U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Probe Reports from the NIH Molecular Libraries Program [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2010-.

Cover of Probe Reports from the NIH Molecular Libraries Program

Probe Reports from the NIH Molecular Libraries Program [Internet].

Show details

Probing Metabolic Requirements for Fungal Virulence — Probe 1

, , , , , , , , , , , , , and .

Author Information and Affiliations

Received: ; Last Update: May 14, 2014.

The role of glycolytic versus mitochondrial respiratory metabolism in supporting fungal virulence and drug-resistance have not been fully investigated due in large part to the lack of appropriate small molecule tool compounds that can reliably discriminate between these two metabolic pathways. This project seeks to identify drug-like molecules that selectively inhibit the growth of fungi when they are cultured under conditions requiring mitochondrial respiration to support metabolic needs. A prior Molecular Libraries Probe Centers Network (MLPCN) project designed to identify compounds capable of chemosensitizing drug-resistant C. albicans clinical isolates to fluconazole also uncovered compounds possessing potent single-agent activity against a range of opportunistic human fungal pathogens. Further experimentation determined their activity is dependent upon culture conditions that require mitochondrial respiratory metabolism for growth and survival. The current project investigated this subset of compounds as selective inhibitors of fungal respiration, and we now report the development of a thiohydantoin probe (ML316, CID 56604860) exhibiting potent antifungal activity. ML316 will be of great value in probing the metabolic requirements for fungal virulence and may provide essential leads for the development of new antifungal drugs that operate in a completely unexploited target space.

Assigned Assay Grant No: 1 R03 MH086456-01

Screening Center Name & PI: Broad institute Probe Development Center, Stuart L. Schreiber, PhD

Chemistry Center Name & PI: Broad institute Probe Development Center, Stuart L. Schreiber, PhD

Assay Submitter & Institution: Susan Lindquist, PhD, Whitehead Institute, Cambridge, MA

PubChem Summary Bioassay Identifier (AID): 588528

Probe Structure & Characteristics

ML316.

ML316

CID/ML No.TargetsIC50 (μM)
[SID, AID]
Anti-TargetIC50 (μM)
[SID, AID]
Fold Selective*
CID 56604860
ML316
CaCi-2 growth inhibition0.04
[SID 134356650, AID 623899]
Fibroblast toxicity>26
[SID 134356650, AID 602394]
>650x
CaCi-17 growth inhibition1.0
[SID 134356650, AID 623897]
>37x
*

Selectivity = Anti-target IC50/Target IC50

CID/ML No.Secondary AssaysIC50 (μM)
[SID, AID]
Fold Selective
CID 56604860
ML316
Inhibition of S. cerevisiae growth on glycerol0.004
[134356650, 623969]
>750x
Inhibition of S. cerevisiae growth on glucose>3.0
[134356650, 623971]
Inhibition of C. glabrata growth on glycerol0.011
[134356650, 623965]
>272x
Inhibition of C. glabrata growth on glucose>3.0
[134356650, 623967]

Selectivity = Glucose IC50/Glycerol IC50

Recommendations for the scientific use of this probe

Intrinsic and acquired drug resistance by medically relevant microorganisms poses a grave threat to human health and has enormous economic consequences. Fungal pathogens present a particular challenge because they are eukaryotes and share many of the same mechanisms that support the growth and survival of the human host cells they infect. Epidemiological studies indicate that nearly 50% of patients that contract invasive fungal infections die within one year, totaling 10,000 deaths per year in the United States alone (1). The high rate of mortality stems in part from the paucity of antifungal agents available to clinicians. Only three classes of antifungal agents are active against systemic infection. In addition, current antifungals are not well-suited for combination therapy, and can even display antagonistic effects in certain settings (2). Agents with single-agent activity against pathogenic fungi and no toxicity to human cells are exceedingly rare. Additional classes of single-agent antifungals would allow for the rational design of combination therapies to limit the emergence of resistance.

A frequently overlooked issue in the search for new antifungal targets is that fungal pathogens face a diverse array of environmental challenges during the establishment of invasive infection within a host animal (3). These can include pH, thermal and osmotic stresses, as well as the need to utilize alternative carbon sources for energy production (4, 5). The role of glycolytic versus respiratory metabolism in supporting fungal virulence and drug-resistance remains unresolved, due in large part to the absence of good tools with which to tackle the question (6, 7). We have developed a small molecule probe ML316 that can reliably discriminate between glycolytic versus respiratory metabolism in supporting fungal virulence and growth.

The probe ML316 can be used in numerous types of research activities. For example, it can be evaluated in genetic studies using genome-wide over-expression and deletion libraries to define the modes of action in Saccharomyces cerevisiae assayed under fermentative versus respiratory growth conditions. To complement genetic approaches, affinity precipitation and proteomic approaches based on Stable Isotope Labeling with Amino acids in Cell culture (SILAC) technology may also be used for target identification. Further medicinal chemistry research of ML316, including optimization of DMPK properties will enable studies in well-established mouse models for infection by drug-resistant Candida glabrata and Candida albicans.

1. Introduction

Scientific Rationale

The various metabolic pathways of medically relevant fungi have recently come under investigation for their roles in fungal pathogenicity and the acquisition of drug resistance (8, 9). Specifically, the mitochondrion and its numerous components and functions are emerging as factors in determining the effectiveness of current antimycotic therapies in controlling human fungal infections. In vitro experiments have shown that loss of mitochondrial DNA (mtDNA) in C. glabrata correlates to increased resistance to azole antifungals (6). Paradoxically, clinical isolates of azole-resistant C. glabrata rarely show impairment of mtDNA function. The significance of the seemingly contradictory in vitro assays remains to be understood (10, 11).

The importance of mitochondrial function to fungal virulence is no better understood. C. glabrata with dysfunctional or missing mtDNA, also known as petite strains, are documented as being less pathogenic than non-petite strains (6). However, this observation is only reproducible when the genetic damage is artificially induced with chemical reagents such as ethidium bromide. The work of Ferrari et al. showed the C. glabrata petite mutant clinical isolate BPY41 was actually more virulent than its non-petite parent BPY40 in murine infection models (12). The 15-day survival rate of mice infected with BPY41 was 25% compared to 65% when infected with BPY40. Similarly, murine fungal loads of BPY41 after 7-day infection were 10- to 100-fold greater on average. Interestingly, when BPY40 was treated with ethidium bromide to generate a petite mutant, this newly generated petite strain was noticeably less pathogenic than BPY41, with a 15-day survival rate of 85% but similar fungal loads to its parent strain BPY40 (13) These conflicting observations clearly underscore the need to better understand the role mitochondria play in pathogenic fungi.

Similar work with Candida albicans has proven even more challenging because C. albicans petite mutants are more difficult to obtain (13, 14). In one exceptional case, Cheng et al. were able to produce a viable petite mutant by passing C. albicans SC5314 through murine spleens by intravenous inoculation (15, 16). After five serial passages, a mutant strain named P5 was isolated and characterized. P5 exhibited several phenotypes characteristic of respiratory mutants, including the inability to proliferate on glucose-deficient media. Subsequent oxygen consumption measurements suggested that while the electron transport chain was intact and functional, it was no longer coupled to ATP synthesis (16). With regards to clinical relevance, P5 was determined to be 10-fold less susceptible to fluconazole and voriconazole, more resistant to phagocytosis and neutrophils, and less sensitive to superoxide generators. In addition, P5 sustained a completely non-lethal infection in mice for up to 60 days. In contrast, SC5314 infection resulted in 100% mortality within 9 days (15).

The reported work with C. glabrata and C. albicans petite mutants suggests that modifying fungal mtDNA in laboratory settings with chemical agents cannot be relied upon to investigate the clinical behavior of these invasive fungi. Similarly, clinically isolated Candida petite mutants may not be amenable to further experimentation. The isolation of petite mutants from patients rarely occurs, and for the few strains harvested thus far, the occurrence of nuclear DNA mutations cannot be ruled out. Identifying specific mitochondrial contributions to Candida clinical behavior is thus complicated because the selective pressures of the host environment cannot be reasonably and reliably controlled to allow modification of only fungal mtDNA.

An alternative, untested approach would be to modulate fungal mitochondria function during an active infection. However, the current toolbox of mitochondria modulators (Figure 1) targets the oxidative phosphorylation pathway and does not discriminate between fungal or mammalian mitochondria (1720). Disruption of this process is typically lethal for both the invading fungi and its mammalian host. A possible fungal-selective mitochondria inhibitor is the antibacterial agent ilicicolin H (Figure 1, compound 1) (21). Studies by Trumpower et al. have shown that ilicicolin H binds to purified S. cerevisiae cytochrome bc1 complex with ~100-fold selectivity over isolated bovine bc1 complex (IC50 = 0.003 μM and 0.200 μM, respectively) (22, 23). However, 1 displays considerable toxicity against HeLa cells (ED50 = 2 μg/mL or 4.6 μM) (21). Follow-up studies with ilicicolin H have been slow to emerge, possibly because there are no longer any commercial sources of this natural product.

Figure 1. Some known inhibitors of fungal mitochondria oxidative phosphorylation.

Figure 1

Some known inhibitors of fungal mitochondria oxidative phosphorylation. Ilicicolin H (1) has demonstrated 100-fold selectivity for fungal mitochondria in biochemical assays, but displays moderate toxicity against HeLa cells. Antimycin (2) and funiculosin (more...)

Our project seeks to identify compounds capable of modulating cellular respiration in fungi, preferably by affecting mitochondrial function, without triggering a similar response in mammalian cells. It has been reported that fungal mitochondria possess several proteins lacking human orthologs (8, 24), and we have adopted a phenotypic assay tree in the hope of identifying modulators of these unique targets. Probes successfully developed within this project will be of great value for interrogating the metabolic requirements of fungal virulence. In addition, discovered probes may provide essential leads for the development of new antifungal drugs that operate in a completely unexploited target space.

2. Materials and Methods

See subsections for a description of the materials and methods used for each assay.

