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

Discovery of ML370, an inhibitor of Vibrio cholerae Quorum Sensing Acting via the LuxO response regulator

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

Author Information and Affiliations

Received: ; Last Update: September 18, 2014.

Quorum sensing (QS) is a process of bacterial cell-to-cell communication that relies upon recognition of extracellular signaling molecules called autoinducers. QS allows bacteria to synchronize their behavior in response to changes in the population density and species composition of the proximal bacterial community. Known behaviors regulated by QS include bioluminescence, sporulation, virulence factor production, and biofilm formation. We carried out a high throughput screen (HTS) to identify small molecules that modulate QS in a modified V. cholerae strain carrying a luciferase operon; activation of the quorum pathway is accompanied by light production. 352,083 compounds from the NIH-MLPCN compound library were evaluated. Potential QS modulators were characterized via additional bacterial epistatic assays to elucidate the mode of action. We report the discovery and medicinal chemistry development of a substituted pyrazoloquinoline (ML370) shown to be an inhibitor of Vibrio cholerae LuxO, a response regulator and intracellular kinase. The probe acts directly on LuxO by inhibiting the ATPase activity. ML370 should greatly expand the general understanding of how QS response regulators relay information from upstream signals that lead to modified gene expression. In addition ML370 and compound analogues could lead to the development of antibacterial drugs designed to interfere with QS that could have enormous ramifications for improving human health.

Assigned Assay Grant No: R03 MH094166-01

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

Chemistry Center Name & PI: University of Kansas Specialized Chemistry Center, Jeffrey Aubé, PhD

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

Assay Submitter & Institution: Bonnie L. Bassler, PhD., Princeton University, Princeton, NJ

PubChem Summary Bioassay Identifier (AID): 588521

Probe Structure & Characteristics

Image ml370f1
PubChem CID/MLTargetsEC50 (µM)
[SID, AID]
Anti-TargetIC50 (µM)
[SID, AID]
Fold Selective*
CID 70680248/ML370V. cholerae BH15782.5
[SID 160655705, AID 652239]
HeLa cytotoxicity>35
[SID 160655705,AID 686928]
>14
V. cholerae BH16512.7
[SID 160655705, AID 652289]
>13
V. cholerae DH2311.7
[SID 160655705, AID 686931
>20
V. cholerae WN11032.3
[SID 160655705, AID 686930]
>15
*

Selectivity = Cytotoxicity IC50/Target EC50

Recommendations for the scientific use of these probes

Discovery of quorum sensing (QS) receptor agonists or other QS pathway modulators will permit more precise control of Vibrio cholerae QS through the perturbation of distinct steps within the signaling cascade. This, in turn, would enable elucidation of the underlying principles governing response regulator interactions utilized by intracellular transcriptional activators, such as the intracellular kinase LuxO. Upon phosphorylation, LuxO undergoes a number of conformational changes. The newly identified inhibitor probe may shed light on the nature of QS activators and possibly provide a clearer understanding of LuxO activities. V. cholerae LuxO is highly homologous to LuxO proteins in other Vibrio sp. and a larger class of NtrC transcriptional activators [1]. If the probe targets a conserved domain, the probe may be useful to study two-component signaling pathways in a larger number of organisms [2].

Crystallography efforts to visualize LuxO are underway, and this high-quality probe will be evaluated for possible co-crystallization. Concurrently, directed mutagenesis will be applied to understand how ML370 interacts with LuxO. Identification of LuxO-inhibitor binding motifs will establish a framework for the development of antibacterial drugs designed to interfere with quorum sensing. This probe can also test the utility of quorum pathway activation as a means of treating cholera via reduction of the pathogen's virulence in vivo. ML370 can be tested in combination with CqsS agonists as a potential antibacterial regiment that could be potent and species-specific. There are several animal models available with which such studies may be conducted [3-5]. If such an approach is efficacious, these potential therapeutics, operating in a completely unexploited target space, would have enormous ramifications upon improving human health. ML370 is currently not optimized for in vivo experiments but medicinal chemistry efforts can be directed towards improving this scaffold's ADME profile to facilitate animal studies.

1. Introduction

Scientific Rationale

Bacteria utilize quorum sensing (QS) to assess the population density and species complexity of their environment and adapt their physiological behavior to the prevailing conditions. A large number of human pathogens utilize QS as a means to produce virulence factors. Therefore, the ability to prevent virulence factor production could have a huge impact on human health. An example of a QS bacterium is the human pathogen Vibrio cholerae. The World Health Organization estimates that reported cases represent only 5–10% of the actual number of cholera incidents. There are an estimated 3 to 5 million cases that occur every year and approximately 120,000 deaths [6]. These high case numbers persist despite the development of a vaccine and a simple treatment regimen. In addition to V. cholerae, there are at least six other Vibrio species that are human pathogens. It has been determined that V. cholerae, as well as other bacteria, rely on a two-component system to relay exogenous information to the cell interior. A transmembrane histidine kinase (i.e. sensor) and an internal response regulator are the primary constituents of these two-component systems [7]. V. cholerae possess two parallel QS pathways. One pathway is responsible for interspecies communication wherein LuxPQ is the transmembrane receptor. The second, genus-specific pathway is controlled by the CqsS sensor [8]. Both pathways converge upon a common intermediary, LuxO [9].

The endogenous ligand of CqsS is the α-hydroxy ketone 1 (Figure 1), also known as cholera autoinducer 1 (CAI-1) [9-10]. Through careful studies of mutant CqsS receptors, it has been determined that CAI-1 inhibits His194 auto-phosphorylation of CqsS [11]. The auto-phosphorylation of His194 is believed to initiate a phosphorelay sequence involving the intermediary transfer protein LuxU and terminates at the response regulator LuxO (Figure 2). LuxO is a member of the NtrC family of response regulators that require interaction with a σ-54 factor to activate transcription [12]. LuxO binds DNA upstream of RNA polymerase and contains an inherent ATPase activity that is needed for changes to the transcriptional complex which involves DNA looping and subsequent formation of an open promoter complex [13]. LuxO integrates the CqsS and AI-2 signaling pathways and a homologue has been found in all Vibrio species examined. There are species-specific differences on the number of pathways that feed into LuxO.

Figure 1. The Endogenous Ligand of the Vibrio cholerae Sensor Histidine Kinase CqsS, Cholera autoinducer-1 (CAI-1).

Figure 1

The Endogenous Ligand of the Vibrio cholerae Sensor Histidine Kinase CqsS, Cholera autoinducer-1 (CAI-1).

Figure 2. The V cholerae CqsS/CAI-1 Quorum Sensing Phosphorelay system.

Figure 2

The V cholerae CqsS/CAI-1 Quorum Sensing Phosphorelay system. At low cell density, there are insufficient amounts of CAI-1 to adequately bind CqsS. This leads to the production of AphA and the adoption of individual cell behaviors. At high cell density, (more...)

When CAI-1 is present in minute quantities, the phosphorelay system favors the phosphorylation of LuxO at Asp47 (Figure 2) [11]. Phosphorylated LuxO up-regulates transcription of the small regulatory RNAs qrr1-4, which subsequently promote translation of AphA while simultaneously suppressing translation of the HapR regulator. AphA regulates genes that are beneficial to individual behaviors, while HapR regulates genes that promote group (quorum) behaviors. Another level of regulating the RNA is the RNA chaperone Hfq that base pairs with and destabilizes the hapR mRNA [12]. When dense populations of V. cholerae synthesize and secrete sufficient quantities of CAI-1, this ligand will bind to CqsS and promote the production of HapR. The induction of HapR expression then allows the colonizing V. cholerae population to detach from the gut epithelium and vacate its host. Among the genes regulated by HapR is the expression of haemagglutinin protease (HapA) that facilitates the expulsion of V. cholerae from its human host [13]. In most QS bacteria, QS induces virulence factor production but Vibrio cholerae is distinct from non-vibrio bacterial species because QS results in repression of virulence factor and biofilm production, [2]. From a therapeutic perspective, the artificial activation of CqsS or inhibition of LuxO or Hfq to promote HapR expression could lead to a new generation of anti-cholera treatments. A potential combinatorial therapeutic approach targeting multiple targets in the QS pathway could ensure effective and genus-specific antimicrobial response.

Antagonism of LuxO has not been well-studied in the literature thus far. The only known small molecule inhibitors of LuxO were reported after the initiation of this project [2]. Derived from an aza-uracil scaffold, these compounds show moderate inhibition of LuxO activity (IC50's range from 5 to 20 µM) in cellular assays. Further investigation of the most potent compound, (Figure 3, Aza-U), determined that these LuxO antagonists block ATPase hydrolysis and prevent subsequent transcriptional activation. Aza-U was selected as the positive control for all cell-based SAR assays in the post-HTS medicinal chemistry phase of this project.

Figure 3. Aza-uracil-derived antagonists of LuxO ATPase activity.

Figure 3

Aza-uracil-derived antagonists of LuxO ATPase activity. Aza-U (4) and its analogs have been shown to inhibit the ATPase activity of LuxO. Quorum-sensing modulation was measured in V. cholera strains BH1578 and BH1651 (see Section 2) and their respective (more...)

It is hoped that the identification of additional small molecule modulators of LuxO could elucidate the mechanisms of the latter steps underpinning the V. cholerae QS phosphorelay system. The additional chemical matter will also assist in the structural characterization of potential binding pockets and, in combination with a clearer understanding of how the transcriptional activation occurs, could guide development of new QS agonists as potential anti-cholera agents. Since all vibrios possess a LuxO homologue, LuxO inhibitors should be efficacious against all pathogenic vibrios. Aza-U compounds were found to inhibit LuxO activity in V. harveyi and V. parahaemolyticus [2].

