number of cells in the oral cavity was 10 CFU (1 log10 CFU). The viable cell counts were performed in duplicate.

Disseminated candidiasis model.ICR mice were immunosuppressed utilizing 5-fluorouracil (5-FU) at 200 mg/kg of body weight subcutaneously administered 6 days prior to infection. These mice were also administered 0.1 mg/ml ciprofloxacin orally via their drinking water, from 2 to 3 days prior to infection to 5 to 7 days after infection, in order to prevent endogenous bacterial infections. C. albicans IFM49971, C. albicans IFM49738, and C. tropicalis E83037 were each cultured on an SDA plate at 35°C for 2 days. The cells from the surface of the agar plate were suspended in sterile normal saline, and the cells were counted with a hemocytometer. The final inoculum was adjusted to the required density using sterile normal saline. Infection was induced in the neutropenic mice by the intravenous administration of 0.2 ml of a C. albicans cell suspension (0.8 to 1.4 × 104 CFU/mouse or 5.3 × 104 CFU/mouse for IFM49971) or of a C. tropicalis cell suspension (3.0 × 105 CFU/mouse) injected into the lateral tail vein. Antifungal therapy was initiated 1 h or 24 h after infection and was continued for three consecutive days (days 0 to 2 or 1 to 3). E1210 or voriconazole was each orally administered two or three times daily, fluconazole was orally administered once daily, and caspofungin or liposomal amphotericin B was intravenously administered once daily. The control group received an equivalent volume of vehicle (5% glucose, 10 ml/kg) orally two or three times daily. In our preliminary studies, the survival curve of control mice receiving vehicle orally was similar to that of mice receiving vehicle intravenously. Therefore, we did not set up the control group to receive vehicle intravenously. The survival rate and survival period were determined over 14 days.

Pulmonary aspergillosis model.DBA/2N mice were immunosuppressed with subcutaneously administered 5-FU at 200 mg/kg, 5 to 6 days prior to infection. The mice were also administered 0.1 mg/ml ciprofloxacin orally via their drinking water, from 3 to 4 days prior to infection until 7 days after infection, in order to prevent endogenous bacterial infections. A. flavus IFM50915 and A. fumigatus IFM51126 were cultured on a PDA plate at 35°C for 7 days. The conidia from the surface of the agar plate were suspended in sterile normal saline containing 0.05% Tween 80, and the cells were counted with a hemocytometer. The final inoculum was adjusted to the required density with sterile normal saline containing 0.05% Tween 80. The mice were anesthetized with 0.1 ml ketamine hydrochloride (4.17 mg/ml) intravenously. Infection was induced in these neutropenic mice by the intranasal inoculation of 0.05 ml of an A. flavus conidial suspension (3.0 × 104 conidia/mouse) or 0.05 ml of an A. fumigatus conidial suspension (6.0 × 104 conidia/mouse). Antifungal therapy was initiated 1 h after infection and was continued for four or seven consecutive days (days 0 to 3 or 0 to 7). E1210 or voriconazole was administered orally twice daily, and caspofungin or liposomal amphotericin B was administered intraperitoneally once daily. The control group received an equivalent volume of vehicle (5% glucose, 10 ml/kg) orally twice daily. In our preliminary studies, the survival curve of control mice receiving vehicle orally was similar to that of mice receiving vehicle intraperitoneally. Therefore, we did not set up the control group to receive vehicle intraperitoneally. The survival rate and survival period were determined over 14 days.

Disseminated fusariosis model.DBA/2N mice were immunosuppressed with 200 mg/kg of subcutaneously administered 5-FU, 6 days prior to infection. The mice were also administered 0.1 mg/ml ciprofloxacin orally in their drinking water, from 3 days prior to infection until 7 days after infection, to prevent bacterial infections. F. solani IFM50956 was cultured on a PDA plate at 30°C for 7 days. The cells from the surface of the agar plate were suspended in sterile normal saline containing 0.05% Tween 80, and the cells were counted using a hemocytometer. The final inoculum was adjusted to the required density using sterile normal saline containing 0.05% Tween 80. Infection was induced in the neutropenic mice by the intravenous inoculation of a 0.2-ml F. solani cell suspension (5.0 × 103 cells/mouse) into the lateral tail vein. Antifungal therapy was initiated 1 h after infection and was continued for five consecutive days (days 0 to 4). E1210 was orally administered three times a day (TID). The control group received an equivalent volume of 5% glucose orally TID. The survival rate and survival period were determined over 14 days.

Pharmacokinetic study.E1210 was intravenously or orally administered to male ICR mice. After administration of E1210, blood samples were drawn from the vena cava of each mouse at designated time points (0.08, 0.25, 0.5, 1, 2, 4, 6, 8 h). Plasma samples were obtained by centrifuging blood. After deproteinization with methanol, the extracted sample was analyzed by liquid chromatography-tandem mass spectrometry (LC/MS/MS). The concentrations of E1210 in plasma were determined by an internal standard method using MassLynx (Waters, Milford, MA). The pharmacokinetic parameters of E1210 were calculated by model independent analysis.

Toxicology study.E1210 was administered orally by gavage once a day for 7 days to male and female Sprague-Dawley rats (3 animals/group/gender) at doses of 100, 300, or 1,000 mg/kg. A control group received an equivalent volume (10 ml/kg) of vehicle (0.4 mol/liter hydrochloric acid). All rats found dead or moribund were necropsied as soon as they were discovered, and all surviving animals were necropsied after 7 days of administration. The following were evaluated: mortality, clinical signs, body weight, food consumption, hematology, blood chemistry, toxicokinetics, hepatic drug-metabolizing enzymes, and macroscopic and microscopic pathologies.