Materials and Reagents

  • Alamar Blue was purchased from Invitrogen (Catalog No. DAL1100).
  • JC-1 dye was obtained from Invitrogen (Catalog No. T-3168).
  • Amphotericin B was purchased from Sigma-Aldrich (Catalog No. A2411).
  • Geldanamycin was obtained from AG Scientific (Catalog No. G-1047).
  • Ilicicolin H was previously purchased from Analyticon Discovery (Catalog No. NP-005728) and was purified by column chromatography prior to use.
  • Carbonyl cyanide m-chlorophenyl hydrazone (CCCP) was purchased from Sigma-Aldrich (Catalog No. C2759).

Cell Lines

The following cell lines were used in this study:

  • Candida albicans CaCi-2; a drug-resistant clinical isolate was provided by the Whitehead Institute (25). This strain was used for the primary assay.
  • Candida albicans CaCi-17; a strongly drug-resistant clinical isolate was provided by the Whitehead Institute (25). This strain was used for the primary assay
  • Saccharyomyces cerevisiae W303-1a (ATCC 208352); a well-characterized Saccharomyces strain used for determination of glycolytic vs. respiratory metabolism, as well as in the mitochondrial disruption assay.
  • Candida glabrata (ATCC 2001); used for determination of glycolytic vs. respiratory metabolism.
  • NIH-3T3 mammalian fibroblasts (ATCC; CRL no.1658); used for mammalian toxicity assays.

2.1. Assays

A summary listing completed assays and corresponding PubChem AID numbers is provided in Appendix A. Refer to Appendix B for the detailed assay protocols.

2.1.1. Primary CaCi-2 (AID 588529) and CaCi-17 (AID 588530), CaCi-2 Dose-Response Retest (AID 602190, AID 623899, AID 623981, AID 624006) and CaCi-17 Dose-Response Retest (AID 602189, AID 623897, AID 623982)

The primary assay measures reduction of viability (as measured by reduced fluorescence in the presence of Alamar Blue) by potential inhibitors. The assay was performed using two Candida albicans clinical isolates to determine the range of the antifungal activity. In this assay, a fungal inoculum is dispensed into 384-well assay plates in which 100 nL of compounds have been pinned. After 48 hours of incubation at 30°C, Alamar Blue is added and fluorescence output is read. Compounds possessing an IC50 value less than 10 μM were selected for further studies.

2.1.2. Counterscreen Mammalian Cell Toxicity Assay (AID 588634, AID 602394)

Quiescent murine 3T3 fibroblasts were subjected to incubation with compounds of interest for 48 hours. Subsequent cell viability was determined via measuring Alamar Blue fluorescence. Toxicity at less than 10 times the effective CaCi-2 IC50 or less than 20 μM was an indication of a poor therapeutic index and resulted in exclusion of the associated compound from further consideration.

2.1.3. Secondary S. cerevisiae Inhibition in Glucose Media (AID 623971, AID 624025), S. cerevisiae Inhibition in Glycerol Media (AID 623969, AID 624012), C. glabrata Inhibition in Glucose Media (AID 623967, AID 624013), and C. glabrata Inhibition in Glycerol Media (AID 623965, AID 624014)

Saccharomyces, while not a major human pathogen, is a useful model with well-developed genetic tools available. Candida glabrata is an increasingly prominent clinical problem for immune-compromised patients but with fewer genetic tools available with which to study it. These organisms can utilize alternative metabolic pathways to support growth and survival depending on the carbon source. By comparing the concentration-dependent inhibition of yeast growth in glycerol relative to glucose media, a quantitative assessment of a compound’s ability to disrupt respiration can be determined. Compounds in this assay must have an IC50 of less than 10 μM in both organisms when grown in glycerol media. There should be at least a 10-fold loss in potency against both organisms when they are grown in glucose media, thus indicating respiratory disruption.

2.1.4. Secondary Mitochondrial Disruption Assay (AID 624062, AID 624068)

The fluorescent dye JC-1 was used to quantify the ability of compounds to disrupt the mitochondrial membrane potential of fungi. Mitochondrial membrane depolarization is indicated by a decrease in the yellow/green fluorescence intensity ratio. This is a non-gating, binning assay that establishes whether inhibition of fungal growth in glycerol results from direct compromise of mitochondrial function or alternative components of fungal respiratory metabolism. Probes that disrupt mitochondrial function as well as those that perturb other metabolic components are desirable.

2.2. Probe Chemical Characterization

After preparation as described in Section 2.3, the probe (ML316) was analyzed by UPLC, 1H, 19F and 13C NMR spectroscopy, and high-resolution mass spectrometry. The data obtained from NMR and mass spectroscopy are consistent with the structure of the probe, and UPLC indicates an isolated purity of greater than 95%. The complete synthetic protocol is provided in Appendix C, and the associated spectral data are provided in Appendix E.

The solubility of the probe (ML316) was experimentally determined to be 1.1 μM in phosphate buffered saline with 1% (v/v) DMSO. The probe is reasonably stable in PBS solution (>60% remaining after a 48-hour incubation). The data from the PBS stability assay is provided in Figure 2. The probe is unstable in human and murine plasma with less than 1% remaining after incubation at 37°C for 5 hours. The ethyl carbamate appears to be the primary source of instability since replacing the carbamate with a methyl group (CID 56604835) significantly increases plasma stability (Table 1). However, the stable analog CID 56604835 shows no activity against the C. albicans test strains (see Section 3.4 for full details).

Figure 2. Stability of the Probe (ML316) and CID 56604835 in PBS Buffer (pH 7.4, 23°C).

Figure 2

Stability of the Probe (ML316) and CID 56604835 in PBS Buffer (pH 7.4, 23°C).

Table 1. Plasma Stability of Probe ML316 and analogs CID3889161 and CID56604835.

Table 1

Plasma Stability of Probe ML316 and analogs CID3889161 and CID56604835.

Experimental procedures for all analytical assays are provided in Appendix D. The physical properties of the probe ML316 are summarized in Table 2.

Table 2. Summary of Probe Properties Computed from Structure ML316.

Table 2

Summary of Probe Properties Computed from Structure ML316.

2.3. Probe Preparation

Probe 1 (ML316) and most of the analogs prepared for structure-activity relationship analysis were prepared by the four-step sequence outlined in Scheme 1. Cyclohexanone was subjected to a Strecker reaction with 4-fluoroaniline and trimethylsilyl cyanide. The resulting adduct 4 was cyclized to the hydantoin 5 with sodium cyanate, then converted to the thiohydanatoin 6, and acylated with ethyl chloroformate to provide the probe 7. Full experimental details are provided in Appendix C.

Scheme 1. Synthesis of the Probe (ML316).

Scheme 1

Synthesis of the Probe (ML316).

3. Results

Probe attributes

  • Inhibits growth in primary screen cell line C. albicans CaCi-2 at an IC50 ≤ 10 μM.
  • Inhibits growth in resistant cell line C. albicans CaCi-17 at an IC50 ≤ 10 μM.
  • Inhibits growth in S. cerevisiae grown on glycerol at an IC50 ≤ 10 μM.
  • Inhibits growth in C. glabrata grown on glycerol at an IC50 ≤ 10 μM.
  • Shows at least 10-fold selectivity between the CaCi-2 test strain and murine 3T3 cells.
  • Shows at least 10-fold selectivity between S. cerevisiae grown on glycerol and S. cerevisiae grown on glucose.
  • Show at least 10-fold selectivity between C. glabrata grown on glycerol and C. glabrata grown on glucose.

The current project emerges from an earlier MLPCN effort to identify compounds capable of reversing antifungal drug-resistance, thereby providing probes to interrogate the mechanisms responsible for drug-resistance in medically relevant fungi (PubChem summary AID 2007). Compounds that directly inhibit fungal growth were outside the scope of the original project, and a secondary assay integrated into that project measured a compound’s intrinsic antifungal activity (AID 2387), rather than its ability to reverse resistance to the widely used antimycotic fluconazole. This secondary assay identified a number of compounds that possessed potent single agent activity against a range of opportunistic human fungal pathogens, but only under culture conditions that require mitochondrial respiratory metabolism for growth and survival.

We mined our data reported in PubChem for growth inhibition of Candida albicans as well as mammalian fibroblasts (AID 2387 and 2327, respectively). Analysis of this data identified 43 compounds that inhibited C. albicans growth with IC50 ≤ 10 μM. Approximately 75% (30 of 43) of these single-agent antimycotics showed modest cytotoxicity against fibroblasts (IC50 ≥ 20 μM). Based on this data mining, seventeen dry powders were obtained and evaluated for C. albicans and fibroblast growth inhibition. After examining the re-test data, a spirocyclic thiohydantoin (CID3889161) was prioritized for further development.

The initial hit (CID 3889161, Figure 3) is a potent inhibitor of C. albicans growth (IC50 = 0.3 μM), with insignificant cytotoxicity against murine 3T3 fibroblasts (IC50 ≥ 26 μM). Analogs of CID3889161 were designed and prepared accordingly, resulting in the identification of the more potent probe ML316 (see Section 3.4).

Figure 3. Dose Response Curves for Initial Hit (CID 3889161).

Figure 3

Dose Response Curves for Initial Hit (CID 3889161). CID 3889161 was used over a range of concentrations up to 26 μM in the primary assay and secondary assays. Dose curves were generated with Genedata Condeseo and show normalized percent activity (more...)

3.1. Summary of Screening Results

As described previously, an earlier high-throughput screen evaluated 302,509 compounds for their ability to chemosensitize the fluconazole-resistant Candida albicans strain CaCi-2 to a sublethal concentration of fluconazole (PubChem AID 1979). A secondary screen of the primary assay hits was included to identify any intrinsic antimycotic activity, and 342 compounds were evaluated in this assay. 43 compounds displayed inhibitory activity against CaCi-2 with IC50 values below 10 μM (AID 2387). While their antifungal activity disqualified these compounds from the prior project, this set of 43 compounds was re-purposed for the current campaign to identify potent single-agent antimycotics.

DMSO stocks of only 39 compounds were available for re-test against the C. albicans strain CaCi-2 (PubChem AID 588529) and the more highly drug-resistant CaCi-17 (PubChem AID 588530). Of the 39 compounds, 23 surpassed the less than 10 μM IC50 screening cutoff for both assays. Unfortunately, the cytotoxicity of many compounds appeared to be non-specific as determined by a murine 3T3 fibroblast counter screen (AID 588634). Of the 23 antifungal compounds, only two compounds (CID 26662188 and CID 3889161) displayed greater than 10-fold selectivity towards fungal cells. One compound (CID 26662188) was deemed chemically intractable and subsequently discarded.