2. Materials and Methods

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

Materials and Reagents

  • CellTiter-Glo® Luminescent Cell Viability Assay was purchased from Promega (Catalog No. G7573; Madison, WI)

Bacterial strains & Cell Lines

The following cell lines were used in this study:

  • Vibrio cholerae BH1578; a genetically modified strain lacking LuxS and CqsA autoinducer synthases that was provided by the Bassler lab. This strain was used in the primary assay.
  • Vibrio cholerae BH1651; a genetically modified strain with constitutively active LuxO that was provided by the Bassler lab.
  • Vibrio cholerae DH231; a genetically modified strain lacking the CqsS receptor that was provided by the Bassler lab.
  • Vibrio cholerae WN1103; a genetically modified strain lacking the LuxQ receptor that was provided by the Bassler lab.
  • HeLa obtained from ATCC (Catalog Number CCL-2; Manassas, VA) is a human epithelial adenocarcinoma cell line used for mammalian cytotoxicity profiling

Note: All of the V. cholerae strains also carry the heterologous V. harveyi luxCDABE luciferase operon

Table 1Strains of V. cholera used in assays

StrainGenotypeReporterSuggested Mechanism for Inducers
BH1578ΔluxS ΔcqsAV. harveyi luciferase operonMultiple mechanisms possible
BH1651luxOD47EV. harveyi luciferase operonLuxO inhibitor or downstream
DH231ΔluxS ΔcqsSV. harveyi luciferase operonLuxPQ agonist or LuxO inhibition
WN1103ΔluxQ ΔcqsAV. harveyi luciferase operonCqsS agonist or LuxO inhibition
SLS353luxOD47EQrr4:GFP reporterLuxO inhibitor or downstream

2.1. Assays

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

2.1.1. Primary assay – V. cholerae BH1578 bioluminescence inducer assay (AID 588346, AID 602243, AID 624270, AID 652239, AID 686929)

A modified strain of Vibrio cholerae used in this assay uses light production to indicate quorum sensing induction. Vibrio cholerae is not naturally bioluminescent but the closely related species Vibrio harveyi produces light when the population is at a high density (i.e. a quorum is sensed). The heterologous Vibrio harveyi luciferase operon (luxCDABE) was cloned into the Vibrio cholerae C6706 O1 El Tor bacterial strain on the pBB1 cosmid to create a bioluminescence assay strain. This operon is activated by the endogenous V. cholerae quorum sensing pathway [2]. The BH1578 strain is a CqsA, LuxS double mutant that lacks both autoinducer synthases (CAI-1 and AI-2). BH1578 does not generate light in the absence of exogenous autoinducers but bioluminescence can be stimulated up to 10,000-fold by adding 1 µM (saturating) CAI-1. On day 0, a colony of bacteria was picked into 50 mL Luria Broth with 10 µg/mL tetracycline and cultured overnight at 30 °C. On day 1, bacterial density was determined by spectrometry, and the OD600 was adjusted to 0.3. 20 µL of Luria Broth with 10 µg/mL tetracycline was added per well into white, opaque 384 well plates. Compounds and controls were added by pin transfer method. The HTS used 150 nL of compound for screening at 20 µM, and the retest assays used 100 nL of compound. After 6 hours, the luminescence signal was determined with the Perkin-Elmer EnVision plate reader. In addition to luminescence, the confluency of each well was measured at an absorbance of 600 nM. Primary HTS data were analyzed in Genedata Screener Assay Analyzer. All values were normalized against DMSO treated samples and the positive control (1 µM CAI-1, CID24892809). For the HTS, the average of two replicates was used to rank order activity and to choose compounds for retests. For dose studies, percent (%) activity was determined for each concentration, and the concentration response curves (CRCs) were generated with Genedata Screener's Condoseo.

2.1.2. Secondary assay – HeLa cytotoxicity assay (AID 624140, AID 686928)

HeLa cells were treated with compounds for 24 hours, and then cell viability was measured using the CellTiter-Glo Assay (Promega), a luciferase-based reagent that measures cellular ATP levels. The compounds were tested at different concentrations to determine IC50 values. Compounds that were inactive (IC50 ≥ 30 µM) in this assay were considered for probe development. Data were normalized against DMSO in Genedata Screener's Assay Analyzer. Curves were generated with Genedata Screener's Condoseo and showed percent (%) activity for the individual doses.

2.1.3. Secondary assay – V. cholerae BH1651 LuxO inhibitor assay (AID 624254, AID 624269, AID 652289)

A modified strain of Vibrio cholerae used in this assay uses light production to indicate quorum sensing induction. Vibrio cholerae is not naturally bioluminescent but the closely related species Vibrio harveyi produces light when the population is at a high density (i.e. a quorum is sensed). The heterologous Vibrio harveyi luciferase operon (luxCDABE) was cloned into the Vibrio cholerae C6706 O1 El Tor bacterial strain on the pBB1 cosmid to create a bioluminescence assay strain. This operon is activated by the endogenous V. cholerae quorum sensing pathway [2]. The BH1651 strain is a luxOD47E mutant where LuxOD47E mimics the behavior of phosphorylated LuxO, rendering LuxOD47E constitutively active within the QS pathway. BH1651 does not generate light but any compound that inhibits LuxO or works downstream of LuxO will induce light production. On day 0, a colony of bacteria was picked into 50 mL Luria Broth with 10 µg/mL tetracycline and cultured overnight at 30 °C. On day 1, bacterial density was determined by spectrometry, and the OD600 was adjusted to 0.3. 20 uL of Luria Broth with 10 µg/mL tetracycline was added per well into white, opaque 384 well plates. Compounds and controls were added by pin transfer method. After 6 hours, the luminescence signal was determined with the Perkin-Elmer EnVision plate reader. In addition to luminescence, the confluency of each well was measured at an absorbance of 600 nM. Data were analyzed in Genedata Screener Assay Analyzer. All values were normalized against DMSO and 40 µM azaU (positive control) treated samples. Percent (%) activity was determined for each concentration, and the concentration response curves (CRCs) were generated with Genedata Screener's Condoseo.

2.1.4. Secondary assay – V. cholerae DH231 sensor mechanism assay (AID 624281, AID 686931)

A modified strain of Vibrio cholerae used in this assay uses light production to indicate quorum sensing induction. Vibrio cholerae is not naturally bioluminescent but the closely related species Vibrio harveyi produces light when the population is at a high density (i.e. a quorum is sensed). The heterologous Vibrio harveyi luciferase operon (luxCDABE) was cloned into the Vibrio cholerae C6706 O1 El Tor bacterial strain on the pBB1 cosmid to create a bioluminescence assay strain. This operon is activated by the endogenous V. cholerae quorum sensing pathway [2]. The DH231 strain is a LuxS and CqsS double deletion mutant. DH231 does not generate light but any compound that agonizes the receptor LuxQ will induce light production, and CqsS agonists will have no activity in this assay. On day 0, a colony of bacteria was picked into 50 mL Luria Broth with 10 µg/mL tetracycline and cultured overnight at 30 °C. On day 1, bacterial density was determined by spectrometry, and the OD600 was adjusted to 0.3. 20 µL of Luria Broth with 10 µg/mL tetracycline was added per well into white, opaque 384 well plates. Compounds and controls were added by pin transfer method. After 6 hours, the luminescence signal was determined with the Perkin-Elmer EnVision plate reader. In addition to luminescence, the confluency of each well was measured at an absorbance of 600 nM. Data were analyzed in Genedata Screener Assay Analyzer. All values were normalized against DMSO and 40 µM azaU (positive control) treated samples. Percent (%) activity was determined for each concentration, and the concentration response curves (CRCs) were generated with Genedata Screener's Condoseo.

2.1.5. Secondary assay – V. cholerae WN1103 sensor mechanism assay (AID 624275, AID 686930)

A modified strain of Vibrio cholerae used in this assay uses light production to indicate quorum sensing induction. Vibrio cholerae is not naturally bioluminescent but the closely related species Vibrio harveyi produces light when the population is at a high density (i.e. a quorum is sensed). The heterologous Vibrio harveyi luciferase operon (luxCDABE) was cloned into the Vibrio cholerae C6706 O1 El Tor bacterial strain on the pBB1 cosmid to create a bioluminescence assay strain. This operon is activated by the endogenous V. cholerae quorum sensing pathway [15]. The WN1103 strain is a luxPQ and CqsA double deletion mutant. DH231 does not generate light but any compound that agonizes the CqsS receptor will induce light production and LuxQ agonists will have no activity in this assay. On day 0, a colony of bacteria was picked into 50 mL Luria Broth with 10 µg/mL tetracycline and cultured overnight at 30 °C. On day 1, bacterial density was determined by spectrometry, and the OD600 was adjusted to 0.3. 20 µL of Luria Broth with 10 µg/mL tetracycline was added per well into white, opaque 384 well plates. Compounds and controls were added by pin transfer method. After 6 hours, the luminescence signal was determined with the Perkin-Elmer EnVision plate reader. In addition to luminescence, the confluency of each well was measured at an absorbance of 600 nM. Data were analyzed in Genedata Screener Assay Analyzer. All values were normalized against DMSO and 40 µM azaU (positive control) treated samples. Percent (%) activity was determined for each concentration, and the concentration response curves (CRCs) were generated with Genedata Screener's Condoseo.