Statistical analysis.In the oropharyngeal candidiasis model, data are expressed as the mean ± standard error of the mean (SEM). The differences in the viable cell counts between the control group and the E1210-treated groups or the fluconazole-treated groups were evaluated using one-way analysis of variance (ANOVA), followed by the Dunnett multiple-comparison test. The dose responsiveness of E1210 or fluconazole was determined using regression analysis.

The differences between the survival curves of the vehicle-treated (control) and antifungal-treated groups over 14 days postinfection were analyzed by the log rank test with the Bonferroni adjustment. Additionally, based on the survival rate at day 14 after infection, the 50% effective dose (ED50) and 95% confidence interval (CI) of each antifungal were estimated with a probit method. For the antifungals for which the 95% CIs were not calculated by a probit method, the 95% CIs of the ED50s were calculated based on the exact 95% confidence limits of the survival rate at each dose. Statistical analyses were performed with SAS version 8.2 software package (SAS Institute Japan Ltd., Tokyo, Japan). A probability (P) value of <0.05 (two-sided) was considered statistically significant.

RESULTS

In vitro antifungal activity.Table 1 shows the MICs of E1210 and reference comparator antifungal compounds against Candida spp., Aspergillus spp., and F. solani used in the in vivo efficacy studies of E1210. E1210 showed potent antifungal activity against C. albicans IFM49971 (MIC = 0.004 μg/ml). E1210 was more active than fluconazole, caspofungin, and amphotericin B and showed activity similar to that of voriconazole against C. albicans IFM49971. E1210 also had potent activity against both azole-resistant Candida strains, C. albicans IFM49738 (MIC = 0.008 μg/ml) and C. tropicalis E83037 (MIC = 0.016 μg/ml); E1210 was more active than all of the reference antifungals tested. E1210 showed potent antifungal activity against A. flavus IFM50915 (MIC = 0.03 μg/ml), A. fumigatus IFM51126 (MIC = 0.03 μg/ml), and F. solani IFM50596 (MIC = 0.06 μg/ml); E1210 was the most active compound tested against these strains of filamentous fungi.

View this table:
  • View inline
  • View popup
Table 1.

Comparative in vitro antifungal susceptibilities of experimental infection strains to E1210 and reference compoundsa

Efficacy in the oropharyngeal candidiasis model.The efficacy of orally administered E1210 compared to that of fluconazole for the treatment of C. albicans-induced oral candidiasis is shown in Fig. 2. In this model, the control group showed a viable oral cavity cell count of 5.62 ± 0.11 log10 CFU. The oral cavity cell counts in mice treated with E1210 (BID) doses of 2.5, 5 and 10 mg/kg were 4.97 ± 0.16, 4.24 ± 0.31, and 3.08 ± 0.27 log10 CFU, respectively. The oral cavity cell counts in mice treated with doses of fluconazole (once daily [QD]) of 2.5, 5, and 10 mg/kg were 5.28 ± 0.18, 3.88 ± 0.23, and 2.75 ± 0.24 log10 CFU, respectively. Mice treated with E1210 or fluconazole at doses of 5 and 10 mg/kg showed significantly better resolution of oral candidiasis than control mice. E1210 or fluconazole reduced the number of viable C. albicans cells in the oral cavity in a dose-dependent manner.

Fig. 2.
  • Open in new tab
  • Download powerpoint
Fig. 2.

Comparative efficacies of E1210 and fluconazole in a murine oropharyngeal candidiasis model. Mice, immunosuppressed with cortisone acetate administered subcutaneously 1 day before and 3 days after infection, were orally infected with 4 × 105 CFU of Candida albicans. E1210 was orally administered twice daily and fluconazole was orally administered once daily for three consecutive days starting 3 days after infection. The oral cavity of each mouse was thoroughly swabbed, and the recovered cells were cultured on SDA plates. After incubation at 35°C overnight, the viable cells were counted and expressed as the number of CFU. The viable cell counts were performed in duplicate. *, P < 0.05 versus control (one-way ANOVA with the Dunnett multiple-comparison test).

Efficacy in the disseminated candidiasis model.The efficacies of E1210, fluconazole, caspofungin, and liposomal amphotericin B on C. albicans-induced mortality in mice are shown in Fig. 3. In this model, all control mice died within 5 days. The mice treated with E1210 (BID) at 5 and 12.5 mg/kg, with fluconazole (QD) at 2 and 5 mg/kg, with caspofungin (QD) at 0.064 and 0.16 mg/kg, and with liposomal amphotericin B (QD) at 0.4 and 1 mg/kg (as amphotericin B) showed significantly higher survival rates than control mice. E1210 at doses of 0.8, 2, 5,and 12.5 mg/kg protected 0%, 11%, 56%, and 89% of the mice at day 14, respectively, and its ED50 was 4.8 mg/kg (95% CI, 3.0 to 7.9 mg/kg). Fluconazole at doses of 0.32, 0.8, 2, and 5 mg/kg protected 0%, 0%, 67%, and 100% of the mice at day 14, respectively, and its ED50 was 1.9 mg/kg (95% CI, 0.80 to 5.0 mg/kg). Caspofungin at doses of 0.01, 0.026, 0.064, and 0.16 mg/kg protected 0%, 11%, 100%, and 100% of the mice at day 14, respectively, and its ED50 was 0.030 mg/kg (95% CI, 0.026 to 0.064 mg/kg). Liposomal amphotericin B at doses of 0.064, 0.16, 0.4, and 1 mg/kg protected 0%, 22%, 56%, and 100% of the mice at day 14, respectively, and its ED50 was 0.31 mg/kg (95% CI, 0.20 to 0.49 mg/kg). The ED50s of all compounds tested in the disseminated candidiasis model are summarized in Table 2.