In order to identify additional candidates, the PubChem data for AID 2387 was revisited. The CaCi-2 inhibitory activity threshold was raised from 10 μM to 15 μM to produce a list of 67 compounds. This hit list was cross-referenced against the 39 compounds previously evaluated as described above and also checked for fibroblast toxicity using available PubChem data (AID 2327). After applying these filters, an additional 17 compounds were obtained.

Dry powders for these 17 compounds, as well as for CID 3889161, were procured for re-testing. In addition, several analogs of promising compounds were obtained to provide preliminary SAR data and validate these scaffolds. After purity analysis and structural confirmation by NMR spectroscopy, a total 24 substances was screened according to the workflow outlined in Figure 4. The screens in this pathway consist of CaCi-2 and CaCi-17 inhibitory activity, mammalian toxicity, and growth media-dependent activity against C. glabrata and S. cerevisiae. The associated PubChem AIDs for these assays are provided in Figure 4. CID 3889161 emerged as the most promising candidate and was subsequently prioritized for further investigation. A number of analogs related to this scaffold were prepared and assayed for fungal selective respiratory inhibition. The results of the SAR investigation are presented in detail in Section 3.4.

Figure 4. Critical Path for Probe Development.

Figure 4

Critical Path for Probe Development.

3.2. Dose Response Curves for Probe

Figure 5. ML316 Selectively Inhibits Growth of C. albicans (A and B) over 3T3 Fibroblasts (C).

Figure 5ML316 Selectively Inhibits Growth of C. albicans (A and B) over 3T3 Fibroblasts (C)

C. albicans CaCi-2 growth inhibition (AID 623899), IC50 = 0.04 μM (A); C. albicans CaCi-17 growth inhibition (AID 623897), IC50 = 1.0 μM (B); Murine 3T3 fibroblasts growth inhibition (AID 602394), IC50 > 26 μM (C); Dose curves were generated with Genedata Condeseo and show normalized percent activity for the individual doses, performed in duplicate.

Figure 6. ML316 Inhibits Growth of S. cerevisiae and C. glabrata When Fed Glycerol but Not Glucose.

Figure 6ML316 Inhibits Growth of S. cerevisiae and C. glabrata When Fed Glycerol but Not Glucose

S. cerevisiae on glycerol growth inhibition (AID 623969), IC50 = 0.004 μM (A); S. cerevisiae on glucose growth inhibition (AID 623971), IC50 > 3.0 μM (B); C. glabrata on glycerol growth inhibition (AID 623965), IC50 = 0.011 μM (C); C. glabrata on glucose growth inhibition (AID 623967), IC50 > 3.0 μM (D). Dose curves were generated with Genedata Condeseo and show normalized percent activity for the individual doses, performed in duplicate.

3.3. Scaffold/Moiety Chemical Liabilities

A potential chemical liability associated with the probe is the possible hydrolysis of the ethyl carbamate. As discussed in Section 2.2, slow hydrolysis in PBS was observed with noticeable loss of the carbamate only occurring after 24 hours. This instability was addressed with the preparation of N-alkyl derivatives and larger, more stable carbamates. However, these analogs displayed diminished activity against the C. albicans test strains, and the complete SAR study results are presented in Section 3.4.

A search of PubChem for the probe (ML316) indicated that the probe has not been previously evaluated in any other assay. A structure-based search in SciFinder and Reaxys did not identify any publications or patents in which the probe appeared.

3.4. SAR Tables

In order to investigate the activity of the hit (CID 3889161), a collection of 51 structurally related analogs were synthesized and evaluated for their ability to inhibit growth in the C. albicans test strains. The biological assay data of these analogs are presented in Tables 38 (PubChem AID 623899, AID 623897, AID 602394). Characterization data (1H NMR spectra and UPLC chromatograms) of these analogs are provided in Appendix G.

Table 3. SAR Analysis of Ethyl Carbamate.

Table 3

SAR Analysis of Ethyl Carbamate.

Table 8. Growth media-dependent activity of C. albicans active analogs.

Table 8

Growth media-dependent activity of C. albicans active analogs.

The hit compound (CID 3889161, Table 3, Entry 1) displayed significant activity against the C. albicans CaCi-2 strain (IC50 = 0.3 μM), but was considerably less effective at inhibiting the growth of the more resistant CaCi-17 strain (IC50 = 8.5 μM). There was also no noticeable toxicity to fibroblasts (IC50 > 26 μM). The weak activity towards CaCi-17 was the primary attribute targeted for improvement during subsequent SAR studies.

The importance of the ethyl carbamate was examined first, and the results are presented in Table 3. Alternative alkyl carbamates displayed moderate activity against CaCi-2 (Table 3, entries 2–5). Potency against this strain correlated with the size of the carbamate, and smaller alkyl chains were more effective. Except for the methyl carbamate derivative which only showed marginal activity, these analogs had no effect on CaCi-17 growth. Phenyl carbamate (Table 3, entry 6) showed no activity against either C. albicans strain. Replacing the nitrogen cap with other functionalities such as methyl sulfonamide, butyl amide, and N-alkyl chains did not confer additional antifungal activity (Table 3, entries 7–10). Similarly, the unsubstituted nitrogen failed to inhibit C. albicans growth (Table 3, entry 11).

While the substituent of the imide nitrogen seemed resistant to modification, the neighboring N-tolyl group was considerably more pliant (Table 4). Substitution at the phenyl ring’s 4-position was not required for activity, and removing the methyl group appeared to improve potency against the more resistant CaCi-17 line (Table 4, entry 2). Conversely, incorporating methyl ether at that position attenuated the compound’s effect on both C. albicans strains (Table 4, entry 3). Progressing to electron-deficient phenyl rings provided the most significant gains in potency (Table 4, entries 4–7). While the trifluoromethyl was the worst performer among the electron-withdrawing substituents examined, this analog was still only 2-fold weaker than the original hit. In comparison, the 4-fluoro, chloro, and cyano replacements were all 4 to 8-fold more effective against both CaCi-2 and CaCi-17. Comparison of positional effects showed that the location of the substituent has only a marginal influence on the compound’s potency (Table 4, entries 8–9), although minor gains in PBS solubility were observed. The 4-ethyl and 4-propyl derivatives retain potency against CaCi-2 (Table 4, entries 10–11), but potency diminishes rapidly with larger substituents at this position (Table 4, entries 12–14). However, significantly smaller substituents are not effective growth inhibitors either (Table 4, entries 15–16). The N-benzyl analog showed a 10-fold drop in activity (Table 4, entry 17). The activity trends of this subset suggest the N-tolyl cap may lie within a sizeable pocket. While excessively large substituents attenuate activity, smaller functionalities (e.g. isopropyl or N-H) seem unable to occupy this space effectively and therefore lower cellular activity.

Table 4. SAR Analysis of Eastern Arene Ring.

Table 4

SAR Analysis of Eastern Arene Ring.

Modification of the spirocyclic cyclohexane led to drastic reductions in activity (Table 5). Incorporating oxygen into the ring system provided a large increase in solubility, but this was accompanied by a 100-fold drop in potency (Table 5, entry 2). Substituting smaller aliphatic rings reiterated this trend; gains in solubility were offset by the inability to affect C. albicans growth (Table 5, entries 3–5).

Table 5. SAR Analysis of Northern Cyclohexane Ring.

Table 5

SAR Analysis of Northern Cyclohexane Ring.

Table 6 summarizes attempts to alter the underlying thiohydantoin core. Exchanging any of the chalcogens for other elements (O → N or S → O) eliminated cellular activity (Table 6, entries 2–3). Similarly, switching the nitrogen substituents produced an inactive derivative (Table 6, entry 4). N-alkyl hydantoins showed no ability to inhibit C. albicans growth (Table 6, entries 5–6) and neither did N-alkylated 4-thiohydantoins (Table 6, entries 7–8). Many intermediates prepared were also evaluated for antifungal activity, but none of these compounds were able to affect C. albicans (Table 7). The results summarized in Table 7 clearly demonstrate the importance of the carbamate (cf. Table 7, entries 7–8 & 10).

Table 6. Modifications of the Thiohydantoin Core.

Table 6

Modifications of the Thiohydantoin Core.

Table 7. Bioactivity of Various Intermediates.

Table 7

Bioactivity of Various Intermediates.

Six of the most potent inhibitors of C. albicans, including hit CID 3889161, were then evaluated against C. glabrata and S. cerevisiae under different growth conditions to investigate their ability to perturb cellular respiration pathways in fungi (Table 8). All of the compounds tested were able to deter growth in both fungal strains at nanomolar concentrations (IC50’s 0.006 – 0.058 μM) when the yeast were cultured in non-fermentable media. Conversely, yeast grown on a glucose-derived media showed considerable resistance to the test compounds (IC50 > 3 μM). Amphotericin B significantly inhibited C. glabrata and S. cerevisiae growth regardless of the growth media (Table 8, entry 7).

Of the analogs evaluated, the 4-fluoro derivative (CID 56604860) demonstrated the most activity against the C. albicans test strains CaCi-2 (IC50 = 0.04 μM) and CaCi-17 (IC50 = 1.0 μM) (Table 4, entry 4). In addition, CID 56604860 appears to disrupt cellular respiration processes of the fermenting yeast C. glabrata and S. cerevisiae. CID 56604860 is a potent growth inhibitor of these species when only non-fermentable carbon sources are available (IC50 = 0.011 and 0.004 μM, respectively), but its antifungal properties can be mitigated by permitting the yeast to ferment glucose (IC50 > 3.0 μM) (Table 8, entry 3). Because of its impressive performance in these cellular assays, CID 56604860 was nominated as the probe (ML316) for the respiration-selective growth inhibition of pathogenic fungi.

3.5. Cellular Activity

Both primary assays were performed with whole cells. One secondary screen evaluating toxicity utilized murine 3T3 fibroblast cells, and growth inhibition assays of whole S. cerevisiae and C. glabrata on different media were included as additional secondary assays. An overview of the assays is provided in Section 2.1, and full experimental details can be found in Appendix B. The probe (ML316) clearly met the established cellular activity criteria specified for this project (refer to Section 4, Table 9).