2.1.6. Secondary assay – V. cholerae SLS353 qrr4:GFP fluorescence reporter assay (AID 652219, AID 686942)

A modified strain of Vibrio cholerae used in this assay uses fluorescence to indicate qrr4 sRNA transcription [16]. The SLS353 strain is a luxOD47E mutant strain containing the pSLS4 plasmid where the qrr4 promoter drives GFP expression. LuxOD47E mimics the behavior of phosphorylated LuxO, rendering LuxOD47E constitutively active. This leads to steady levels of qrr4 promoter-driven GFP. Any compound that inhibits LuxO or works downstream of LuxO will decrease qrr4:GFP expression. Since the GFP protein tends to be more stable than luciferase, the compound incubation is increased from 6 to 16 hours. On day 0, a colony of bacteria was picked into 50 mL Luria Broth with 100 µg/mL kanamycin and cultured overnight at 30 °C. On day 1, bacterial density was determined by spectrometry, and the OD600 was adjusted to 0.3. 20 µL of Luria Broth with 100 µg/mL kanamycin was added per well into white, opaque 384 well plates. Compounds and controls were added by pin transfer method. After 16 hours, the fluorescence signal was determined with the Perkin-Elmer EnVision plate reader using 480 nM excitation and 509 nM emission wavelength spectra. All values were compared to DMSO and 40 µM azaU (positive control) treated samples. The concentration response curves (CRCs) were generated with GraphPad Prism 6.

2.1.7. Secondary assay – LuxO in vitro ATPase assay (AID 686936)

A modified coupled-enzyme assay measured the rate of ATP hydrolysis by LuxOD47E [2]. Briefly, ADP released from ATP by LuxOD47E is reacted with phosphoenolpyruvate (PEP) to form pyruvate using pyruvate kinase (PK). Pyruvate is reacted with NADH to form NAD and lactate using lactate dehydrogenase (LDH). The rate of NAD production is followed at 340 nm using a spectrophotometer. ATP hydrolysis rates were inferred from the absorbance change observed (εNADH,340NAD,340 = 6220 M-1 cm-1 for NADH) [2]. The rates of ATP hydrolysis by LuxOD47E were measured in reactions containing 100 mM Sodium phosphate buffer pH 7.4, 5 mM MgCl2, 0.2 mM NADH, 1 mM PEP, 5–20 units of PK/LDH mix (Sigma), and 10 mM LuxOD47E. ATP and inhibitors were added to the reactions at indicated concentrations. The rate of ATP hydrolysis was monitored for 5 minutes. Data were fitted using GraphPad Prism to obtain the kinetic parameters. Percent ATPase inhibition was calculated.

2.2. Probe Chemical Characterization

Probe ML370 was prepared as described in Section 2.3. Data from 1H NMR, 13C NMR and high resolution mass spectrometry are all consistent with the proposed structure. The purity of the compound is 96.1 area% at 214 nm by UPLC. Synthetic procedures for the preparation of the probe from commercially available m-anisidine are found in Appendix C and spectral data for the probe are in Appendix E.

The solubility of ML370 was experimentally determined to be 33.5 µM in phosphate buffered saline (PBS) with 1% (v/v) DMSO. Solubility was also measured to be 49.4 µM in the Luria Broth (LB) media used in the primary assay. Table 2 summarizes the solubility of ML370 and several analogs in both PBS and LB media. In general, LB media is a better solvent than PBS for these compounds.

Table 2. Solubility of ML370 and Analogs in PBS and LB Media.

Table 2

Solubility of ML370 and Analogs in PBS and LB Media.

The probe ML370 is stable in 1:1 acetonitrile:PBS solution (>99% remaining after 48-hour incubation at 23 °C). No significant reaction with DTT was observed for ML370 (>99% remaining after 8-hour incubation). The data from the PBS stability and DTT stability assays is provided in Figure 4. The probe is also stable to human and murine plasma with greater than 98% and 85% remaining respectively after incubation at 37 °C for 5 hours. Table 3 summarizes the results for the various stability assays performed. Experimental procedures for all analytical assays are provided in Appendix D.

Figure 4. Stability of Probe ML370 in 1:1 Acetonitrile:PBS (pH 7.4, 23 °C).

Figure 4

Stability of Probe ML370 in 1:1 Acetonitrile:PBS (pH 7.4, 23 °C). MLS004820389 (CID 70680248, SID 160655705) was tested over a time course in aqueous stability assay and thiol stability assay. (A) Stability of the probe over 8 hours in 1:1 acetonitrile:PBS; (more...)

Table 3. Thiol Stability (DTT) and Plasma Stability for ML370.

Table 3

Thiol Stability (DTT) and Plasma Stability for ML370.

The physical properties of probe ML370 are summarized in Table 4

Table 4. Summary of Probe Properties Computed for ML370.

Table 4

Summary of Probe Properties Computed for ML370.

2.3. Probe Preparation

Probe ML370 was synthesized in five steps and 7.5% overall yield as shown in Scheme 1. m-Anisidine was acetylated using neat acetic acid under microwave irradiation to afford acetanilide KU2. A Meth-Cohn quinolone synthesis using this material gave aldehyde KU3, which was converted to the nitrile (KU4) in one step using hydroxylamine and propylphosphonic anhydride [17]. Reaction of nitrile KU4 with methylhydrazine afforded the 1H-pyrazolo[3,4-b]quinolone core (penultimate compound KU5), which was acylated using furoyl chloride to give probe ML370.

Scheme 1. Synthesis of Probe ML370.

Scheme 1

Synthesis of Probe ML370.

Full experimental details for the preparation of ML370 are provided in Appendix C.

3. Results

Probe attributes

  • Induces light production in V. cholerae strain BH1578 with EC50 ≤ 5 µM
  • Induces light production in V. cholerae strains BH1651, DH231 & WN1103
  • Non-toxic to HeLa cells with IC50 ≥ 35 µM

The current project utilizes several genetically modified strains of the human pathogen Vibrio cholerae in an effort to identify potential activators of the V. cholerae quorum sensing (QS) network. The MLSMR collection of 352,083 compounds was examined for possible QS activators, using V. cholerae BH1578 as the primary test strain. In the V. cholerae BH1578 strain, the V. harveyi luxCDABE (luciferase) operon was introduced as a cosmid [16], and the operon is activated by endogenous mechanisms to elicit light upon reaching a quorum. In addition, the BH1578 strain is a cqsA, luxS double deletion mutant that lacks the ability to produce any endogenous autoinducers of QS. Therefore, light production can only occur in response to a screening compound. Secondary assays utilizing additional V. cholerae mutants were included in order to triage active compounds by their potential mode of action. Based on their behavior in the various secondary assays, compounds were categorized as an agonist of either CqsS or LuxQ; an inhibitor of LuxO; or as an inhibitor of Hfq.

Analysis of the primary screening data revealed that approximately 500 compounds could activate the V. cholerae QS pathway. Re-evaluation of these compounds in a dose-response format led to the selection of 26 compounds for subsequent examination as dry powders. Based on their cellular potency in BH1578 and low toxicity towards HeLa cells, two scaffolds (CID 665390 & CID 4443990) were prioritized for further development as LuxO inhibitors. The optimization of CID 4443990 to ML366 is reportedly separately.

The pyrazoloquinoline CID 665390 (Figure 5) is an activator of V. cholera QS with a measured AC50 = 12 µM with no observable toxicity in HeLa cells (IC50 > 35 µM). Analogs of CID 665390 were designed, prepared, and evaluated for QS activation. The structure-activity relationship (SAR) was quite steep; however, CID 70680248 proved to be a more efficacious compound (see Section 3.4).

Figure 5. Dose Response Curves for initial hit (CID 665390) and optimized probe (CID 70680248).

Figure 5

Dose Response Curves for initial hit (CID 665390) and optimized probe (CID 70680248). CID 665390 & CID 70680248 were tested across a range of concentrations up to 35 µM in the primary assay and the BH1651 assay. Concentration response (more...)

3.1. Summary of Screening Results

As described above, the Molecular Libraries and Small Molecules Repository (MLSMR) collection of 352,083 unique compounds were tested at a single concentration (20 µM) for their ability to induce the production of luciferase by BH1578, thus indicating successfully activation of the QS signaling pathway. 553 compounds were active at 20 µM, representing a nominal hit rate of 0.15%.

The active compounds were then re-tested in a dose-response format, wherein only 2 compounds actually met or exceeded the project requirement of AC50 ≤ 1.0 µM. However, the low success rate of the primary assay prompted us to expand the primary assay cut-off to include substances with AC50 ≤ 10 µM.

Subsequently, dry powders for 26 compounds were procured for re-testing. After purity analysis and structural confirmation by NMR spectroscopy, these substances were screened according to the workflow outlined below in Figure 6.

Figure 6. Critical Path for Probe Development.

Figure 6

Critical Path for Probe Development.

The assay progression outlined in Figure 6 assessed the activation of QS pathways in several V. cholerae strains: BH1578, BH1651, and SLS353. CID 665390 emerged as a promising LuxO inhibitor and was subsequently prioritized for development. A number of analogs were prepared and assayed for QS activation. The results of the SAR investigation are presented in detail in Section 3.4.

3.2. Representative Dose Response Curves for Probe ML370

Figure 7. ML370 Induces Quorum Sensing via LuxO ATPase Inhibition without Toxicity.

Figure 7ML370 Induces Quorum Sensing via LuxO ATPase Inhibition without Toxicity

ML370 was tested across a range of concentrations up to 35 µM in the primary BH1578 assay, the SLS353 qrr4 reporter assay, the in vitro LuxO ATPase assay and a HeLa cytotoxicity assay. Concentration response curves were generated with Genedata Screener Condeseo and show normalized percent activity for the individual doses. ML370 DH231 assay (AID 686931), IC50 = 1.8 µM (A); ML370 and ML366 SLS353 assay, (B); ML370 in vitro LuxO ATPase assay (C); ML370 HeLa cytotoxicity (AID 686928), IC50 ≥ 35 µM (D). ○ = replicate 1, △ = replicate 2.

3.3. Scaffold/Moiety Chemical Liabilities

A search of PubChem for ML370 returns no biological assay results (as of April 8, 2013). A structure-based search (80% similarity to ML370) of the Chemical Abstracts database using the SciFinder software returned 12 analogs of ML370 with biological studies reported (as of April 8, 2013). All of the compound analogs were reported in the same patent application, “Method for altering the lifespan of eukaryotic organisms” [18]. An exact structure search of the Chemical Abstracts database using the SciFinder software for ML370 did not return a hit.