Fig. 3.
  • Open in new tab
  • Download powerpoint
Fig. 3.

Efficacy of E1210 compared to that of reference antifungals in a murine model of disseminated candidiasis caused by C. albicans IFM49971. Mice (n = 9) were immunosuppressed with 5-FU administered subcutaneously 6 days prior to infection and then intravenously infected with 0.8 × 104 CFU of Candida albicans. E1210 was administered orally twice daily, fluconazole was administered orally once daily, and liposomal amphotericin B or caspofungin was administered intravenously once daily for three consecutive days starting 1 h after infection. The survival rate and survival period were determined over 14 days. (A) E1210; (B) fluconazole; (C) caspofungin; (D) liposomal amphotericin B. *, P < 0.05 versus control group (log rank test with Bonferroni's adjustment).

View this table:
  • View inline
  • View popup
Table 2.

ED50s of E1210 and reference compounds based on day 14 survival ratesa

The efficacies of E1210 and voriconazole on azole-resistant C. albicans-induced mortality in mice are shown in Fig. 4. In this model, all of the control mice died within 5 days. The mice treated with E1210 (TID) at 2.5 mg/kg showed a significantly higher survival rate than the control mice. E1210 at doses of 0.313, 0.625, 1.25, 2.5, and 5 mg/kg protected 0%, 13%, 38%, 88%, and 100% of the mice at day 14, respectively, and its ED50 was 1.3 mg/kg (95% CI, 0.93 to 2.0 mg/kg). Voriconazole (TID) at a dose of 5 mg/kg protected 25% of the mice at day 14.

Fig. 4.
  • Open in new tab
  • Download powerpoint
Fig. 4.

Efficacy of E1210 compared to that of voriconazole in a murine model of disseminated candidiasis caused by azole-resistant C. albicans IFM49738. Mice (n = 8) were immunosuppressed with 5-FU administered subcutaneously 6 days prior to infection and then intravenously infected with 5.3 × 104 CFU of Candida albicans. E1210 or voriconazole was administered orally three times daily for three consecutive days starting 1 h after infection. The survival rate and survival period were determined over 14 days. *, P < 0.05 versus control group (log rank test with Bonferroni's adjustment).

In addition, E1210 was effective in the model of disseminated candidiasis caused by C. tropicalis (data not shown). In this model, all of the control mice died within 4 days. The mice treated with E1210 (TID) at 2.5 mg/kg showed a significantly higher survival rate than the control mice. E1210 at doses of 1.25 and 2.5 mg/kg protected 20% and 100% of the mice at day 14, respectively.

The impact of E1210 administered orally for 3 consecutive days starting 1 h or 24 h after infection on C. albicans-induced mortality in mice is shown in Fig. 5. In this model, all of the control mice died within 5 days. The mice treated with E1210 (TID) at 2.5 mg/kg showed a significantly higher survival rate than the control mice regardless of the time that treatment was initiated. When therapy was started 1 h after infection, E1210 at doses of 0.625, 1.25, and 2.5 mg/kg protected 0%, 50%, and 100% of the mice at day 14, respectively, and its ED50 was 1.3 mg/kg (95% CI, 0.63 to 2.5 mg/kg). When therapy was started 24 h after infection, E1210 at doses of 0.625, 1.25, and 2.5 mg/kg protected 0%, 38%, and 75% of the mice at day 14, respectively, and its ED50 was 1.7 mg/kg (95% CI, 1.1 to 2.8 mg/kg). The efficacy of E1210 for the treatment of disseminated candidiasis was minimally affected by a treatment delay of 24 h.

Fig. 5.
  • Open in new tab
  • Download powerpoint
Fig. 5.

Effect of treatment delay on the efficacy of E1210 in a murine model of disseminated candidiasis. Mice (n = 8) were immunosuppressed with 5-FU administered subcutaneously 6 days prior to infection and then intravenously infected with 1.4 × 104 CFU of C. albicans IFM49971. E1210 was administered orally three times daily for three consecutive days starting 1 h or 24 h after infection. The survival rate and survival period were determined over 14 days.

Efficacy in the pulmonary aspergillosis model.The efficacies of E1210, voriconazole, caspofungin, and liposomal amphotericin B on A. flavus-induced mortality in mice are shown in Fig. 6. In this model, all of the control mice died within 6 days. The mice treated with E1210 (BID) at 10 and 25 mg/kg, voriconazole (BID) at 4 and 10 mg/kg, and caspofungin (QD) at 0.4 and 1 mg/kg showed significantly higher survival rates than the control mice. E1210 at doses of 1.6, 4, 10, and 25 mg/kg protected 0%, 11%, 33%, and 100% of the mice at day 14, respectively, and its ED50 was 10 mg/kg (95% CI, 6.8 to 16 mg/kg). Voriconazole at doses of 0.64, 1.6, 4, and 10 mg/kg protected 0%, 11%, 44%, and 100% of the mice at day 14, respectively, and its ED50 was 3.7 mg/kg (95% CI, 2.5 to 5.9 mg/kg). Caspofungin at doses of 0.064, 0.16, 0.4, and 1 mg/kg protected 11%, 22%, 44%, and 78% of the mice at day 14, respectively, and its ED50 was 0.41 mg/kg (95% CI, 0.21 to 1.3 mg/kg). Liposomal amphotericin B (QD) at a dose of 10 mg/kg did not protect the mice against mortality. The ED50s of all compounds tested in the pulmonary aspergillosis model are summarized in Table 2.