Table 9. Comparison of Probe ML316 to Project Criteria.

Table 9

Comparison of Probe ML316 to Project Criteria.

3.6. Profiling Assays

The probe was evaluated for binding to a broad panel of receptors, ion channels and enzymes. ML316 exhibits a remarkable selectivity profile against the 67 targets screened. When tested at 10 μM, the dopamine transporter showed approximately 80% inhibition with an estimated IC50 of 1.7 μM. The remaining 66 targets showed less than 50% inhibition when treated with 10 μM ML316 (Figure 7).

Figure 7. Selectivity profiling of ML316.

Figure 7

Selectivity profiling of ML316.

4. Discussion

Publicly available data from the NIH’s PubChem repository facilitated an abbreviated screening campaign for the current project. 302,509 compounds from the NIH’s screening collection had been previously evaluated for inhibitory activity against C. albicans CaCi-2 (AID 1979, AID 2387) as well as 3T3 mammalian fibroblasts (AID 2327), and cross-referencing this data identified 67 compounds demonstrating acceptable potency against CaCi-2 that were also reasonably non-toxic towards fibroblasts. After validation of these candidates from dry powder, thiohydantoin CID3889161 was selected as a priority scaffold for further investigation.

Over 50 derivatives of CID 3889161 were synthetically prepared and evaluated for antifungal activity against two Candida albicans clinical isolates, CaCi-2 and CaCi-17. Both strains have demonstrated resistance towards the commonly prescribed antimycotic fluconazole, with CaCi-17 being significantly more resistant than CaCi-2. In addition to fungal growth inhibition, these analogs were screened for non-specific toxicity using murine 3T3 fibroblasts. Figure 8 summarizes the various modifications explored during SAR studies. Three primary diversity points (highlighted in purple, blue, and green) were investigated, and the number of analogs prepared for each location is reported in Figure 8. Several analogs do not fit into these three clusters and are classified as “miscellaneous” analogs.

Figure 8. Summary of Analogs Prepared to Investigate the SAR profile of hit CID 3889161.

Figure 8

Summary of Analogs Prepared to Investigate the SAR profile of hit CID 3889161. Key SAR findings for each site of diversification are provided in italics.

From the SAR study, it was determined that carbamates were the preferred substituent for the imide nitrogen and that the ethyl carbamate was optimal. Electron-deficient phenyl rings are superior to the original N-tolyl group, while ortho and meta substituents do not appear to significantly influence cellular activity. Contracting the cyclohexyl ring system provides increases in solubility while diminishing antifungal potency. While some variation of the nitrogen substituents is tolerated, the modification of the underlying thiohydantoin motif is not. The 4-fluorophenyl analog (CID 56604860) emerged from these studies as the most potent antifungal of this class. Subsequent investigation of CID 56604860 revealed that its antifungal properties could be modulated by the specific growth media given to the fungal test strains, as desired by the project criteria. Consequently, CID 56604860 was nominated as the probe ML316.

Based on the project goals summarized in Table 9, the probe ML316 clearly surpasses all of the specified criteria. ML316 possesses potent activity against the C. albicans test strains CaCi-2 (IC50 = 0.04 μM) and CaCi-17 (IC50 = 1.0 μM). In addition, ML316 is a potent growth inhibitor of C. glabrata and S. cerevisiae when only non-fermentable carbon sources (e.g. glycerol) are available (IC50 = 0.011 and 0.004 μM, respectively), but its antifungal properties are lost when the yeasts ferment glucose (IC50 > 3.0 μM). There is no apparent toxicity against mammalian 3T3 fibroblasts at concentrations up to 26 μM, indicating that ML316 may be a selective inhibitor of fungal mitochondrial respiration.

According to PubChem, ML316 has not been tested in any other assay reported to its database. The original hit, CID 3889161, has been evaluated in 7 assays. All seven assays are associated with the original screening campaign (AID 2007) from which the current project is derived. The cellular activity of CID 3889161 reported in Section 3.4 is in agreement with the published PubChem data.

4.1. Comparison to existing art and how the new probe is an improvement

A search of the available literature identified several compounds capable of disrupting fungal respiration (1720). Details of this prior art search are provided in Appendix F. With the exception of ilicicolin H, these compounds do not show appreciable selectivity towards fungi over mammalian targets or cells. Hence, we decided to compare the probe ML316 with ilicicolin H.

Previous work with the antifungal antibacterial ilicicolin H has shown that this natural product is a potent inhibitor of the mitochondrial cytochrome bc1 complex (22, 23). Using purified proteins, Trumpower et al. determined that ilicicolin H binds to the S. cerevisiae enzyme complex with almost 100-fold greater selectivity than the corresponding bovine homolog. To our knowledge, this selectivity has not been reproduced within a cell-based assay.

A commercial sample of ilicicolin H was procured from Analyticon Discovery and determined to be approximately 85% pure by HPLC. The commercial sample was purified by column chromatography over silica gel prior to testing in cellular assays. We recently attempted to obtain additional quantities of ilicicolin H but discovered that this compound is no longer commercially available.

Probe ML316 was tested alongside ilicicolin H in a direct comparison of biological activity. The results are summarized in Table 10. ML316 clearly shows a stronger inhibitory effect upon Candida albicans growth than ilicicolin H. While both compounds show differential activity relative to growth media, the distinction is far less pronounced with ilicicolin H regardless of the fungal test strain.

Table 10. Comparison of Probe ML316 to Prior Art.

Table 10

Comparison of Probe ML316 to Prior Art.

Given the complex structure of ilicicolin H, optimization of biological and physical properties through synthetic methods will be difficult. Despite having been isolated 40 years ago (21), the first and only total synthesis of the ilicicolin H was reported in 1985 (26). That this pioneering synthesis produced racemic material further underscores the inherent difficulties of preparing analogs of this compound. While semi-synthetic analogs are possible (27, 28), the lack of a commercial feedstock renders this option untenable at the present time.

As a small molecule probe, ML316 is the superior compound because of its cellular potency and selectivity, and its simple preparation and chemical tractability facilitate further optimization.

4.2. Mechanism of Action Studies

A series of secondary assays were integrated into the current project to interrogate the possible mode of action. ML316 appears to disrupt cellular respiration of fungal organisms as demonstrated by the distinctly different growth patterns of C. glabrata and S. cerevisiae when they are fed with fermentable and non-fermentable carbon sources. In both species, ML316 is most effective when the yeast are grown in glycerol-containing media and displays a greater than 250-fold reduction in potency when the yeast are grown in glucose-containing media.

Additionally, direct disruption of mitochondrial membrane potential in C. glabrata and S. cerevisiae was assessed using a fluorescence probe JC-1. JC-1 is a lipophilic, cationic dye that can selectively enter mitochondria and reversibly changes from red to green when homeostatic mitochondrial membrane potential is lost. Application of JC-1 dye to living yeast treated with 1 μM ML316 determined that depolarization of the mitochondrial membrane does not occur (Figures 9 and 10) (PubChem AIDs 624062, 624068). Comparison of the DMSO controls (Figures 9 and 10, A) with ML316 (Figures 9 and 10, B) suggests the yeast can still maintain their normal mitochondrial membrane potential; it appears that the molecular target of ML316 may be integrated within an alternative metabolic pathway that fungi utilize for respiration (10).

Figure 9. Evaluating Loss of S. cerevisiae Mitochondrial Membrane Potential with JC-1.

Figure 9

Evaluating Loss of S. cerevisiae Mitochondrial Membrane Potential with JC-1. Δψm depolarization was monitored by FACS analysis with JC-1 mitochondrial staining. DMSO negative control in S. cerevisiae (A); S. cerevisiae treated with 1.0 (more...)

Figure 10. Evaluating Loss of C. glabrata Mitochondrial Membrane Potential with JC-1.

Figure 10

Evaluating Loss of C. glabrata Mitochondrial Membrane Potential with JC-1. Δψm depolarization was monitored by FACS analysis with JC-1 mitochondrial staining. DMSO negative control in C. glabrata (A); C. glabrata treated with 1.0 μM (more...)

4.3. Planned Future Studies

Target identification of ML316 will be the main priority. Current evidence suggests ML316 disrupts a critical respiratory pathway independent of mitochondrial membrane potential. ML316 will now be evaluated in genetic studies using genome-wide over-expression and deletion libraries to define its mode(s) of action in S. cerevisiae cultured under fermentative versus respiratory growth conditions. To complement genetic approaches, affinity precipitation and proteomic approaches based on Stable Isotope Labeling with Amino acids in Cell culture (SILAC) technology may also be used for target identification.

The probe will be submitted to the Center for Medical Mycology, an affiliate of the University Hospitals of Cleveland and Case Western Reserve University, to evaluate its antifungal activity against a panel of pathogenic fungi.

Further chemical optimization of ML316 will focus on optimization of aqueous solubility and plasma stability. The N-tolyl substituent appears to be the most tractable position, and the introduction of heteroarenes like pyridines at this location may afford the desirable solubility. Analogously, embedding the ethyl carbamate into an oxazole or similar ring systems might address the observed plasma instability. By addressing the solubility and plasma stability issues, we seek to prepare the probe for application in well-established mouse models of human disease in order to determine whether mitochondrial function is required for infection by drug-resistant C. glabrata and C. albicans.