A search of PubChem for CID 665390 on August 06, 2013 (the original hit compound, entry 1 in Table 6) shows that this compound has been evaluated in a total of 732 assays. There are 16 assays unrelated to this project in which CID 665390 showed dose-dependent activity with a reported AC50 below 20 µM. Table 5 lists these assays and the measured activity of CID 665390.

Table 6. Structural Modifications of the Core Region.

Table 6

Structural Modifications of the Core Region.

Table 5. PubChem Assays Reporting Dose-Dependent Activity of CID 665390 (the hit compound).

Table 5

PubChem Assays Reporting Dose-Dependent Activity of CID 665390 (the hit compound).

ML370 (CID 70680248) possesses no obvious chemical liabilities, and a 1:1 acetronitrile:phosphate-buffered saline solution of the probe showed no significant decomposition over a 48-hour period (see Figure 4B). In addition DTT did not react with the probe appreciably over 8 hours (see Figure 4C). See Appendix D for the experimental procedures for the solubility, PBS stability, GSH/DTT stability and plasma stability measurements.

3.4. SAR Tables

The 1H-pyrazolo[3,4-b]quinolone core was one of a small number of cores chosen after analysis of the high throughput data (AID 588436) and subsequent confirmatory analyses (data not shown). One derivative of this structure, N-(1,7-dimethyl-1H-pyrazolo[3,4-b]quinolin-3-yl)furan-2-carboxamide (CID 665390), was the ninth most active compound in the high throughput screen. Confirmatory and related screens were run on this compound (Figure 5A & B) and, given the acceptable results and drug-likeness of the compound, this compound was chosen as a lead structure for a new chemotype.

A search of AID 588436 (350,317 substances tested) for the 1H-pyrazolo[3,4-b]quinoline substructure (SMILES C12=CC=CC=C1C=C3C(NN=C3)=N2) showed that 543 substances with this substructure were tested, of which two were active (CID 665390 and N-(1-ethyl-1H-pyrazolo[3,4-b]quinolin-3-yl)furan-2-carboxamide (CID 663294)). A broader search using the simpler substructure 1H-pyrazolo[3,4-b]pyridine (SMILES C12=NC=CC=C1C=NN2) showed 2,894 substances with this substructure, of which only the same two compounds noted above were active. (The overall hit rate for AID 588436 was 0.16%.) Given the structural similarities between CIDs 665390 and 663294, and the data for CID 665390, we proceeded with CID 665390 as a lead structure.

In order to investigate the activity of the hit compound CID 665390, 20 structurally related analogs were synthesized and evaluated for their ability to initiate the V. cholera QS pathway in the BH1578 V. cholera strain.

The biological assay data of these analogs are presented in Table 6-7. Characterization data (1H and 13C NMR spectra and RP HPLC chromatograms and HRMS data) for these analogs are provided in Appendix F.

Table 7. Structural Modifications of the Acyl Group.

Table 7

Structural Modifications of the Acyl Group.

With respect to structural modifications to the pyrazoloquinoline core, while holding the 2-furoyl amide constant, only entries 5 and 6 (Table 6) gave desirable activity. Truncating the quinoline ring, as in entries 2 through 4 (Table 6) significantly diminished activity. Di-substitution on the far-left portion of the pyrazoloquinoline core uniformly led to diminished activity, as shown for entries 10 through 13 (Table 6). Depending on the nature of the mono-substitution on the far-left portion of the pyrazoloquinoline core, activity was increased or decreased, as seen for entries 6 and 7 through 9 (Table 6), respectively. With the limited number of analogs tested, in this regard, inferring causation is difficult and unwarranted.

In Table 7, structural modifications to the 2-furoyl moiety, while holding the pyrazoloquinoline core constant, uniformly decreased activity for the compounds tested.

Given the above results, entry 6, Table 6, was nominated as the probe compound.

3.5. Cellular Activity

The primary assay and several secondary assays were performed with intact Vibrio cholerae bacteria and a few compounds were observed to be capable of inhibiting an intracellular kinase, LuxO. In addition, compounds were tested in a cytotoxicity screen utilizing mammalian HeLa cells without apparent cytotoxicity. An overview of these assays is provided above in Section 2.1, and full experimental details can be found in Appendix B. The probe ML370 satisfies the cellular activity criteria specified for this project (refer to Section 4, Table 8).

Table 8. Comparison of New & Existing Cholera QS Probes to Project Criteria.

Table 8

Comparison of New & Existing Cholera QS Probes to Project Criteria.

3.6. Profiling Assays

ML370 was submitted to Eurofins Panlabs for evaluation in their LeadProfilingScreen (Catalog No. 68) to determine possible off-target effects against a broad panel of receptors, ion channels, and transporters. The probe compound was determined to be inactive against most of the 68 biological targets, and showed significant activity against three targets, the Adenosine A1 (85% Inh at 10 µM), Adenosine A2A (68% Inh at 10 µM), and Adenosine A3 (86% Inh at 10 µM) receptors. This potential for activity at the adenosine receptors should be kept in mind when contemplating using ML370 in animal studies. The complete listing of the results for the Eurofins Panlabs LeadProfilingScreen for ML370 can be found in Appendix H.

Because its tentative mode of action is ATPase inhibition, ML370 was also tested in Merck Millipore's KinaseProfiler screen for potential cross-reactivity towards a panel of 59 human kinase targets. The probe compound was determined to be inactive against all of the 59 kinase targets. The complete listing of the results for the Merck Millipore KinaseProfiler panel assay for ML370 can be found in Appendix I.

4. Discussion

The MLSMR collection, containing 352,083 unique compounds, was screened for activity against the Vibrio cholerae quorum sensing pathway. After validation of activity using purified dry powders and numerous secondary cell-based assays, one scaffold was advanced for chemical optimization by the Broad Institute and SAR investigation that yielded ML366, which is reported elsewhere. A second LuxO inhibitor scaffold was advanced by the University of Kansas Specialized Chemistry Center to generate probe ML370.

The potent scaffold comprises a pyrazoloquinoline core substituted with a furanyl moiety via an amide bond. Figure 8 depicts the compound hit CID 665390 and summarizes the various modifications explored during SAR studies. While four diversity points were earmarked for examination (highlighted in blue, mauve, green and orange), the regions highlighted in blue and orange were the primary diversity points investigated with a collection of 20 synthetically prepared analogs. As a result of these studies, probe ML370 was identified, wherein the original 7-methyl group substitution on the pyrazoloquinoline core was replaced by a methoxy group.

Figure 8. Summary of analogs prepared to investigate the SAR profile of hit CID 665390.

Figure 8

Summary of analogs prepared to investigate the SAR profile of hit CID 665390.

ML370 activates the quorum sensing pathway of Vibrio cholerae when present at micromolar concentrations (EC50 = 2.5 µM, BH1578). Furthermore, it does not appear to be toxic to HeLa cells and therefore satisfies the desired probe criteria for this project (Table 8).

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

A series of aza-uracil-containing compounds are the only known, reported inhibitors of V. cholerae LuxO [2]. The most potent member of this family, designated Aza-U (cf. Figure 3), was synthetically prepared and used as the positive control for all cell-based assays. ML370 is more potent than Aza-U in the primary assay when compared directly (EC50 = 2.5 µM and > 35 µM, respectively). Consistently across all of the assays, ML370 performed better than Aza-U (Figure 5C, Figure 5D, Table 9). In BH1578, BH1651, and DH231, only the positive control dose of 40 µM induces light production and the lower doses do not generate a consistent dose-dependent response (see Figures 5C & 5D). In the WN1103 assay, ML370 is more potent compared to Aza-U (1.7 to 21 µM, respectively, Table 9).In addition to Aza-U, ML370 was compared to the natural QS ligand, CAI-1, as well as two other small molecule CqsS agonists and the results of this comparison are summarized in Table 9. The discovery and optimization of the CqsS agonist probes, ML343 and ML344, were previously reported by the Broad in late 2012. It is anticipated that the current LuxO probes, ML366 and ML370, will provide valuable information regarding available binding pockets of V. cholera LuxO and guide the development of the next generation of quorum sensing focused therapeutics.

Table 9. Probe Activation of Quorum Sensing in Vibrio cholerae Mutant Strains.

Table 9

Probe Activation of Quorum Sensing in Vibrio cholerae Mutant Strains. Table 9. ML370, positive controls and the previously submitted probes were evaluated in alternative V. cholerae mutants to establish possible modes of actions. The reported EC50 values (more...)

4.2. Mechanism of Action Studies

ML370 was evaluated for its ability to activate the quorum sensing pathways of additional V. cholerae strains (Table 9). Strain BH1651 expresses a constitutively active LuxOD47E mutant (Pubchem AID 651847) as described above in Section 2.1.3. For this particular bacterial strain, LuxO is locked into an active conformation and thus the signaling processes downstream of LuxO are actively repressed. The production of luciferase is repressed independent of CqsS or LuxPQ signaling. The ability of ML370 to promote light production in BH1651 suggests the compound acts upon or downstream of LuxO, consistent with the putative activity of LuxO inhibitors. V. cholerae DH231 and WN1103 are mutant strains lacking CqsS and LuxQ membrane receptors, respectively. If ML370 is working downstream of either CqsS or LuxPQ, the DH231 and WN1103 cells should activate light production with compound treatment. This predicted phenotype is precisely what is observed (Table 9, entries 1 & 4). This activity profile appears to be conserved across both LuxO inhibitor scaffolds (ML370 and ML366); QS agonists like CAI-1, ML343 and ML344 activate V. cholerae QS only if the cells express the CqsS receptor.