Fig. 6.
  • Open in new tab
  • Download powerpoint
Fig. 6.

Efficacies of E1210 and reference antifungals in a murine model of pulmonary aspergillosis caused by A. flavus IFM50915. Mice (n = 9) were immunosuppressed with 5-FU administered subcutaneously 5 days prior to infection and then intranasally infected with 3.0 × 104 of Aspergillus flavus conidia. E1210 or voriconazole was administered orally twice daily and caspofungin or liposomal amphotericin B was administered intraperitoneally once daily for four consecutive days starting 1 h after infection. The survival rate and survival period were determined over 14 days. (A) E1210; (B) voriconazole; (C) caspofungin; (D) liposomal amphotericin B. *, P < 0.05 versus control group (log rank test with Bonferroni's adjustment).

E1210 was also effective in treating pulmonary aspergillosis caused by A. fumigatus (data not shown). In this model, all of the control mice died within 6 days. The mice treated with E1210 (TID) at 10 and 20 mg/kg showed significantly higher survival rates than the control mice. E1210 at doses of 2.5, 5, 10, and 20 mg/kg protected 0%, 14%, 43%, and 100% of the mice at day 14, respectively, and its ED50 was 9.3 mg/kg (95% CI, 6.4 to 15 mg/kg).

Efficacy in the disseminated fusariosis model.The efficacy of oral E1210 on F. solani-induced mortality in mice is shown in Fig. 7. In this model, 80% of the control mice died within 6 days and 20% of the control mice survived at day 14. E1210 at doses of 5, 10, and 20 mg/kg protected 25%, 63%, and 100% of the mice at day 14, respectively. The mice treated with E1210 (TID) at 20 mg/kg showed significantly higher survival rates than the control mice.

Fig. 7.
  • Open in new tab
  • Download powerpoint
Fig. 7.

Efficacy of E1210 in a murine model of disseminated fusariosis caused by F. solani IFM50956. Mice (n = 8–10) were immunosuppressed with 5-FU administered subcutaneously 6 days prior to infection and then intravenously infected with 5.0 × 103 of F. solani conidia. E1210 was administered orally three times daily for five consecutive days starting 1 h after infection. The survival rate and survival period were determined over 14 days. *, P < 0.05 versus control group (log rank test with Bonferroni's adjustment).

Pharmacokinetic profile.Mean plasma concentrations of E1210 after oral and intravenous administrations to mice are shown in Fig. 8. In mice, after intravenous administration, E1210 exhibited moderate clearance and volume of distribution and the elimination half-life was 2.2 h. E1210 dosed as an oral solution was rapidly absorbed and achieved a maximum concentration at 0.5 h after dosing. Oral bioavailability was calculated at 57.5% in mice.

Fig. 8.
  • Open in new tab
  • Download powerpoint
Fig. 8.

Time-plasma concentration profiles of E1210 after oral and intravenous administrations of a single dose of 1 mg/kg to mice. After administration of E1210, blood samples were drawn from the vena cava of mice at designated time points. After deproteinization with methanol, the extracted sample was analyzed by LC/MS/MS. Each value represents the mean of the results for two animals.

Toxicological profile.At an E1210 dose of 1,000 mg/kg, mortality (1 male) and morbidity (1 male) were observed, which were caused by anorexia and gastrointestinal lesion. Adaptive hepatocellular hypertrophy resulting from liver enzyme induction was observed at 300 mg/kg and higher. No E1210-related change was observed at 100 mg/kg. E1210 was generally well tolerated when given orally to rats for 1 week at doses up to 300 mg/kg.

DISCUSSION

E1210 is a first-in-class, broad-spectrum antifungal with a novel mechanism of action—inhibition of fungal GPI biosynthesis. E1210 has a broad spectrum of potent activity against major pathogenic fungi, such as Candida spp., Aspergillus spp., and other filamentous fungi which are resistant to existing antifungals (24, 25). Invasive fungal infections, including disseminated candidiasis, pulmonary aspergillosis, and disseminated fusariosis, are serious, life-threatening infections recognized increasingly more frequently in immunocompromised patients (17, 19, 28, 30, 45). Oropharyngeal candidiasis, the most commonly encountered opportunistic infection in human immunodeficiency virus-infected patients (10), is becoming increasingly more resistant to fluconazole and, soon, other azoles. We therefore developed experimental models of these fungal infections in immunosuppressed mice to evaluate the therapeutic efficacy of E1210. In our previous studies to evaluate the efficacy of ravuconazole, we showed that 5-fluorouracil treatment elicited a considerable reduction in the number of circulating leukocytes, especially neutrophils, in mice (41), and disseminated and pulmonary infections caused by Candida spp. or Aspergillus spp. were then readily induced in these mice (11, 12). The neutropenic disseminated or pulmonary infection model appears to more closely mimic the immunocompromised patient situation in clinical settings. Furthermore, a disseminated fusariosis model was also able to be established in these neutropenic mice, as part of this study. Oropharyngeal infection caused by C. albicans was able to be induced in cortisone acetate-treated mice.