5. References

1.
Pfaller MA, Diekema DJ. Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev. 2007 Jan;20(1):133–63. [PMC free article: PMC1797637] [PubMed: 17223626]
2.
Lewis RE, Lund BC, Klepser ME, Ernst EJ, Pfaller MA. Assessment of antifungal activities of fluconazole and amphotericin B administered alone and in combination against Candida albicans by using a dynamic in vitro mycotic infection model. Antimicrob Agents Chemother. 1998 Jun;42(6):1382–6. [PMC free article: PMC105608] [PubMed: 9624480]
3.
Cowen LE. Hsp90 orchestrates stress response signaling governing fungal drug resistance. PLoS Pathog. 2009 Aug;5(8):e1000471. Epub 2009 Aug 28. [PMC free article: PMC2726949] [PubMed: 19714223]
4.
Becker JM, Kauffman SJ, Hauser M, Huang L, Lin M, Sillaots S, Jiang B, Xu D, Roemer T. Pathway analysis of Candida albicans survival and virulence determinants in a murine infection model. Proc Natl Acad Sci U S A. 2010 Dec 21;107(51):22044–9. Epub 2010 Dec 6. [PMC free article: PMC3009777] [PubMed: 21135205]
5.
Robbins N, Collins C, Morhayim J, Cowen LE. Metabolic control of antifungal drug resistance. Fungal Genet Biol. 2010 Feb;47(2):81–93. Epub 2009 Jul 10. [PubMed: 19595784]
6.
Brun S, Dalle F, Saulnier P, Renier G, Bonnin A, Chabasse D, Bouchara JP. Biological consequences of petite mutations in Candida glabrata. J Antimicrob Chemother. 2005;56:307–314. [PubMed: 15958498]
7.
Cheng S, Clancy CJ, Zhang Z, Hao B, Wang W, Iczkowski KA, Pfaller MA, Nguyen MH. Uncoupling of oxidative phosphorylation enables Candida albicans to resist killing by phagocytes and persist in tissue. Cell Microbiol. 2007;9:492–501. [PubMed: 16987332]
8.
Shingu-Vazquez M, Traven A. Mitochondria and fungal pathogenesis: drug tolerance, virulence, and potential for antifungal therapy. Eukaryot Cell. 2011 Nov;10(11):1376–83. Epub 2011 Sep 16. [PMC free article: PMC3209048] [PubMed: 21926328]
9.
de Martins VP, Dinamarco TM, Curti C, Uyemura SA. Classical and alternative components of the mitochondrial respiratory chain in pathogenic fungi as potential therapeutic targets. J Bioenerg Biomembr. 2011 Feb;43(1):81–8. Review. [PubMed: 21271279]
10.
Bouchara JP, Zouhair R, Le Boudouil S, Renier G, Filmon R, Chabasse D, Hallet JN, Defontaine A. In-vivo selection of an azole-resistant petite mutant of Candida glabrata. J Med Microbiol. 2000 Nov;49(11):977–84. [PubMed: 11073151]
11.
Ferrari S, Ischer F, Calabrese D, Posteraro B, Sanguinetti M, Fadda G, Rohde B, Bauser C, Bader O, Sanglard D. Gain of function mutations in CgPDR1 of Candida glabrata not only mediate antifungal resistance but also enhance virulence. PLoS Pathog. 2009 Jan;5(1):e1000268. Epub 2009 Jan 16. [PMC free article: PMC2607542] [PubMed: 19148266]
12.
Ferrari S, Sanguinetti M, De Bernardis F, Torelli R, Posteraro B, Vandeputte P, Sanglard D. Loss of mitochondrial functions associated with azole resistance in Candida glabrata results in enhanced virulence in mice. Antimicrob Agents Chemother. 2011 May;55(5):1852–60. Epub 2011 Feb 14. [PMC free article: PMC3088236] [PubMed: 21321146]
13.
Abuhatab MA, Whittaker PA. Respiration deficiency in the pathogenic yeast, Candida albicans. Biochem Soc Trans. 1992 Feb;20(1):63S. [PubMed: 1633984]
14.
Aoki S, Ito-Kuwa S. Induction of petite mutation with acriflavine and elevated temperature in Candida albicans. J Med Vet Mycol. 1987 Aug;25(4):269–77. [PubMed: 3312559]
15.
Cheng S, Clancy CJ, Zhang Z, Hao B, Wang W, Iczkowski KA, Pfaller MA, Nguyen MH. Uncoupling of oxidative phosphorylation enables Candida albicans to resist killing by phagocytes and persist in tissue. Cell Microbiol. 2007 Feb;9(2):492–501. Epub 2006 Sep 20. [PubMed: 16987332]
16.
Cheng S, Clancy CJ, Nguyen KT, Clapp W, Nguyen MH. A Candida albicans petite mutant strain with uncoupled oxidative phosphorylation overexpresses MDR1 and has diminished susceptibility to fluconazole and voriconazole. Antimicrob Agents Chemother. 2007 May;51(5):1855–8. Epub 2007 Feb 26. [PMC free article: PMC1855545] [PubMed: 17325226]
17.
von Jagow G, Link TA. Use of specific inhibitors on the mitochondrial bc1 complex. Methods Enzymol. 1986;126:253–71. [PubMed: 2856132]
18.
Ueki M, Machida K, Taniguchi M. Antifungal inhibitors of mitochondrial respiration: discovery and prospects for development. Curr Opin Anti-Infective Invest Drugs. 2000;2(4):387–398.
19.
Sridhara AM, Reddy KRV, Keshavayya J, Vadiraj SG, Bose P, Ambika DS, Raju CK, Shashidhara S, Raju NH. Synthesis, antimicrobial and cytotoxicity studies of some novel phthalazine-methoxyacrylate derivatives. J Pharm Res. 2011;4(2):496–500.
20.
Mathre DE. Mode of action of oxathiin systemic fungicides. III. Effect on mitochondrial activities. Pest Biochem Physiol. 1971;1(2):216–224.
21.
Hayakawa S, Minato H, Katagiri K. The ilicicolins, antibiotics from Cylindrocladium ilicicola. J Antibiot (Tokyo). 1971 Sep;24(9):653–4. [PubMed: 5167226]
22.
Gutierrez-Cirlos EB, Merbitz-Zahradnik T, Trumpower BL. Inhibition of the yeast cytochrome bc1 complex by ilicicolin H, a novel inhibitor that acts at the Qnsite of the bc1 complex. J Biol Chem. 2004 Mar 5;279(10):8708–14. Epub 2003 Dec 10. [PubMed: 14670947]
23.
Rotsaert FA, Ding MG, Trumpower BL. Differential efficacy of inhibition of mitochondrial and bacterial cytochrome bc1 complexes by center N inhibitors antimycin, ilicicolin H and funiculosin. Biochim Biophys Acta. 2008 Feb;1777(2):211–9. Epub 2007 Nov 1. [PMC free article: PMC2677292] [PubMed: 18022381]
24.
Okamoto K, Shaw JM. Mitochondrial morphology and dynamics in yeast and multicellular eukaryotes. Annu Rev Genet. 2005;39:503–36. Review. [PubMed: 16285870]
25.
Redding S, Smith J, Farinacci G, Rinaldi M, Fothergill A, Rhine-Chalberg J, Pfaller M. Resistance of Candida albicans to fluconazole during treatment of oropharyngeal candidiasis in a patient with AIDS: documentation by in vitro susceptibility testing and DNA subtype analysis. Clin Infect Dis. 1994 Feb;18(2):240–2. [PubMed: 8161633]
26.
Williams DR, Bremmer ML, Brown DL, D’Antuono J. Total synthesis of (±)-ilicicolin H. J Org Chem. 1985 July;50(15):2807–9.
27.
Liu W, Guan Z, Singh SB. A new method for the synthesis of 1,4,5-oxadiazocines and its application in the structure modification of natural products. Tetrahedron Lett. 2005 Nov 14;46(46):8009–12.
28.
Singh SB, Li X, Chen T. Biotransformation of antifungal ilicicolin H. Tetrahedron Lett. 2011 Nov 16;52(46):6190–1.

Appendix A. Assay Summary Table

Table A1Summary of Completed Assays and AIDs

PubChem AIDTypeTargetConcentration Range (μM)Samples Tested
588528SummaryNANANA
558529PrimaryCaCi-2 growth inhibition26 – 0.0139
588530PrimaryCaCi-17 growth inhibition26 – 0.0139
602190Confirmatory (Powder)CaCi-2 growth inhibition26 – 0.124
602189Confirmatory (Powder)CaCi-17 growth inhibition26 – 0.124
623899AnalogsCaCi-2 growth inhibition26 – 0.0155
623897AnalogsCaCi-17 growth inhibition26 – 0.0155
623981
624006
Analogs (low dose)CaCi-2 growth inhibition3 – 0.0000067
623982Analogs (low dose)CaCi-17 growth inhibition3 – 0.0000067
588634SecondaryFibroblast Toxicity26 – 0.0139
602394Secondary (Analogs)Fibroblast Toxicity26 – 0.0155
623967
624013
Secondary (Analogs)C glabrata on glucose3 – 0.0000067
623965
624014
Secondary (Analogs)C glabrata on glycerol3 – 0.0000067
623971
624025
Secondary (Analogs)S cerevisiae on glucose3 – 0.0000067
623969
624012
Secondary (Analogs)S cerevisiae on glycerol3 – 0.0000067
624062Secondary (Analogs)C. glabrata mitochondria inhibition1.07
624068Secondary (Analogs)S. cerevisiae mitochondria inhibition1.07

Appendix B. Detailed Assay Protocol

CaCi-2 Dose-Response Primary (AID 558529, AID 602190, AID 623899, AID 623981, AID 624006)
CaCi-17 Dose-Response Primary (AID 588530, AID 602189, AID 623897, AID 623982)

Materials and Reagents

Clear, flat bottom, black, 384-well plates (Corning, Catalog No. 3712BC Lot No. 35808016); Amphotericin (Sigma, Catalog No. A2411) 15 mM stock solution in DMSO; Pen/Strep (Gibco, Catalog No.10378-016; Lot No. 21040170) 100X in PBS; Alamar Blue (AG Scientific, Catalog no.DAL 1100; Lot No.151016SA); PBS without Calcium and Magnesium (Cellgro, Catalog No. 21-040-CV)

Synthetic Defined Growth Medium

RPMI 1640 medium, (powder without sodium bicarbonate; Invitrogen, Catalog No. 31800-089; Lot No. 648072); Uridine 8 mg/mL in water (Sigma, Catalog No. U3750, Lot No. 028KO760); Glucose 40% (w/v) in water (Sigma, Catalog No. G-5400); MOPS Buffer (Sigma, Catalog No. M-1254; Lot No. 098K0033)

  1. Prepare 1X RPMI medium by dissolving 10.4 grams powdered medium in 800 mL water.
  2. Add 34.52 g MOPS. While stirring, adjust pH to 7.0 with 10 N NaOH.
  3. Add 10 mL uridine solution, 50 mL glucose solution, adjust final volume to 1000 mL. Filter sterilize.
Fungal Inoculum

Test Strains: C. albicans CaCi-2, C. albicans CaCi-17

  1. Inoculate 500 μL of strain from cryopreserved stock into 250 mL shaker flask containing 30 mL growth medium. Shake at 30°C overnight.
  2. Read OD 600 of 1 mL fungal culture in a cuvette using a standard optical density reader (Eppendorf BioPhotometer Plus), with growth medium as a background blank.
  3. Dilute to desired volume of fungal inoculums according to the following formula: (1/OD measured) X (Desired Final Volume of Inoculum) X 0.3 = Volume of fungal culture (μL) to add to desired volume of growth medium. When added to media in wells, this yields a calculated starting OD of the fungal inoculum of 0.00015.