Compared to DMSO, ML370 significantly decreases LuxO ATPase activity in vitro (Figure 6C). This suggests that ML370 inhibits LuxO in a similar manner to azaU by directly targeting LuxO itself [2].

4.3. Planned Future Studies

In addition to V. cholerae, there are at least six Vibrio species that cause disease in humans that are contracted through contaminated water or ingested during consumption of seafood [19]. Aza-U inhibits LuxO in multiple Vibrio species, including V. harveyi and V. haemolyticus [2]. ML370 will be tested against these and multiple vibrios for the ability to shutdown virulence factor production. Combinatorial therapies have proven efficacious in a number of infectious disease treatment regimens and could also be applicable in treating cholera. ML370 will be tested in combination with several CqsS agonists and the natural CAI-1 ligand. ML370 and the other probes will also be tested in animal models of cholera. Identification and characterization of the binding site(s) occupied by ML370 will be undertaken. These studies will include analysis with several LuxO mutants in cells and using in vitro kinase and ATPase assays. Other experiments could include structural efforts. This structural information will undoubtedly be beneficial in the development of additional LuxO inhibitors as potential anti-cholera therapeutics. Co-crystallization of LuxO with the probe is the most attractive possibility to obtain structural information, but protein NMR binding studies are also an option. Other methods to determine probe-protein interaction, albeit with less precision, are biophysical experiments to determine direct binding of the compound to the LuxO protein. Differential scanning fluorimetry (DSF) or thermal shift is a means of detecting binding of a ligand to purified protein. This technique is routinely used in our laboratories and could be applied to LuxO.

ML370 will be assessed for pharmacokinetic properties in anticipation of evaluating the therapeutic value of QS agonists in animal models of cholera infection [3-5]. One attribute of a potential cholera therapeutic is that it should be well distributed within the host gut, but systemic exposure is not required. In addition, ML370 could be tested in combination with CqsS agonists as a possible therapeutic regiment. Such a combination therapy would be potent and species selective.

ML370 might benefit from additional chemical optimization investigating substitution on the pyrazolo moiety and bioisosteric replacement of the amide linker (Figure 9).

Figure 9. Possible Directions for Future SAR Investigation of ML370.

Figure 9

Possible Directions for Future SAR Investigation of ML370.

Our recommendations for future SAR expansion around the probe ML370 involve: (1) exploring additional electron-donating and electron-withdrawing groups on the far-left of the pyazoloquinoline core, (2) replacing the amide with the sulfonamide group, or reducing the amide, or N-alkylating the amide or sulfonamide, (3) replacing the furan using bioisosteric groups that may be less prone to metabolism, and (4) exploring a variety of replacements for the N-methyl group. The regions shaded in mauve and green, especially, could benefit from SAR exploration.

5. References

1.
Rombel I, North A, Hwang I, Wyman C, Kustu S. The bacterial enhancer-binding protein NtrC as a molecular machine. Cold Spring Harb Symp Quant Biol. 1998;63:157–66. Review. [PubMed: 10384279]
2.
Ng WL, Perez L, Cong J, Semmelhack MF, Bassler BL. Broad spectrum pro-quorum sensing molecules as inhibitors of virulence in Vibrios. PLOS Pathog. 8(6):e1002767. [PMC free article: PMC3386246] [PubMed: 22761573] [CrossRef]
3.
Dutta NK, Habbu MK. Experimental cholera in infant rabbits: a method for chemotherapeutic investigation. Br J Pharmacol Chemother. 1955 Jun;10(2):153–9. [PMC free article: PMC1509487] [PubMed: 14389652]
4.
Aziz KM, Mohsin AK, Hare WK, Phillips RA. Using the rat as a cholera “model” Nature. 1968 Nov 23;220(5169):814–5. [PubMed: 5725345]
5.
Klose KE. The suckling mouse model of cholera. Trends Microbiol. 2000 Apr;8(4):189–91. Review. [PubMed: 10754579]
6.
Ali M, Lopez AL, You YA, Kim YE, Sah B, Maskery B, Clemens J. The global burden of cholera. Bull World Health Organ. 2012 Mar 1;90(3):209–218A. Epub 2012 Jan 24. [PMC free article: PMC3314202] [PubMed: 22461716] [CrossRef]
7.
Perez LJ, Ng WL, Marano P, Brook K, Bassler BL, Semmelhack MF. Role of the CAI-1 Fatty Acid tail in Casino P, Rubio V, Marina A. The mechanism of signal transduction by two-component systems. Curr Opin Struct Biol. 2010 Dec;20(6):763–71. Epub 2010 Oct 13. Review. [PubMed: 20951027]
8.
Rutherford ST, Bassler BL. Bacterial quorum sensing: Its role in virulence and possibilities for its control. Cold Spring Harb Perspect Med. 2012;2:a012427. [PMC free article: PMC3543102] [PubMed: 23125205]
9.
Miller MB, Skorupski K, Lenz DH, Taylor RK, Bassler BL. Parallel quorum sensing systems converge to regulate virulence in Vibrio cholerae. Cell. 2002 Aug 9;110(3):303–14. [PubMed: 12176318]
10.
Higgins DA, Pomianek ME, Kraml CM, Taylor RK, Semmelhack MF, Bassler BL. The major Vibrio cholerae autoinducer and its role in virulence factor production. Nature. 2007 Dec 6;450(7171):883–6. Epub 2007 Nov 14. [PubMed: 18004304]
11.
Wei Y, Ng WL, Cong J, Bassler BL. Ligand and antagonist driven regulation of the Vibrio cholerae quorum sensing receptor CqsS. Mol Microbiol. 2012 Mar;83(6):1095–108. Epub 2012 Feb 14. [PMC free article: PMC3310172] [PubMed: 22295878] [CrossRef]
12.
Lenz DH, Mok KC, Linney BN, Kulkarni RV, Wingreen NS, Bassler BL. The small chaperone Hfq and multiple small RNAs control quorum sensing in Vibrio harveyi and Vibrio cholerae. Cell. 2004;118:69–82. [PubMed: 15242645]
13.
Studholme DJ, Dixon R. Domain architectures of sigma54-dependent transcriptional activators. J Bacteriol. 2003 Mar;185(6):1757–67. Review. [PMC free article: PMC150144] [PubMed: 12618438]
14.
Zhu J, Mekalanos JJ. Quorum sensing-dependent biofilms enhance colonization in Vibrio cholerae. Dev Cell. 2003 Oct;5(4):647–56. [PubMed: 14536065]
15.
Ng WL, Perez LJ, Wei Y, Kraml C, Semmelhack MF, Bassler BL. Signal production and detection specificity in Vibrio CqsA/CqsS quorum sensing systems. Mol Microbiol. 2011 Mar;79(6):1407–17. Epub 2011 Jan 26. [PMC free article: PMC3285556] [PubMed: 21219472] [CrossRef]
16.
Svenningsen SL, Waters CM, Bassler BL. A negative feedback loop involving small RNAs accelerates Vibrio cholerae's transition out of quorum-sensing mode. Genes Dev. 2008;22:226–238. [PMC free article: PMC2192756] [PubMed: 18198339]
17.
Augustine JK, Atta RN, Ramappa BK, Boodappa C. Propylphosphonic Anhydride (T3P®): A Remarkably Efficient Reagent for the One-Pot Transformation of Aromatic, Heteroaromatic, and Aliphatic Aldehydes to Nitriles. Synlett. 2009;20:3378–3382.
18.
Goldfarb DS. Method using lifespan-altering compounds for altering the lifespan of eukaryotic organisms, and screening for such compounds. US 200901635 A1 20090625. U.S. Pat. Appl. 2009
19.
Shinoda S, Miyoshi SI. Proteases produced by vibrios. Biocontrol Science. 2011;16(1):1–11. [PubMed: 21467624]
20.

Appendix A. Assay Summary Table

Table A1Summary of Completed Assays and AIDs

PubChem
AID
TypeTargetConcentration Range (µM)Samples Tested
588521SummaryNANANA
588346PrimaryV. cholerae BH1578 bioluminescence20350,317
602243PrimaryV. cholerae BH1578 bioluminescence0.024-50623
624270PrimaryV. cholerae BH1578 bioluminescence0.00006-3521
651854PrimaryV. cholerae BH1578 bioluminescence0.00006-3521
651809PrimaryV. cholerae BH1578 bioluminescence0.135-35101
651816PrimaryV. cholerae BH1578 bioluminescence0.0000001-8047
652239PrimaryV. cholerae BH1578 bioluminescence0.0000001-8097
686929PrimaryV. cholerae BH1578 bioluminescence0.0000001-8097
624140SecondaryHeLa mammalian cytotoxicity0.00006-3527
651774SecondaryHeLa mammalian cytotoxicity0.135-35101
651864SecondaryHeLa mammalian cytotoxicity0.0000001-8047
686928SecondaryHeLa mammalian cytotoxicity0.0000001-8097
624254SecondaryV. cholerae BH1651 bioluminescence0.024-50623
624269SecondaryV. cholerae BH1651 bioluminescence0.00006-3521
651847SecondaryV. cholerae BH1651 bioluminescence0.0000001-8047
652289SecondaryV. cholerae BH1651 bioluminescence0.0000001-8097
624281SecondaryV. cholerae DH231 bioluminescence0.00006-3521
651808SecondaryV. cholerae DH231 bioluminescence0.0000001-8047
686931SecondaryV. cholerae DH231 bioluminescence0.0000001-8097
624275SecondaryV. cholerae WN1103 bioluminescence0.00006-3521
651841SecondaryV. cholerae WN1103 bioluminescence0.0000001-8047
686930SecondaryV. cholerae WN1103 bioluminescence0.0000001-8097
652219SecondarySLS353 qrr4:GFP reporter assay0.24-50010
686942SecondarySLS353 qrr4:GFP reporter assay0.02-504
686936SecondaryLuxO in vitro ATPase assay1-1002