In order to better evaluate the comparative efficacies of E1210 and reference antifungals, the experimental infection models were studied using various numbers of doses per day for each antifungal compound. It has been reported that the plasma half-lives of voriconazole (2), fluconazole (14), caspofungin (9), and liposomal amphotericin B (8) were 0.7 to 2.9 h, 5.1 h, 7.6 h, and 10.1 to 12.5 h, respectively, in mice. Therefore, the plasma half-life of E1210 (2.2 h) was noted to be similar to that of voriconazole and about two to five times shorter than those of the reference antifungals other than voriconazole. From these data, we determined the optimal frequency of dosing for E1210 and voriconazole to be BID or TID and those for other drugs to be QD for these murine models of infection. Thus, E1210 had a relatively shorter plasma half-life than those of the reference drugs, except for voriconazole, in mice. Also, the addition of serum (90%) greatly affected the MICs of E1210 for the strains tested; the MICs of E1210 increased 64-fold (24, 25). This result indicates that E1210 may have a high plasma protein binding ratio. For all these reasons, we expect that the higher doses of E1210 were needed for effective treatment in mouse models of infection. We are currently conducting further pharmacokinetic and metabolic studies for the future clinical development of E1210 in rats, dogs, and monkeys.

Orally administered E1210 demonstrated clear dose-dependent therapeutic responses in the various experimental infection models in mice. First, E1210 showed efficacy in treating oropharyngeal candidiasis in mice. Treatment with E1210 significantly reduced the number of Candida CFU in the oral cavity in comparison to that of the control treatment (P < 0.05), with the extent of eradication comparable to that of fluconazole. Second, E1210 increased the survival time in a dose-dependent manner in mice infected with Candida spp. or Aspergillus spp. E1210 was consistently effective in treating disseminated candidiasis caused by azole-susceptible C. albicans. Furthermore, E1210 had the additional benefit of an efficacy against azole-resistant C. albicans infections in mice comparable to that against azole-susceptible C. albicans infections. The frequency of clinical reports of azole-resistant C. albicans and azole-resistant Candida spp. other than C. albicans is increasing (22, 33, 35). E1210 proved to be effective against disseminated candidiasis caused by azole-resistant strains of C. albicans and C. tropicalis in mice. E1210 is further characterized by its efficacy against invasive pulmonary aspergillosis caused by A. flavus and A. fumigatus in mice. Invasive aspergillosis currently constitutes the most common cause of infectious pneumonic mortality in patients undergoing hematopoietic stem cell transplantation (HSCT) and is an important cause of opportunistic infection in other immunocompromised patients (3, 4, 17, 19, 30, 32). For primary treatment of invasive pulmonary aspergillosis, intravenous or oral voriconazole is recommended for most patients based on the Infectious Diseases Society of America's guideline published in 2008 (43). Interestingly, E1210 at doses of >20 mg/kg showed a maximum therapeutic efficacy (100% survival) that was comparable to that of voriconazole at 10 mg/kg in murine invasive pulmonary aspergillosis models. In addition, we confirmed that E1210 was effective against disseminated aspergillosis caused by A. fumigatus in mice (data not shown). In the future, it will be very important to evaluate the antifungal activity of E1210 against azole-resistant Aspergillus strains, which are increasingly being recognized in the clinic (34, 42).

From the viewpoint of antifungal spectrum of activity, one of the important characteristics of E1210 is its activity against non-Aspergillus filamentous fungi, such as F. solani and Scedosporium prolificans, which are intrinsically resistant to all currently approved or available antifungals (15, 21, 28). We therefore conclude that it will be very important to evaluate the in vivo efficacy of E1210 in animal models of infections attributable to these pathogens. We showed the efficacy of E1210 in a disseminated fusariosis model in this study and have already started to investigate the efficacy of E1210 in the same model for S. prolificans pulmonary infections.

Furthermore, E1210 proved to be effective in treating disseminated candidiasis, even if treatment was started 24 h after infection (Fig. 5). We also confirmed that E1210 was effective against disseminated aspergillosis caused by A. fumigatus even if treatment was started 24 h after infection in mice (data not shown). These results strongly suggest that E1210 has the potential for being effective if given during acute or subacute infections.

As a preliminary toxicological study, we have conducted a 7-day oral dose range toxicity study in rats. E1210 was generally well tolerated when given orally to rats for 1 week at doses up to 300 mg/kg. Further toxicological studies are needed to confirm the safety of E1210 in rats and other animals, such as dogs and monkeys.

In conclusion, these results suggest that E1210 has the potential for efficacy as a single-agent therapy in the treatment of oropharyngeal candidiasis, disseminated candidiasis, pulmonary aspergillosis, and disseminated fusariosis. E1210 is thus a very promising drug for the treatment of fungal infections, and therefore, further studies on its pharmacokinetics/pharmacodynamics and toxicological characteristics are warranted.

ACKNOWLEDGMENTS

We thank Katsuhiko Kamei, Medical Mycology Research Center, Chiba University, Chiba, Japan, and Hiroshige Mikamo, Department of Infection Control and Prevention, Aichi Medical University, Aichi, Japan, for providing the fungal strains. We thank Takashi Owa, Eisai Product Creation Systems, Eisai Inc., NJ, for making constructive suggestions to the antifungal project team. We thank Frederick P. Duncanson, Eisai Product Creation Systems, Eisai Inc., NJ, for editing and proofreading this article.