Procedures

  1. Add Pen/Strep at 0.1 mL per 10 mL media (1% v/v).
  2. Use a Thermo Combi nL to dispense 20 μL/well of assay media into all wells.
  3. Pin 100 nL test compound from compound plates into assay plates using CyBi-Well pin tool.
  4. Dispense 20 μL/well of culture into the assay media in all wells.
  5. Incubate plates in a humidified (90% humidity) Liconic incubator at 37°C without agitation for 48 hours.
  6. Dilute Alamar Blue Reagent 1:40 in Ca/Mg-free PBS.
  7. To all plates, add 5 μL/well of the diluted Alamar to a final dilution factor of 1:200.
  8. Incubate the plates for an additional 2 hours.
  9. Read the Relative Fluorescence Intensity (RFU) of wells on a standard plate reader as a measure of relative fungal growth. Envision (Perkin Elmer) plate reader set-up: Ex 544 nm, Em 590 nm, Bandwidth 12 nm, Top read.

Counter screen Mammalian Cell Toxicity Dose Response (AID 588634, AID 602394)

Materials and Reagents

Clear, flat bottom, black 384-well plates (Corning, Catalog No. 3712BC; Lot No. 35808016); Geldanamycin (AG Scientific, Catalog No. G-1047) 15 mM stock solution in DMSO; Alamar Blue (AG Scientific, Catalog No. DAL1100, Lot No. 151016SA); PBS without Calcium and Magnesium (Cellgro, Catalog No. 21-040-CV)

Assay Medium

Optimem medium (Invitrogen, Catalog No. 31985-070; Lot No. 548536); 2.5% (v/v) Fetal Bovine Serum (Hyclone, Catalog No.30071.03; Lot No. ARF26748); 1% (v/v) Pen/Strep solution (Invitrogen, Catalog No.15140-122; Lot No. 529891)

Cell Inoculum

Test Strain: NIH-3T3 mammalian fibroblasts (ATCC CRL No. 1658)

  1. Plate cells in 384-well plates at 1,000 cells/well in 40 μL assay medium.
  2. Incubate plates overnight at 37°C under 5% CO2.

Procedures

  1. After overnight culture, pin compounds into wells at 100 nL/well using the CyBio CyBi-Well pinning instrument.
  2. Return the plates to the tissue culture incubator and incubate the culture for an additional 48 hours at 37°C under 5% CO2.
  3. At the completion of this incubation, add Alamar Blue solution diluted 1:40 in PBS to each well (10 μL/well) to achieve a final dilution of 1:200.
  4. Incubate the plates for an additional 2–3 hours at 37°C under 5% CO2.
  5. Read the Relative Fluorescence Intensity (RFU) of wells on a standard plate reader as a measure of relative cell growth. Envision (Perkin Elmer) plate reader set-up: Ex 544 nm, Em 590 m, Bandwidth12 nm, Top read.

Secondary Glycerol Assay Protocol (AID 623969, AID 623965, AID 624014, AID 624012)

Materials and Reagents

Clear, flat bottom, black 384-well plates (Corning Catalog no. 3712BC; Lot no. 35808016); Amphotericin B (Sigma Catalog no. A2411) 15 mM stock solution in DMSO; Pen/Strep (Gibco Catalog no. 10378-016; Lot no21040170) 100X in PBS.

Synthetic Defined Growth Medium

Complete Supplement Mixture; (Sunrise Science, Catalog No. 1001-100);Yeast Nitrogen Base without ammonium sulfate (MP Biomedicals, Catalog No. 4027-012); Glycerol (Sigma Catalog No. G-9012).

  1. Prepare medium by dissolving 0.79 g Complete Supplement Mixture and 1.7 g Yeast Nitrogen Base in 800 mL water.
  2. Add 20 mL glycerol, adjust final volume to 1000 mL. Filter sterilize.
Fungal Inoculum

Test Strains: C. glabrata (ATCC 2001), Saccharomyces cerevisiae W303

  1. Inoculate 100 μL of yeast from cryopreserved stock into 250 mL shaker flask containing 20 mL growth medium. Shake at 30°C overnight (16 hours).
  2. Read OD 600 of 1 mL of fungal culture in a cuvette using a standard optical density reader (Eppendorf BioPhotometer Plus), with growth medium as a background blank.
  3. Dilute to desired volume of fungal inoculum to produce an OD600 reading of 0.005 (S. cerevisiae) or 0.004 (C. glabrata).

Procedures

  1. Use a Thermo Combi nL to dispense 20 μL/well of assay media into all wells.
  2. Add 2 μL Amphotericin B to control wells.
  3. Pin 100 nL of test compound from compound plates into assay plates using a CyBi-Well pin tool.
  4. Dispense 20 μL/well of culture into the assay media in all wells.
  5. Incubate the plates in a humidified (90% humidity) Liconic incubator at 37°C without agitation for 48 hours.
  6. Shake plates briefly.
  7. Read OD600 of wells on a standard plate reader as measure of relative fungal growth. Envision (Perkin Elmer) plate reader settings: Ex Photometric 600, Top read.

Secondary Glucose Assay Protocol (AID 623971, AID 623967, AID 624013, AID 624025)

Materials and Reagents

Clear, flat bottom, black 384-well plates (Corning, Catalog No. 3712BC; Lot No. 35808016); Amphotericin B (Sigma, Catalog no. A2411) 15 mM stock solution in DMSO; Pen/Strep (Gibco, Catalog No. 10378-016; Lot no21040170) 100X in PBS.

Synthetic Defined Growth Medium

Complete Supplement Mixture; (Sunrise Science, Catalog No. 1001-100);Yeast Nitrogen Base without ammonium sulfate (MP Biomedicals, Catalog No. 4027-012); Glucose (Sigma, Catalog No. G7021).

  1. Prepare medium by dissolving 0.79 g Complete Supplement Mixture and 1.7 g Yeast Nitrogen Base in 800 mL water.
  2. Add 20 g glucose, adjust final volume to 1000 mL. Filter sterilize.
Fungal Inoculum

Test Strains: C. glabrata (ATCC 2001), Saccharomyces cerevisiae W303

  1. Inoculate 100 μL of yeast from cryopreserved stock into 250 mL shaker flask containing 20 mL growth medium. Shake at 30°C overnight (16 hours).
  2. Read OD 600 of 1 mL of fungal culture in a cuvette using a standard optical density reader (Eppendorf BioPhotometer Plus), with growth medium as a background blank.
  3. Dilute to desired volume of fungal inoculum to produce an OD600 reading of 0.005 (S. cerevisiae) or 0.004 (C. glabrata).

Procedures

  1. Use a Thermo Combi nL to dispense 20 μL/well of assay media into all wells.
  2. Add 2 μL Amphotericin B to control wells.
  3. Pin 100 nL of test compound from compound plates into assay plates using a CyBi-Well pin tool.
  4. Dispense 20 μL/well of culture into the assay media in all wells.
  5. Incubate the plates in a humidified (90% humidity) Liconic incubator at 37°C without agitation for 48 hours.
  6. Shake plates briefly.
  7. Read OD600 of wells on a standard plate reader as measure of relative fungal growth. Envision (Perkin Elmer) plate reader settings: Ex Photometric 600, Top read.

Secondary Mitochondrial Disruption Assay (AID 624062, AID 624068)

Materials and Reagents

JC-1 Dye (Invitrogen, Catalog No. T-3168); Carbonyl cyanide m-chlorophenyl hydrazone (CCCP) (Sigma Catalog No. C2759)

Synthetic Defined Growth Medium

Complete Supplement Mixture; (Sunrise Science, Catalog No. 1001-010);Yeast Nitrogen Base without ammonium sulfate (BD, Catalog No. 239210); Glucose (Sigma, Catalog No. G7021).

  1. Prepare medium by dissolving 0.79 g Complete Supplement Mixture and 6.7 g Yeast Nitrogen Base in 800 mL water.
  2. Add 20 g glucose, adjust final volume to 1000 mL. Filter sterilize.

Test Strains: Saccharomyces cerevisiae strain W303-1a (ATCC# 208352)

Candida glabrata (ATCC# 2001)

Procedures

  1. Grow a 2 mL overnight culture of desired strain in SD-CSM media at 30°C.
  2. Dilute to an OD600 of 0.05 in 2 mL of fresh media (one tube per compound replicate).
  3. Incubate at 30°C with agitation for 90 minutes.
  4. Add compounds at desired concentrations.
  5. Incubate 4 hr at 30°C with agitation.
  6. Aliquot 0.9 mL of SD-CSM media into a microcentrifuge tube.
  7. Add compounds at desired concentrations into microcentrifuge tubes
  8. Add JC-1 dye stock to tube at a final concentration of 4 μg/mL.
  9. Add 0.1 mL of fungal culture to each microcentrifuge tube and vortex.
  10. Incubate 20 minutes in the dark with agitation at 30°C.
  11. Pass stained cells through a 70 micron cell strainer into a FACS tube.
  12. Analyze by flow cytometry using a 488nM excitation laser and detect emission on FL1 and FL2 channels. Polarized mitochondria will emit strongly at ~590 nm in the FL2 channel; while depolarized mitochondria will emit mainly at ~525 nm in the FL1 channel.