Appendix B. Detailed Assay Protocols

V. cholerae BH1578 primary bioluminescence assay (2132-01) (AID 588346, AID 602243, AID 624270, AID 651854, AID 651809, AID 651816, AID 652239, AID 686929)

Materials and Reagents

  • Bacterial strain: BH1578 (V. cholerae ΔcqsA ΔluxS carrying pBB1 cosmid, which contains the V. harveyi luxCDABE luciferase operon)
  • LB Medium: Dissolve in 10 g/L Tryptone, 5 g/L Yeast Extract, and 10 g/L NaCl in distilled water, autoclave, store at room temperature
  • Tetracycline (10 mg/mL): Dissolve 10 mg tetracycline in 1 mL 100% ethanol, store at -20 °C, protect from light.
  • LB/tet: add 1 mL tetracycline (10 mg/mL) to 1 L of LB medium. Final concentration of tetracycline is 10 µg/mL. Make it fresh for every use.
  • CAI-1 stock: Dissolve CAI-1 in DMSO to 50 mM (10.7 mg/mL), store at -20 °C

Procedure

  1. Grow up BH1578 reporter strain in 50 mL LB/Tet at 30 °C for >16 hours with shaking (200 rpm). The final OD600 of each culture should be > 3.0.
  2. Adjust culture to OD600 = 0.3, mix well. (Note: a low speed centrifugation (200 rpm for 1 min) removed most biofilm aggregates).
  3. Add 20 µL LB/tet per well with Thermo Combi MultiDrop fluid dispenser into a 384 well plate. (Greiner black, clear bottom plates were used for HTS while Corning 8867BC white opaque 384 plates were used for subsequent assays).
  4. Pin transfer 100 nL compound or 1 µM CAI-1 as positive control per well from compound source plate to assay plate (pinning volume was 150 nL for the primary HTS).
  5. Dispense 10 µL of diluted culture into each well of a 384 well plate.
  6. Incubate the plates at 30 °C without shaking for 6 hours.
  7. Measure bioluminescence (Lum(384)) and OD600 in a Perkin-Elmer Envision Multilabel Reader

For dose retests, compounds were arrayed so that 2 rows of DMSO were located between each test compound. This minimized any signal from a neighboring well yielding a false positive due to an extremely high bioluminescence signal.

V. cholerae BH1651 secondary LuxO inhibitor bioluminescence assay (2132-03) (AID 624254, AID 624269, AID 651847, AID 652289)

Materials and Reagents

  • Bacterial strain: BH1651 (V. cholerae luxOD47E carrying pBB1 cosmid, which contains the V. harveyi luxCDABE luciferase operon)
  • LB Medium: Dissolve in 10 g/L Tryptone, 5 g/L Yeast Extract, and 10 g/L NaCl in distilled water, autoclave, store at room temperature
  • Tetracycline (10 mg/mL): Dissolve 10 mg tetracycline in 1 mL 100% ethanol, store at -20 °C, protect from light.
  • LB/tet: add 1 mL tetracycline (10 mg/mL) to 1 L of LB medium. Final concentration of tetracycline is 10 µg/mL. Make it fresh for every use.

Procedure

  1. Grow up BH1651 reporter strain in 50 mL LB/Tet at 30 °C for >16 hours with shaking (200 rpm). The final OD600 of each culture should be > 3.0.
  2. Adjust culture to OD600 = 0.3, mix well. (Note: a low speed centrifugation (200 rpm for 1 min) removed most biofilm aggregates).
  3. Add 20 µL LB/tet per well with Thermo Combi MultiDrop fluid dispenser into a 384 well plate. (Greiner black, clear bottom plates were used for HTS while Corning 8867BC white opaque 384 plates were used for subsequent assays).
  4. Pin transfer 100 nL compound per well from compound source plate to assay plate.
  5. Dispense 10 µL of diluted culture into each well of a 384 well plate.
  6. Incubate the plates at 30 °C without shaking for 6 hours.
  7. Measure bioluminescence (Lum(384)) and OD600 in a Perkin-Elmer Envision Multilabel Reader

For dose retests, compounds were arrayed so that 2 rows of DMSO were located between each test compound. This minimized any signal from a neighboring well yielding a false positive due to an extremely high bioluminescence signal.

V. cholerae DH231 secondary sensor mechanism bioluminescence assay (2132-04) (AID 624281, AID 651808, AID 686931)

Materials and Reagents

  • Bacterial strain: DH231 (V. cholerae ΔcqsS ΔluxS carrying pBB1 cosmid, which contains the V. harveyi luxCDABE luciferase operon)
  • LB Medium: Dissolve in 10 g/L Tryptone, 5 g/L Yeast Extract, and 10 g/L NaCl in distilled water, autoclave, store at room temperature
  • Tetracycline (10 mg/mL): Dissolve 10 mg tetracycline in 1 mL 100% ethanol, store at -20 °C, protect from light.
  • LB/tet: add 1 mL tetracycline (10 mg/mL) to 1 L of LB medium. Final concentration of tetracycline is 10 µg/mL. Make it fresh for every use.
  • CAI-1 stock: Dissolve CAI-1 in DMSO to 50 mM (10.7 mg/mL), store at -20 °C

Procedure

  1. Grow up reporter strain in 50 mL LB/Tet at 30 °C for >16 hours with shaking (200 rpm). The final OD600 of each culture should be > 3.0.
  2. Adjust culture to OD600 = 0.3, mix well. (Note: a low speed centrifugation (200 rpm for 1 min) removed most biofilm aggregates).
  3. Add 20 µL LB/tet per well with Thermo Combi MultiDrop fluid dispenser into a 384 well plate. (Corning 8867BC white opaque 384 plates were used).
  4. Pin transfer 100 nL compound per well or 1 µM CAI-1 as positive control from compound source plate to assay plate.
  5. Dispense 10 µL of diluted culture into each well of a 384 well plate.
  6. Incubate the plates at 30 °C without shaking for 6 hours.
  7. Measure bioluminescence (Lum(384)) and OD600 in a Perkin-Elmer Envision Multilabel Reader

For dose retests, compounds were arrayed so that 2 rows of DMSO were located between each test compound. This minimized any signal from a neighboring well yielding a false positive due to an extremely high bioluminescence signal.

V. cholerae WN1103 Secondary sensor mechanism bioluminescence assay (2132-05) (AID 624275, AID 651841, AID 686930)

Materials and Reagents

  • Bacterial strain: WN1103 (V. cholerae ΔcqsA ΔluxQ carrying pBB1 cosmid, which contains the V. harveyi luxCDABE luciferase operon)
  • LB Medium: Dissolve in 10 g/L Tryptone, 5 g/L Yeast Extract, and 10 g/L NaCl in distilled water, autoclave, store at room temperature
  • Tetracycline (10 mg/mL): Dissolve 10 mg tetracycline in 1 mL 100% ethanol, store at -20 °C, protect from light.
  • LB/tet: add 1 mL tetracycline (10 mg/mL) to 1 L of LB medium. Final concentration of tetracycline is 10 µg/mL. Make it fresh for every use.
  • CAI-1 stock: Dissolve CAI-1 in DMSO to 50 mM (10.7 mg/mL), store at -20 °C

Procedure

  1. Grow up reporter strain in 50 mL LB/Tet at 30 °C for >16 hours with shaking (200 rpm). The final OD600 of each culture should be > 3.0.
  2. Adjust culture to OD600 = 0.3, mix well. (Note: a low speed centrifugation (200 rpm for 1 min) removed most biofilm aggregates).
  3. Add 20 µL LB/tet per well with Thermo Combi MultiDrop fluid dispenser into a 384 well plate. (Corning white opaque 8867BC 384 plates were used).
  4. Pin transfer 100 nL compound or 1 µM CAI-1 as positive control per well from compound source plate to assay plate.
  5. Dispense 10 µL of diluted culture into each well of a 384 well plate.
  6. Incubate the plates at 30 °C without shaking for 6 hours.
  7. Measure bioluminescence (Lum(384)) and OD600 in a Perkin-Elmer Envision Multilabel Reader

For dose retests, compounds were arrayed so that 2 rows of DMSO were located between each test compound. This minimized any signal from a neighboring well yielding a false positive due to an extremely high bioluminescence signal.

V. cholerae SLS353 secondary qrr4:GFP fluorescence reporter assay (2132-06) (AID 652219, AID 686942)

Materials and Reagents

  • Bacterial strain: SLS353 (V. cholerae luxOD47E carrying qrr4:GFP plasmid)
  • LB Medium: Dissolve in 10 g/L Tryptone, 5 g/L Yeast Extract, and 10 g/L NaCl in distilled water, autoclave, store at room temperature
  • Kanamycin (100 mg/mL): Dissolve 100 mg kanamycin in 1 mL water, store at -20 °C
  • LB/Kan: add 1 mL Kanamycin (100 mg/mL) to 1 L of LB medium. Final concentration of Kanamycin is 100 µg/mL. Make it fresh for every use.

Procedure

8.

Grow up strain in 50 mL LB/Kan at 30 °C for >16 hours with shaking (200 rpm). The final OD600 of each culture should be > 3.0.

9.

Adjust culture to OD600 = 0.3, mix well. (Note: a low speed centrifugation (200 rpm for 1 min) removed most biofilm aggregates).

10.

Add 20 µL LB/Kan per well with Thermo Combi MultiDrop fluid dispenser into a 384 well plate. (Corning black opaque 384 plates were used).

11.

Pin transfer 100 nL compound per well from compound source plate to assay plate.

12.

Dispense 10 µL of diluted SLS353 culture into each well of a 384 well plate.

13.

Incubate the plates at 30 °C without shaking for 16 hours.

14.