FOOTNOTES

    • Received 18 March 2011.
    • Returned for modification 18 May 2011.
    • Accepted 17 July 2011.
    • Accepted manuscript posted online 25 July 2011.
  • Copyright © 2011, American Society for Microbiology. All Rights Reserved.

REFERENCES

  1. 1.↵
    1. Abruzzo G. K.,
    2. et al
    . 1997. Evaluation of the echinocandin antifungal MK-0991 (L-743,872): efficacies in mouse models of disseminated aspergillosis, candidiasis, and cryptococcosis. Antimicrob. Agents Chemother. 41:2333–2338.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    1. Andes D.,
    2. Marchillo K.,
    3. Stamstad T.,
    4. Conklin R.
    . 2003. In vivo pharmacokinetics and pharmacodynamics of a new triazole, voriconazole, in a murine candidiasis model. Antimicrob. Agents Chemother. 47:3165–3169.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. Baddley J. W.,
    2. et al
    . 2010. Factors associated with mortality in transplant patients with invasive aspergillosis. Clin. Infect. Dis. 50:1559–1567.
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    1. Baddley J. W.,
    2. Pappas P. G.,
    3. Smith A. C.,
    4. Moser S. A.
    . 2003. Epidemiology of Aspergillus terreus at a university hospital. J. Clin. Microbiol. 41:5525–5529.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    Clinical and Laboratory Standards Institute. 2008. Reference method for broth dilution antifungal susceptibility testing of filamentous fungi. Approved standard. CLSI document M38-A2. Clinical and Laboratory Standards Institute, Wayne, PA.
  6. 6.↵
    Clinical and Laboratory Standards Institute. 2008. Reference method for broth dilution antifungal susceptibility testing of yeasts. Approved standard. CLSI document M27-A3. Clinical and Laboratory Standards Institute, Wayne, PA.
  7. 7.↵
    1. Denning D. W.,
    2. Hope W. W.
    . 2010. Therapy for fungal diseases: opportunities and priorities. Trends Microbiol. 18:195–204.
    OpenUrlCrossRefPubMedWeb of Science
  8. 8.↵
    1. Gondal J. A.,
    2. Swarts R. P.,
    3. Rahman A.
    . 1989. Therapeutic evaluation of free and liposome-encapsulated amphotericin B in the treatment of systemic candidiasis in mice. Antimicrob. Agents Chemother. 33:1544–1548.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Hajdu R.,
    2. et al
    . 1997. Preliminary animal pharmacokinetics of the parenteral antifungal agent MK-0991 (L-743,872). Antimicrob. Agents Chemother. 41:2339–2344.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Hamza O. J. M.,
    2. et al
    . 2008. Single-dose fluconazole versus standard 2-week therapy for oropharyngeal candidiasis in HIV-infected patients: a randomized, double-blind, double-dummy trial. Clin. Infect. Dis. 47:1270–1276.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Hata K.,
    2. et al
    . 1996. Efficacy of ER-30346, a novel oral triazole antifungal agent, in experimental models of aspergillosis, candidiasis, and cryptococcosis. Antimicrob. Agents Chemother. 40:2243–2247.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Hata K.,
    2. et al
    . 1996. In vitro and in vivo antifungal activities of ER-30346, a novel oral triazole with a broad antifungal spectrum. Antimicrob. Agents Chemother. 40:2237–2242.
    OpenUrlAbstract/FREE Full Text
  13. 12a.↵
    1. Hata K.,
    2. Horii T.,
    3. Miyazaki M.,
    4. Watanabe N.
    . 2010. Efficacy of E1210, a new broad-spectrum antifungal, in murine models of oropharyngeal candidiasis, disseminated candidiasis, and pulmonary aspergillosis, abstr. F1-842. Abstr. 50th Intersci. Conf. Antimicrob. Agents Chemother. American Society for Microbiology, Washington, DC.
  14. 13.↵
    1. Horii T.,
    2. Okubo M.,
    3. Miyazaki M.,
    4. Hata K.,
    5. Watanabe N.
    . 2010. In vivo pharmacodynamic correlates of success for E1210 treatment of disseminated candidiasis. Abstr. 50th Intersci. Conf. Antimicrob. Agents Chemother., abstr. F1-843.
  15. 14.↵
    1. Humphrey M. J.,
    2. Jevons S.,
    3. Tarbit M. H.
    . 1985. Pharmacokinetic evaluation of UK-49,858, a metabolically stable triazole antifungal drug, in animals and humans. Antimicrob. Agents Chemother. 28:648–653.
    OpenUrlAbstract/FREE Full Text
  16. 15.↵
    1. Imhof A.,
    2. Balajee S. A.,
    3. Fredricks D. N.,
    4. Englund J. A.,
    5. Marr K. A.
    . 2004. Breakthrough fungal infections in stem cell transplant recipients receiving voriconazole. Clin. Infect. Dis. 39:743–746.
    OpenUrlCrossRefPubMedWeb of Science
  17. 16.↵
    1. Kamai Y.,
    2. et al