Data Analysis

For the primary screen and other assays, negative-control (NC) wells and positive-control (PC) wells were included on every plate. The raw signals of the plate wells were normalized using the ‘Stimulators Minus Neutral Controls’ method in GeneData Assay Analyzer (v7.0.3). The median raw signal of the intra-plate NC wells was set to a normalized activity value of 0, while the median raw signal of the intra-plate PC wells was set to a normalized activity value of 100. Experimental wells were scaled to this range, resulting in an activity score representing the percent change in signal relative to the intra-plate controls. The mean of the replicate percent activities were presented as the final ‘PubChem Activity Score’. The ‘PubChem Activity Outcome’ class was assigned as described below, based on an activity threshold of 70%:

  • Activity_Outcome = 1 (inactive), less than half of the replicates fell outside the threshold.
  • Activity_Outcome = 2 (active), all of the replicates fell outside the threshold, OR at least half of the replicates fell outside the threshold AND the ‘PubChem Activity Score’ fell outside the threshold.
  • Activity_Outcome = 3 (inconclusive), at least half of the replicates fell outside the threshold AND the ‘PubChem Activity Score did not fall outside the threshold.

Appendix C. Experimental Procedures for the Synthesis of the Probe

General Details. All reagents and solvents were purchased from commercial vendors and used as received. NMR spectra were recorded on a Bruker 300 MHz or Varian UNITY INOVA 500 MHz spectrometer as indicated. Proton, fluorine, and carbon chemical shifts are reported in parts per million (ppm;δ) relative to tetramethylsilane or CFCl3 internal standards, or CDCl3 solvent (1H δ 0, 19F δ 0, 13C δ 77.16, respectively). NMR data are reported as follows: chemical shifts, multiplicity (obs = obscured, app = apparent, br = broad, s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet); coupling constant(s) in Hz; integration. Unless otherwise indicated, NMR data were collected at 25°C. Flash chromatography was performed using 40–60 μM Silica Gel (60 Å mesh) on a Teledyne Isco Combiflash Rf system. Tandem liquid chromatography/mass spectrometry (LCMS) was performed on a Waters 2795 separations module and Waters 3100 mass detector. Analytical thin layer chromatography (TLC) was performed on EM Reagent 0.25 mm silica gel 60-F plates. Visualization was accomplished with UV light and aqueous potassium permanganate (KMnO4) stain followed by heating. High-resolution mass spectra were obtained at the MIT Mass Spectrometry Facility with a Bruker Daltonics APEXIV 4.7 Tesla Fourier Transform Ion Cyclotron Resonance mass spectrometer. Compound purity and identity were determined by UPLC-MS (Waters, Milford, MA). Purity was measured by UV absorbance at 210 nm. Identity was determined on a SQ mass spectrometer by positive electrospray ionization. Mobile Phase A consisted of either 0.1% ammonium hydroxide or 0.1% trifluoroacetic acid in water, while mobile Phase B consisted of the same additives in acetonitrile. The gradient ran from 5% to 95% mobile Phase B over 0.8 minutes at 0.45 mL/min. An Acquity BEH C18, 1.7 μM, 1.0 × 50 mm column was used with column temperature maintained at 65°C. Compounds were dissolved in DMSO at a nominal concentration of 1 mg/mL, and 0.25 μL of this solution was injected.

Image ml316fu70

1-((4-Fluorophenyl)amino)cyclohexanecarbonitrile (4): Cyclohexanone (1.0 g, 10.2 mmol) was dissolved in glacial acetic acid (15 mL) and cooled to 10°C. 4-Fluoroaniline (1.4 g, 12.2 mmol, 1.2 equiv) was added in portions to the reaction, and the resulting mixture was stirred at 10°C for 15 minutes. Trimethylsilyl cyanide (1.4 mL, 11.2 mmol, 1.1 equiv) was then added. The reaction was warmed to room temperature and stirred for 15 hours. The reaction was poured into ice-cold ammonium hydroxide solution (30 mL, 20% v/v in water) to give a basic solution (pH > 10). This mixture was then extracted with dichloromethane (2 × 60 mL). The combined organic extracts were washed with brine (80 mL), shaken over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude residue was diluted with hexanes, and the precipitated solids were collected by filtration. After washing with more hexanes, the solids were air-dried and used without further purification (1.7 g, 76%). 1H NMR (300 MHz, CDCl3): δ 7.02-6.88 (m, 4H), 3.46 (br. s, 1H), 2.29-2.19 (m, 2H), 1.85-1.54 (m, 7H), 1.41-1.22 (m, 1H); MS (ESI+): 219 (M+H).

Image ml316fu71

1-(4-Fluorophenyl)-1,3-diazaspiro[4.5]decane-2,4-dione (5): Compound 4 (1.7 g, 7.8 mmol) was suspended in glacial acetic acid (20 mL) and treated with sodium cyanate (0.81 g, 12.5 mmol, 1.6 equiv). The resulting mixture was heated to 60°C and stirred at this temperature for 3 hours. A solution of concentrated hydrochloric acid (3 mL) in water (2 mL) was added to the reaction. The resulting mixture was then heated to 90°C and stirred at this temperature for 30 minutes. The reaction was cooled to room temperature then poured into water (50 mL). The precipitated solids were collected by filtration, washed with water, and air-dried (1.13 g, 55%). 1H NMR (300 MHz, CDCl3): δ 8.24 (s, 1H), 7.22-7.10 (m, 4H), 2.15-1.92 (m, 4H), 1.73-1.45 (m, 5H), 1.07-0.90 (m, 1H); MS (ESI+): 263 (M+H).

Image ml316fu72

1-(4-Fluorophenyl)-2-thioxo-1,3-diazaspiro[4.5]decan-4-one (6): A solution of compound 5 (0.54 g, 2.1 mmol) in toluene (17 mL) was treated with Lawesson’s reagent (0.46 g, 1.1 mmol, 0.55 equiv). This mixture was heated to reflux and stirred for 5 hours. The reaction was cooled to room temperature then concentrated under reduced pressure. The crude material was purified by column chromatography over silica gel (hexanes/ethyl acetate: 100/0 to 90/10) to give the title compound as a solid (0.50 g, 87%). 1H NMR (300 MHz, CDCl3): δ 9.13 (s, 1H), 7.23-7.15 (m, 4H), 2.17-1.96 (m, 4H), 1.75-1.48 (m, 5H), 1.07-0.91 (m, 1H); MS (ESI+): 279 (M+H).

Image ml316fu73

Ethyl 1-(4-fluorophenyl)-4-oxo-2-thioxo-1,3-diazaspiro[4.5]decane-3-carboxylate (7): Compound 6 (0.26 g, 0.93 mmol) was dissolved in dichloromethane (9.3 mL) and cooled to 0°C. Triethylamine (0.18 mL, 1.9 mmol, 2.0 equiv) was added, followed by ethyl chloroformate (0.26 mL, 1.9 mmol, 2.0 equiv). The reaction was stirred at 0°C for 2 hours before quenching with water (10 mL). The layers were separated and the aqueous phase was extracted with dichloromethane (3 × 10 mL). The combined organic layers were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The crude material was purified by column chromatography over silica gel (hexanes/ethyl acetate: 100/0 to 80/20) to give the title compound as a white solid (87.1 mg, 27%). 1H NMR (500 MHz, CDCl3): δ 7.25-7.17 (m, 4H), 4.53 (q, J = 7.1 Hz, 2H), 2.13-2.02 (m, 4H), 1.77-1.65 (m, 3H), 1.57 (td, J = 13.5, 4.2 Hz, 2H), 1.47 (t, J = 7.1 Hz, 3H), 1.06-0.95 (m, 1H); 19F NMR (282 MHz, CDCl3): δ -110.88 (tt, J = 7.6, 5.3 Hz); 13C NMR (125 MHz, CDCl3): δ 177.3, 171.7, 162.9 (d, JC-F = 250.6 Hz), 148.5, 132.4 (d, JC-CF = 8.9 Hz), 130.4, 116.9 (d, JC-CF = 22.9 Hz), 67.3, 65.8, 32.4, 23.9, 20.6, 13.9; HRMS (ESI+): calculated for C17H19FN2O3SNa [M + Na] 373.0993, found 373.1100.

Appendix D. Experimental Procedure for Additional Analytical Assays

Solubility. Solubility was determined in phosphate buffered saline (PBS) pH 7.4 with 1% DMSO. Each compound was prepared in duplicate at 100 μM in both 100% DMSO and PBS with 1% DMSO. Compounds were allowed to equilibrate at room temperature with a 250 rpm orbital shake for 24 hours. After equilibration, samples were analyzed by UPLC-MS (Waters, Milford, MA) with compounds detected by SIR detection on a single quadrupole mass spectrometer. The DMSO samples were used to create a two-point calibration curve to which the response in PBS was fit.

PBS Stability. Stability was determined in the presence of PBS pH 7.4 with 0.1% DMSO. Each compound was prepared in duplicate on six separate plates and allowed to equilibrate at room temperature with a 250-rpm orbital shake for 48 hours. One plate was removed at each time point (0, 2, 4, 8, 24, and 48 hours). An aliquot was removed from each well and analyzed by UPLC-MS (Waters, Milford, MA) with compounds detected by SIR detection on a single quadrupole mass spectrometer. Additionally, to the remaining material at each time point, acetonitrile was added to force dissolution of compound (to test for recovery of compound). An aliquot of this was also analyzed by UPLC-MS.

Plasma Protein Binding. Plasma protein binding was determined by equilibrium dialysis using the Rapid Equilibrium Dialysis (RED) device (Pierce Biotechnology, Rockford, IL) for both human and mouse plasma. Each compound was prepared in duplicate at 5 μM in plasma (0.95% acetonitrile, 0.05% DMSO) and added to one side of the membrane (200 μL) with PBS pH 7.4 added to the other side (350 μL). Compounds were incubated at 37ºC for 5 hours with a 250-rpm orbital shake. After incubation, samples were analyzed by UPLC-MS (Waters, Milford, MA) with compounds detected by SIR detection on a single quadrupole mass spectrometer.

Plasma Stability. Plasma stability was determined at 37ºC at 5 hours in both human and mouse plasma. Each compound was prepared in duplicate at 5 μM in plasma diluted 50/50 (v/v) with PBS pH 7.4 (0.95% acetonitrile, 0.05% DMSO). Compounds were incubated at 37ºC for 5 hours with a 250-rpm orbital shake with time points taken at 0 hours and 5 hours. Samples were analyzed by UPLC-MS (Waters, Milford, MA) with compounds detected by SIR detection on a single quadrupole mass spectrometer.