Measure GFP fluorescence in a Perkin-Elmer Envision Multilabel Reader

LuxO in vitro ATPase assay (AID 686936)

A modified coupled-enzyme assay was used to measure the rate of ATP hydrolysis by LuxOD47E. Briefly, ADP released from ATP by LuxO D47E is reacted with phosphoenolpyruvate (PEP) to form pyruvate using pyruvate kinase (PK). Pyruvate is reacted with NADH to form NAD and lactate using lactate dehydrogenase (LDH). The rate of NAD production is followed at 340 nm using a spectrophotometer. The rates of ATP hydrolysis by LuxOD47E were measured in reactions containing 100 mM sodium phosphate buffer pH 7.4, 5 mM MgCl2, 0.2 mM NADH, 1 mM PEP, 5–20 units of PK/LDH mix (Sigma), and 10 µM LuxOD47E. ATP was added at a final concentration of 2.5 mM. The rate of ATP hydrolysis was monitored for 5 minutes. The values reported are ΔOD340/min.

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’ or the ‘Neutral Controls’ method (when no positive control was available) in GeneData Screener Assay Analyzer (v7.0.3 & v10.0.2). 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 Probe ML370

General Details: All solvents and reagents were used as received from commercial suppliers. The 1H NMR spectra were recorded on a 400 MHz Bruker Avance spectrometer equipped with a broadband observe probe or a 500 MHz Bruker AVIII spectrometer equipped with a dual cryoprobe. The 13C NMR spectra were recorded on a 500 MHz Bruker AVIII spectrometer equipped with a dual cryoprobe (at 125 MHz). Microwave reactions were carried out in a Biotage Initiator instrument. Column chromatography separations were performed using the Teledyne Isco CombiFlash Rf using RediSep Rf silica gel or RediSep Rf C18 High Performance Gold columns. The analytical RPLC method used an Agilent 1200 RRLC system with UV detection (Agilent 1200 DAD SL) and mass detection (Agilent 6224 TOF). The analytical method conditions included a Waters Aquity BEH C18 column (2.\1 × \2 mm, 1.7 µm) and elution with a linear gradient of 5% acetonitrile in pH 9.8 buffered aqueous ammonium formate to 100% acetonitrile at 0.4 mL/min flow rate. Compound purity was measured on the basis of peak integration (area under the curve) from UV-vis absorbance at 214 nm, and compound identity was determined on the basis of mass spectral and NMR analyses. All compounds used for biological studies have purities of ≥ 93%.

Image ml370f12

N-(3-Methoxyphenyl)acetamide (KU2): A solution of m-anisidine (616 mg; 5.00 mmol) in acetic acid (4.0 mL) was reacted under microwave irradiation at 250 °C for 3 min. Water (40 mL) was added and the solution was then basified with 10 M NaOH (ca. 7.0 mL). The crude product was extracted into DCM (3 × 5 mL) and this solution was dried over MgSO4. Pure product was obtained as a tan solid (779 mg; 94%) after column chromatography (SiO2 / gradient of 0-7% methanol in DCM). 1H NMR (400 MHz, CDCl3) δ 7.30 – 7.16 (m, 3H), 6.96 (d, J = 7.9 Hz, 1H), 6.66 (dd, J = 8.2, 2.0 Hz, 1H), 3.80 (s, 3H), 2.17 (s, 3H).

2-Chloro-7-methoxyquinoline-3-carbaldehyde (KU3): Phosphoryl trichloride (4.12 mL, 44.2 mmol) was added dropwise to DMF (1.21 ml, 15.6 mmol) whilst maintaining the temperature at 0-5 °C. The mixture was allowed to stir for about 5 min. N-(3-methoxyphenyl)acetamide (771 mg, 4.67 mmol) was then added and the resulting solution heated for 8 hr at 80 °C. The reaction mixture was cooled to room temperature and then poured into crushed ice with stirring. A pale yellow precipitate appeared immediately and was filtered and washed with water and then dried. The crude product was purified by column chromatography (SiO2 / gradient of 0 - 5% MeOH in DCM) to afford 2-chloro-7-methoxyquinoline-3-carbaldehyde (607 mg, 59% yield) as a pale yellow solid. 1H NMR (400 MHz, CDCl3) δ 10.51 (s, 1H), 8.66 (s, 1H), 7.85 (d, J = 9.0 Hz, 1H), 7.38 (d, J = 2.4 Hz, 1H), 7.27 (dd, J = 9.0, 2.5 Hz, 1H), 3.99 (s, 3H).

2-Chloro-7-methoxyquinoline-3-carbonitrile (KU4): 1-Propanephosphonic anhydride [17] (50% in DMF; 1.69 mL, 2.86 mmol) was added to a mixture of 2-chloro-7-methoxyquinoline-3-carbaldehyde (577 mg, 2.60 mmol), hydroxylamine hydrochloride (199 mg, 2.86 mmol) and triethylamine (0.40 mL, 2.9 mmol) in DMF (3.0 mL); then the mixture was stirred at 100 °C for 2 hrs. The completion of the reaction was confirmed by TLC (SiO2 / 5% EtOAc in hexane). The mixture was then cooled and carefully poured onto sat. aq NaHCO3 (15 mL) and extracted with DCM (2 × 10 mL). The combined organic phase was washed with water (10 mL) and brine (10 mL) and dried (MgSO4), filtered and the solvent was removed under reduced pressure to afford the crude product which was purified by column chromatography (SiO2 / gradient of 0 - 10% EtOAc in hexanes) yielding 2-chloro-7-methoxyquinoline-3-carbonitrile (285 mg, 50% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.43 (s, 1H), 7.76 (d, J = 9.0 Hz, 1H), 7.37 (d, J = 2.4 Hz, 1H), 7.31 (dd, J = 9.0, 2.5 Hz, 1H), 3.99 (s, 3H).

7-Methoxy-1-methyl-1H-pyrazolo[3,4-b]quinolin-3-amine (KU5): Methylhydrazine (0.338 mL, 6.41 mmol) was added to a stirred suspension of 2-chloro-7-methoxyquinoline-3-carbonitrile (275 mg, 1.26 mmol) in EtOH (7.5 mL). The mixture was stirred at reflux for 2 hrs and then cooled to 0 °C. Water (12 mL) was added followed by DCM (2 × 35 mL). The combined organic phase was dried (MgSO4), filtered and the solvent was removed under reduced pressure to afford the crude product which was purified by column chromatography (SiO2 / gradient of 0 - 5% MeOH in DCM) yielding 7-methoxy-1-methyl-1H-pyrazolo[3,4-b]quinolin-3-amine (208 mg, 0.911 mmol, 73% yield) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.30 (s, 1H), 7.75 (d, J = 9.1 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 7.04 (dd, J = 9.1, 2.5 Hz, 1H), 4.26 (s, 2H), 3.99 (s, 3H), 3.98 (s, 3H).

N-(7-Methoxy-1-methyl-1H-pyrazolo[3,4-b]quinolin-3-yl)furan-2-carboxamide (ML370): 2-Furoyl chloride (0.087 mL, 0.89 mmol) was added to a solution of 7-methoxy-1-methyl-1H-pyrazolo[3,4-b]quinolin-3-amine (202 mg, 0.885 mmol) and triethylamine (0.247 mL, 1.77 mmol) in DCM (5 mL) at 0 °C. The reaction was allowed to warm to room temperature and left to stir overnight. Sat. NaHCO3 (10 mL) was added and this was extracted with DCM (2 × 10 mL). The combined organic phase was washed with brine (15 mL) and dried (MgSO4), filtered and the solvent was removed under reduced pressure to afford the crude product which was purified by column chromatography (SiO2 / gradient of 0 - 50% EtOAc in hexanes) yielding N-(7-methoxy-1-methyl-1H-pyrazolo[3,4-b]quinolin-3-yl)furan-2-carboxamide (104 mg, 0.323 mmol, 37% yield) as a yellow solid. 1H NMR (500 MHz, CDCl3) δ 9.24 (s, 1H), 8.92 (s, 1H), 7.86 (d, J = 9.1 Hz, 1H), 7.59 (dd, J = 1.8, 0.8 Hz, 1H), 7.36 (dd, J = 3.5, 0.8 Hz, 1H), 7.31 (d, J = 2.6 Hz, 1H), 7.08 (dd, J = 9.1, 2.5 Hz, 1H), 6.63 (dd, J = 3.5, 1.7 Hz, 1H), 4.13 (s, 3H), 3.99 (s, 3H); 13C NMR (126 MHz, CDCl3) δ 162.39, 155.32, 150.97, 150.94, 147.08, 144.98, 138.14, 135.22, 131.17, 120.02, 118.11, 116.11, 112.81, 108.05, 104.51, 55.59, 33.62; HRMS (ESI+) m/z calcd for [C17H14N4O3 + H]+: 323.1139, found: 323.1170. UPLC purity at 214 nm is 96.1%.

Appendix D. Experimental Procedure for Additional Analytical Assays

PBS 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.

LB Solubility. Solubility was determined as described for PBS solubility. LB media was prepared as described above and substituted for phosphate buffered saline pH 7.4.

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, methanol was added to force dissolution of compound (to test for recovery of compound). An aliquot of this was also analyzed by UPLC-MS.

GSH Stability. Stability was determined in the presence of PBS pH 7.4 µM and 50 µM glutathione 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 6 hours. One plate was removed at each time point (0, 0.5, 1, 2, 4, and 6 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, methanol was added to force dissolution of compound (to test for recovery of compound). An aliquot of this was also analyzed by UPLC-MS.