    . 2001. New model of oropharyngeal candidiasis in mice. Antimicrob. Agents Chemother. 45:3195–3197.
    OpenUrlAbstract/FREE Full Text
  18. 17.↵
    1. Kontoyiannis D. P.,
    2. et al
    . 2010. Prospective surveillance for invasive fungal infections in hematopoietic stem cell transplant recipients, 2001–2006: overview of the Transplant Associated Infection Surveillance Network (TRANSNET) Database. Clin. Infect. Dis. 50:1091–1100.
    OpenUrlCrossRefPubMedWeb of Science
  19. 18.↵
    1. Law D.,
    2. Dobb K.,
    3. Fothergill A.,
    4. Payne L. J.,
    5. Birch M.
    . 2009. Abstr. 49th Intersci. Conf. Antimicrob. Agents Chemother., abstr. F1-850.
  20. 19.↵
    1. Marr K. A.,
    2. Carter R. A.,
    3. Crippa F.,
    4. Wald A.,
    5. Corey L.
    . 2002. Epidemiology and outcome of mould infections in hematopoietic stem cell transplant recipients. Clin. Infect. Dis. 34:909–917.
    OpenUrlCrossRefPubMedWeb of Science
  21. 20.↵
    1. Martin G. S.,
    2. Mannino D. M.,
    3. Eaton S.,
    4. Moss M.
    . 2003. The epidemiology of sepsis in the United States from 1979 through 2000. N. Engl. J. Med. 348:1546–1554.
    OpenUrlCrossRefPubMedWeb of Science
  22. 21.↵
    1. Meletiadis J.,
    2. et al
    . 2002. In vitro activities of new and conventional antifungal agents against clinical Scedosporium isolates. Antimicrob. Agents Chemother. 46:62–68.
    OpenUrlAbstract/FREE Full Text
  23. 22.↵
    1. Messer S. A.,
    2. Jones R. N.,
    3. Fritsche T. R.
    . 2006. International Surveillance of Candida spp. and Aspergillus spp.: report from the SENTRY Antimicrobial Surveillance Program (2003). J. Clin. Microbiol. 44:1782–1787.
    OpenUrlAbstract/FREE Full Text
  24. 23.↵
    1. Mitsuyama J.,
    2. et al
    . 2008. In vitro and in vivo antifungal activities of T-2307, a novel arylamidine. Antimicrob. Agents Chemother. 52:1318–1324.
    OpenUrlAbstract/FREE Full Text
  25. 24.↵
    1. Miyazaki M.,
    2. Horii T.,
    3. Hata K.,
    4. Watanabe N.
    . 2010. In vitro antifungal activity of E1210: a novel antifungal with activity against clinically important yeasts and moulds. Abstr. 50th Intersci. Conf. Antimicrob. Agents Chemother., abstr. F1-840.
  26. 25.↵
    1. Miyazaki M.,
    2. et al
    . 2011. In vitro activity of E1210, a novel antifungal, against clinically important yeasts and molds. Antimicrob. Agents Chemother. 55:4652–4658.
    OpenUrlAbstract/FREE Full Text
  27. 26.↵
    1. Motyl M. R.,
    2. et al
    . 2010. Abstr. 50th Intersci. Conf. Antimicrob. Agents Chemother., abstr. F1-847.
  28. 27.↵
    1. Nakamoto K.,
    2. et al
    . 2010. Synthesis and evaluation of novel antifungal agents-quinoline and pyridine amide derivatives. Bioorg. Med. Chem. Lett. 20:4624–4626.
    OpenUrlCrossRefPubMed
  29. 28.↵
    1. Nucci M.,
    2. Anaissie E.
    . 2007. Fusarium infections in immunocompromised patients. Clin. Microbiol. Rev. 20:695–704.
    OpenUrlAbstract/FREE Full Text
  30. 29.↵
    1. Okubo M.,
    2. Toritsuka N.,
    3. Horii T.,
    4. Hata K.,
    5. Sonoda J.
    . 2010. Preclinical pharmacokinetics and toxicology of E1210, a new broad-spectrum antifungal. Abstr. 50th Intersci. Conf. Antimicrob. Agents Chemother., abstr. F1-844.
  31. 30.↵
    1. Pappas P. G.,
    2. et al
    . 2010. Invasive fungal infections among organ transplant recipients: results of the Transplant Associated Infection Surveillance Network (TRANSNET). Clin. Infect. Dis. 50:1101–1111.
    OpenUrlCrossRefPubMedWeb of Science
  32. 31.↵
    1. Pappas P. G.,
    2. et al
    . 2009. Clinical practice guidelines for the management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin. Infect. Dis. 48:503–535.
    OpenUrlCrossRefPubMedWeb of Science
  33. 32.↵
    1. Perfect J. R.,
    2. et al
    . 2001. The impact of culture isolation of Aspergillus species: a hospital-based survey of aspergillosis. Clin. Infect. Dis. 33:1824–1833.
    OpenUrlCrossRefPubMedWeb of Science
  34. 33.↵
    1. Pfaller M. A.,
    2. et al
    . 2010. Results from the ARTEMIS DISK global antifungal surveillance study, 1997 to 2007: a 10.5-year analysis of susceptibilities of Candida species to fluconazole and voriconazole as determined by CLSI standardized disk diffusion. J. Clin. Microbiol. 48:1366–1377.
    OpenUrlAbstract/FREE Full Text
  35. 34.↵
    1. Pfaller M. A.,
    2. et al
    . 2008. In vitro survey of triazole cross-resistance among more than 700 clinical isolates of Aspergillus species. J. Clin. Microbiol. 46:2568–2572.
    OpenUrlAbstract/FREE Full Text