Appendix E. Chemical Characterization Data of Probe

1H NMR Spectrum (500 MHz, CDCl3) of Probe ML316.

1H NMR Spectrum (500 MHz, CDCl3) of Probe ML316

19F NMR Spectrum (282 MHz, CDCl3) of Probe ML316.

19F NMR Spectrum (282 MHz, CDCl3) of Probe ML316

13C NMR Spectrum (125 MHz, CDCl3) of Probe ML316.

13C NMR Spectrum (125 MHz, CDCl3) of Probe ML316

UPLC-MS Chromatogram of Probe ML316.

UPLC-MS Chromatogram of Probe ML316

Appendix F. Prior Art Search

Investigation into relevant prior art entailed searching the following databases: SciFinder, Web of Science, Reaxys, PubChem, PubMed, US Patent and Trademark Office (USPTO PatFT and AppFT), and World Intellectual Property Organization (WIPO). The search terms applied and hit statistics are provided in Table A2. As indicated, abstracts were obtained for the references returned and were analyzed for relevance to the current project. The searches were originally performed on August 13, 2011. SciFinder searches were performed again and updated on March 29, 2012.

Table A2. Search Strings and Databases Employed in Prior Art Search.

Table A2

Search Strings and Databases Employed in Prior Art Search.

Appendix G. Chemical Characterization Data for All Analogs

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID3889161.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID3889161

UPLC-MS Chromatogram of Analog CID3889161.

UPLC-MS Chromatogram of Analog CID3889161

FW = 346.44

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID3951437.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID3951437

UPLC-MS Chromatogram of Analog CID3951437.

UPLC-MS Chromatogram of Analog CID3951437

FW = 332.42

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604849.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604849

UPLC-MS Chromatogram of Analog CID56604849.

UPLC-MS Chromatogram of Analog CID56604849

FW = 360.47

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604859.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604859

UPLC-MS Chromatogram of Analog CID56604859.

UPLC-MS Chromatogram of Analog CID56604859

FW = 360.47

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604875.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604875

UPLC-MS Chromatogram of Analog CID56604875.

UPLC-MS Chromatogram of Analog CID56604875

FW = 394.49

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604866.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604866

UPLC-MS Chromatogram of Analog CID56604866.

UPLC-MS Chromatogram of Analog CID56604866

FW = 408.51

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604886.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604886

UPLC-MS Chromatogram of Analog CID56604886.

UPLC-MS Chromatogram of Analog CID56604886

FW = 352.47

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604869.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604869

UPLC-MS Chromatogram of Analog CID56604869.

UPLC-MS Chromatogram of Analog CID56604869

FW = 344.47

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604814.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604814

UPLC-MS Chromatogram of Analog CID56604814.

UPLC-MS Chromatogram of Analog CID56604814

FW = 330.49

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604835.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604835

UPLC-MS Chromatogram of Analog CID56604835.

UPLC-MS Chromatogram of Analog CID56604835

FW = 288.41

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604892.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604892

UPLC-MS Chromatogram of Analog CID56604892.

UPLC-MS Chromatogram of Analog CID56604892

FW = 274.38

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604821.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604821

UPLC-MS Chromatogram of Analog CID56604821.

UPLC-MS Chromatogram of Analog CID56604821

FW = 332.42

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604819.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604819

UPLC-MS Chromatogram of Analog CID56604819.

UPLC-MS Chromatogram of Analog CID56604819

FW = 362.44

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604860.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604860

UPLC-MS Chromatogram of Analog CID56604860.

UPLC-MS Chromatogram of Analog CID56604860

FW = 350.41

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604865.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604865

UPLC-MS Chromatogram of Analog CID56604865.

UPLC-MS Chromatogram of Analog CID56604865

FW = 366.86

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604813.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604813

UPLC-MS Chromatogram of Analog CID56604813.

UPLC-MS Chromatogram of Analog CID56604813

FW = 400.42

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604847.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604847

UPLC-MS Chromatogram of Analog CID56604847.

UPLC-MS Chromatogram of Analog CID56604847

FW = 357.43

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID24052146.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID24052146

UPLC-MS Chromatogram of Analog CID24052146.

UPLC-MS Chromatogram of Analog CID24052146

FW = 346.44

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604823.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604823

UPLC-MS Chromatogram of Analog CID56604823.

UPLC-MS Chromatogram of Analog CID56604823

FW = 346.44

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604836.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604836

UPLC-MS Chromatogram of Analog CID56604836.

UPLC-MS Chromatogram of Analog CID56604836

FW = 360.47

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604858.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604858

UPLC-MS Chromatogram of Analog CID56604858.

UPLC-MS Chromatogram of Analog CID56604858

FW = 374.50

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604817.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604817

UPLC-MS Chromatogram of Analog CID56604817.

UPLC-MS Chromatogram of Analog CID56604817

FW = 374.50

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604837.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604837

UPLC-MS Chromatogram of Analog CID56604837.

UPLC-MS Chromatogram of Analog CID56604837

FW = 388.52

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604882.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604882

UPLC-MS Chromatogram of Analog CID56604882.

UPLC-MS Chromatogram of Analog CID56604882

FW = 408.51

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604830.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604830

UPLC-MS Chromatogram of Analog CID56604830.

UPLC-MS Chromatogram of Analog CID56604830

FW = 256.32

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604896.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604896

UPLC-MS Chromatogram of Analog CID56604896.

UPLC-MS Chromatogram of Analog CID56604896

FW = 298.40

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604878.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604878

UPLC-MS Chromatogram of Analog CID56604878.

UPLC-MS Chromatogram of Analog CID56604878

FW = 346.44

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604818.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604818

UPLC-MS Chromatogram of Analog CID56604818.

UPLC-MS Chromatogram of Analog CID56604818

FW = 348.42

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604843.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604843

UPLC-MS Chromatogram of Analog CID56604843.

UPLC-MS Chromatogram of Analog CID56604843

FW = 332.42

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604894.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604894

UPLC-MS Chromatogram of Analog CID56604894.

UPLC-MS Chromatogram of Analog CID56604894

FW = 318.39

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604873.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604873

UPLC-MS Chromatogram of Analog CID56604873.

UPLC-MS Chromatogram of Analog CID56604873

FW = 306.38

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604827.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604827

UPLC-MS Chromatogram of Analog CID56604827.

UPLC-MS Chromatogram of Analog CID56604827

FW = 330.38

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604867.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604867

UPLC-MS Chromatogram of Analog CID56604867.

UPLC-MS Chromatogram of Analog CID56604867

FW = 435.58

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604852.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604852

UPLC-MS Chromatogram of Analog CID56604852.

UPLC-MS Chromatogram of Analog CID56604852

FW = 346.44

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604845.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604845

UPLC-MS Chromatogram of Analog CID56604845.

UPLC-MS Chromatogram of Analog CID56604845

FW = 272.34

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604877.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604877

UPLC-MS Chromatogram of Analog CID56604877.

UPLC-MS Chromatogram of Analog CID56604877

FW = 314.42

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604857.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604857

UPLC-MS Chromatogram of Analog CID56604857.

UPLC-MS Chromatogram of Analog CID56604857

FW = 330.49

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604883.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604883

UPLC-MS Chromatogram of Analog CID56604883.

UPLC-MS Chromatogram of Analog CID56604883

FW = 258.32

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604884.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604884

UPLC-MS Chromatogram of Analog CID56604884.

UPLC-MS Chromatogram of Analog CID56604884

FW = 220.25

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604829.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604829

UPLC-MS Chromatogram of Analog CID56604829.

UPLC-MS Chromatogram of Analog CID56604829

FW = 256.32

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604856.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604856

UPLC-MS Chromatogram of Analog CID56604856.

UPLC-MS Chromatogram of Analog CID56604856

FW = 274.38

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604841.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604841

UPLC-MS Chromatogram of Analog CID56604841.

UPLC-MS Chromatogram of Analog CID56604841

FW = 184.26

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604862.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604862

UPLC-MS Chromatogram of Analog CID56604862.

UPLC-MS Chromatogram of Analog CID56604862

FW = 278.35

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID823120.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID823120

UPLC-MS Chromatogram of Analog CID823120.

UPLC-MS Chromatogram of Analog CID823120

FW = 260.36

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604820.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604820

UPLC-MS Chromatogram of Analog CID56604820.

UPLC-MS Chromatogram of Analog CID56604820

FW = 226.34

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID14297244.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID14297244

UPLC-MS Chromatogram of Analog CID14297244.

UPLC-MS Chromatogram of Analog CID14297244

FW = 274.38

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604885.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604885

UPLC-MS Chromatogram of Analog CID56604885.

UPLC-MS Chromatogram of Analog CID56604885

FW = 276.35

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604876.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604876

UPLC-MS Chromatogram of Analog CID56604876.

UPLC-MS Chromatogram of Analog CID56604876

FW = 260.36

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604811.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604811

UPLC-MS Chromatogram of Analog CID56604811.

UPLC-MS Chromatogram of Analog CID56604811

FW = 246.33

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604879.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604879

UPLC-MS Chromatogram of Analog CID56604879.

UPLC-MS Chromatogram of Analog CID56604879

FW = 234.32

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604844.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID56604844

UPLC-MS Chromatogram of Analog CID56604844.

UPLC-MS Chromatogram of Analog CID56604844

FW = 206.26

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID759335.

1H NMR Spectrum (300 MHz, CDCl3) of Analog CID759335

UPLC-MS Chromatogram of Analog CID759335.

UPLC-MS Chromatogram of Analog CID759335

FW = 190.20

Appendix H. Compounds Provided to Evotec

Table A3Probe and Analog Information

BRDSIDCIDP/AMLSIDML
BRD-K83072125-001-01-213435665056604860PMLS004256613ML316
BRD-K97464451-001-02-71314047603889161AMLS004256614N/A
BRD-K51126930-001-01-913435663956604827AMLS004256611N/A
BRD-K54655126-001-01-813435664656604856AMLS004256609N/A
BRD-K86579583-001-01-813435662056604819AMLS004256610N/A
BRD-K65907975-001-01-213435663456604859AMLS004256612N/A

A = analog; N/A = not applicable; P = probe

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this page (4.6M)

Related information

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...