  36. 35.↵
    1. Presterl E.,
    2. Daxböck F.,
    3. Graninger W.,
    4. Willinger B.
    . 2007. Changing pattern of candidaemia 2001-2006 and use of antifungal therapy at the University Hospital of Vienna, Austria. Clin. Microbiol. Infect. 13:1072–1076.
    OpenUrlCrossRefPubMed
  37. 36.↵
    1. Richardson K.,
    2. Brammer K. W.,
    3. Marriott M. S.,
    4. Troke P. F.
    . 1985. Activity of UK-49,858, a bistriazole derivative, against experimental infection with Candida albicans and Trichophyton mentagrophytes. Antimicrob. Agents Chemother. 27:832–835.
    OpenUrlAbstract/FREE Full Text
  38. 37.↵
    1. Takakura N.,
    2. et al
    . 2003. A novel murine model of oral candidiasis with local symptoms characteristic of oral thrush. Microbiol. Immunol. 47:321–326.
    OpenUrlCrossRefPubMedWeb of Science
  39. 38.↵
    1. Tanaka K.,
    2. et al
    . 2010. An effective synthesis of a (pyridine-3-yl)isoxazole via 1,3-dipolar cycloaddition using ZnCl2: synthesis of a (2-aminopyridin-3-yl)isoxazole derivative and its antifungal activity. Chem. Lett. 39:1033–1035.
    OpenUrlCrossRef
  40. 39.↵
    1. Tsukahara K.,
    2. et al
    . 2003. Medicinal genetics approach towards identifying the molecular target of a novel inhibitor of fungal cell wall assembly. Mol. Microbiol. 48:1029–1042.
    OpenUrlCrossRefPubMedWeb of Science
  41. 40.↵
    1. Umemura M.,
    2. et al
    . 2003. GWT1 gene is required for inositol acylation of glycosylphosphatidylinositol anchors in yeast. J. Biol. Chem. 278:23639–23647.
    OpenUrlAbstract/FREE Full Text
  42. 41.↵
    1. Van Zant G.
    1984. Studies of hematopoietic stem cells spared by 5-fluorouracil. J. Exp. Med. 159:679–690.
    OpenUrlAbstract/FREE Full Text
  43. 42.↵
    1. Verweij P. E.,
    2. Mellado E.,
    3. Melchers W. J. G.
    . 2007. Multiple-triazole-resistant aspergillosis. N. Engl. J. Med. 356:1481–1483.
    OpenUrlCrossRefPubMedWeb of Science
  44. 43.↵
    1. Walsh T. J.,
    2. et al
    . 2008. Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America. Clin. Infect. Dis. 46:327–360.
    OpenUrlCrossRefPubMedWeb of Science
  45. 44.↵
    1. Watanabe N.,
    2. Horii T.,
    3. Miyazaki M.,
    4. Hata K.
    . 2010. E1210, a new broad-spectrum antifungal, inhibits glycosylphosphatidylinositol (GPI) biosynthesis and affects Candida albicans cell characteristics. Abstr. 50th Intersci. Conf. Antimicrob. Agents Chemother., abstr. F1-841.
  46. 45.↵
    1. Wisplinghoff H.,
    2. et al
    . 2004. Nosocomial bloodstream infections in US hospitals: analysis of 24,179 cases from a prospective nationwide surveillance study. Clin. Infect. Dis. 39:309–317.
    OpenUrlCrossRefPubMedWeb of Science
PreviousNext
Back to top
Download PDF
Citation Tools
Efficacy of Oral E1210, a New Broad-Spectrum Antifungal with a Novel Mechanism of Action, in Murine Models of Candidiasis, Aspergillosis, and Fusariosis
Katsura Hata, Takaaki Horii, Mamiko Miyazaki, Nao-aki Watanabe, Miyuki Okubo, Jiro Sonoda, Kazutaka Nakamoto, Keigo Tanaka, Syuji Shirotori, Norio Murai, Satoshi Inoue, Masayuki Matsukura, Shinya Abe, Kentaro Yoshimatsu, Makoto Asada
Antimicrobial Agents and Chemotherapy Sep 2011, 55 (10) 4543-4551; DOI: 10.1128/AAC.00366-11

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Antimicrobial Agents and Chemotherapy article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Efficacy of Oral E1210, a New Broad-Spectrum Antifungal with a Novel Mechanism of Action, in Murine Models of Candidiasis, Aspergillosis, and Fusariosis
(Your Name) has forwarded a page to you from Antimicrobial Agents and Chemotherapy
(Your Name) thought you would be interested in this article in Antimicrobial Agents and Chemotherapy.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Efficacy of Oral E1210, a New Broad-Spectrum Antifungal with a Novel Mechanism of Action, in Murine Models of Candidiasis, Aspergillosis, and Fusariosis
Katsura Hata, Takaaki Horii, Mamiko Miyazaki, Nao-aki Watanabe, Miyuki Okubo, Jiro Sonoda, Kazutaka Nakamoto, Keigo Tanaka, Syuji Shirotori, Norio Murai, Satoshi Inoue, Masayuki Matsukura, Shinya Abe, Kentaro Yoshimatsu, Makoto Asada
Antimicrobial Agents and Chemotherapy Sep 2011, 55 (10) 4543-4551; DOI: 10.1128/AAC.00366-11
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

About

  • About AAC
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • AAC Podcast
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #AACJournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0066-4804; Online ISSN: 1098-6596