PBS Stability (KU): Compound was dissolved at 10 µM in PBS/acetonitrile (1/1) at pH 7.4 (1% DMSO) and incubated at room temperature. The mixtures were sampled every hour for eight hours or every 8 hours for 48 hours and analyzed by RP HPLC/UV/HRMS. The analytical RP HPLCUV/HRMS system utilized for the analysis was a Waters Acquity system with UV-detection and mass-detection (Waters LCT Premier). The analytical method conditions included a Waters Acquity HSS T3 C18 column (2.\1 × \2mm, 1.8um) and elution with a linear gradient of 99% water to 100% CH3CN at 0.6 mL/min flow rate. Peaks on chromatograms were integrated using the Waters OpenLynx software. Absolute areas under the curve (214 nm) were compared at each time point to determine relative percent compound remaining in supernatant.

DTT Stability (KU): Compound was dissolved at 10 µM in PBS/acetonitrile (1/1) at pH 7.4 (1% DMSO) and incubated at room temperature with either no thiol source as a negative control or 50 µM dithiothreitol (DTT). The mixtures were sampled every hour for eight hours or every 8 hours for 48 hours and analyzed by RP HPLC/UV/HRMS. The analytical RP HPLCUV/HRMS system utilized for the analysis was a Waters Acquity system with UV-detection and mass-detection (Waters LCT Premier). The analytical method conditions included a Waters Acquity HSS T3 C18 column (2.\1 × \2mm, 1.8um) and elution with a linear gradient of 99% water to 100% CH3CN at 0.6 mL/min flow rate. Peaks on chromatograms were integrated using the Waters OpenLynx software. Absolute areas under the curve (214 nm) were compared at each time point to determine relative percent compound remaining in supernatant. The masses of potential adducts were searched for in the samples to determine if any detectable adduct formed. All samples were prepared in duplicate. Ethacrynic acid, a known Michael acceptor, was used as a positive control and was tested in PBS/acetonitrile (1/1).

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 37 ºC 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 37 ºC 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 37 ºC 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 for the Probe

Probe CID 70680248; SID 160655705

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3).

13C NMR Spectrum (126 MHz, CDCl3)

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H14N4O3 + H]+: 323.1139, found: 323.1170.

Appendix F. Chemical Characterization Data for All Analogs

Analog CID 665390; SID 160655702

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.34, 150.76, 149.19, 147.08, 144.98, 141.84, 137.97, 135.12, 129.66, 126.39, 126.11, 122.46, 116.12, 112.81, 109.17, 33.62, 22.25.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H14N4O2 + H]+: 307.1190, found: 307.1234.

Analog CID 70680249; SID 160655703

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.83, 155.39, 150.78, 149.18, 145.38, 141.82, 138.29, 135.38, 129.70, 126.35, 126.04, 122.44, 117.43, 109.31, 109.17, 33.60, 22.25, 13.94.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C18H16N4O2 + H]+: 321.1346, found: 321.1357.

Analog CID 1385812; SID 160655704

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 159.30, 150.75, 149.22, 141.89, 138.40, 137.93, 135.13, 131.74, 129.66, 129.43, 128.15, 126.38, 126.15, 122.48, 109.18, 77.27, 77.01, 76.76, 33.59, 22.25.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H14N4OS + H]+: 323.0961, found: 323.0984.

Analog CID 70680252; SID 160655706

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.37, 148.82, 147.03, 145.02, 138.00, 135.61, 131.19, 130.11, 127.74, 124.13, 123.41, 116.20, 112.84, 109.76, 77.26, 77.22, 77.01, 76.76, 33.69.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C16H12N4O2 + H]+: 293.1033, found: 293.1048.

Analog CID 70680251; SID 160655707

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.35, 150.75, 149.31, 147.97, 147.08, 144.98, 137.99, 135.17, 129.76, 125.23, 124.90, 122.67, 116.13, 112.82, 109.20, 77.27, 77.01, 33.64, 29.38, 14.84.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C18H16N4O2 + H]+: 321.1346, found: 321.1360.

Analog CID 70680254; SID 160655708

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.72, 147.44, 144.60, 141.39, 138.14, 127.07, 123.36, 120.23, 116.55, 115.67, 112.58, 108.75, 35.34.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C13H11N3O2 + H]+: 242.0924, found: 242.0942.

Analog CID 19405046; SID 160655709

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.27, 147.50, 146.21, 144.36, 130.97, 115.20, 112.47, 97.40, 77.28, 77.22, 77.02, 76.77, 38.82.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C9H9N3O2 + H]+: 192.0768, found: 192.0766.

Analog CID 70680250; SID 160655710

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.35, 150.70, 149.65, 147.10, 144.85, 137.48, 134.13, 116.32, 115.97, 112.72, 108.22, 77.27, 77.22, 77.02, 76.76, 33.58.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C12H10N4O2 + H]+: 243.0877, found: 243.0914.

Analog CID 70680253; SID 160655711

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 159.94, 150.77, 149.23, 145.80, 144.43, 141.91, 138.35, 135.07, 129.65, 126.39, 126.16, 122.47, 121.97, 109.24, 108.43, 77.26, 77.21, 77.01, 76.75, 33.60, 22.25.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H14N4O2 + H]+: 307.1190, found: 307.1198.

Analog CID 70680247; SID 160655712

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 174.17, 150.74, 149.15, 141.76, 138.86, 135.18, 129.65, 126.33, 125.98, 122.34, 109.25, 45.98, 33.51, 30.58, 26.09, 22.23.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C18H20N4O + H]+: 309.1710, found: 309.1717.

Analog CID 1400696; SID 160655713

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 161.77, 150.78, 149.14, 149.00, 148.34, 141.73, 138.35, 137.72, 135.03, 129.67, 126.84, 126.42, 126.02, 122.46, 122.38, 109.31, 33.65, 22.24.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C18H15N5O + H]+: 318.1349, found: 318.1250.

Analog CID 70698449; SID 160778928

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 160.18, 158.14, 155.34, 146.94, 145.03, 138.61, 122.04, 116.36, 113.93, 113.79, 112.88, 109.00, 108.85, 33.86, 22.52, 22.51.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H13FN4O2 + H]+: 325.1095, found: 325.1093.

Analog CID 70698444; SID 160778929

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.36, 147.01, 146.11, 145.02, 137.52, 134.11, 134.06, 129.27, 129.22, 116.20, 112.85, 111.24, 111.06, 109.52, 33.63, 16.08, 16.05.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H13FN4O2 + H]+: 325.1095, found: 325.1131.

Analog CID 70698450; SID 160778930

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.87, 155.07, 150.76, 149.23, 147.70, 141.91, 137.82, 135.13, 129.68, 127.66, 127.57, 126.43, 126.19, 124.08, 122.96, 122.50, 112.15, 111.98, 109.07, 33.68, 22.26.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C21H16N4O2 + H]+: 357.1346, found: 357.1347.

Analog CID 70698445; SID 160778931

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.32, 150.53, 148.23, 147.14, 144.93, 142.20, 137.79, 133.97, 133.34, 128.66, 126.78, 123.09, 116.04, 112.78, 109.17, 33.62, 20.82, 19.94.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C18H16N4O2 + H]+: 321.1346, found: 321.1351.

Analog CID 70698443; SID 160778932

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 164.98, 162.98, 155.32, 150.75, 150.51, 150.40, 147.00, 145.01, 139.96, 139.87, 138.38, 132.31, 121.12, 116.18, 115.32, 115.11, 112.83, 108.89, 108.73, 108.70, 33.59, 19.65, 19.64.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C17H13FN4O2 + H]+: 325.1095, found: 325.1134.

Analog CID 70698446; SID 160778933

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 166.35, 149.15, 147.63, 143.14, 141.84, 138.08, 134.79, 129.61, 126.38, 126.12, 122.39, 111.05, 109.47, 109.17, 36.66, 33.53, 22.24.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C18H16N4O2 + H]+: 321.1346, found: 321.1379.

Analog CID 70698448; SID 160778934

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.34, 152.52, 150.77, 149.34, 147.08, 144.98, 138.00, 135.16, 129.81, 124.12, 123.44, 122.80, 116.12, 112.82, 109.24, 34.56, 33.65, 23.54.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C19H18N4O2 + H]+: 335.1503, found: 335.1519.

Analog CID 70698447; SID 160778935

1H NMR Spectrum (500 MHz, CDCl3).

1H NMR Spectrum (500 MHz, CDCl3)

13C NMR Spectrum (126 MHz, CDCl3) δ 155.33, 154.71, 150.81, 149.21, 147.08, 144.99, 137.99, 134.95, 129.47, 122.88, 122.55, 122.38, 116.13, 112.82, 109.34, 35.43, 33.69, 30.96.

UPLC-MS Chromatogram.

UPLC-MS Chromatogram

HRMS calcd for [C20H20N4O2 + H]+: 349.1659, found: 349.1675.

Appendix G. Prior Art Search

Investigation into relevant prior art entailed searching the following databases: SciFinder, Reaxys, PubChem, PubMed, US Patent and Trademark Office (USPTO PatFT and AppFT), World Intellectual Property Organization (WIPO), and Thomson Reuters Integrity. The search terms applied and hit statistics are provided in Table A2. As indicated below, abstracts were obtained for the references returned and were analyzed for relevance to the current project. The searches were originally performed on May 9, 2011. Searches were performed again and updated on April 2, 2013.

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

Table A2

Search Strings and Databases Employed in Prior Art Search.

Appendix H. Eurofins Panlabs LeadProfiling Screen Report for ML370

A sample of the probe compound ML370 (CID 70680248, SID 160655705, KUC112612N, KSC-329-029-1) was submitted to Eurofins Panlabs for screening across a panel of 68 receptors.

Study Objective

To evaluate, in radioligand binding assays, the activity of probe compound ML370 (KUC112612N) across a panel of 68 receptors.

Methods

Methods employed in this study performed at Eurofins Panlabs [20] have been adapted from the scientific literature to maximize reliability and reproducibility. Reference standards were run as an integral part of each assay to ensure the validity of the results obtained. Assay results are presented as the percent inhibition of specific binding or activity (for n = 2 replicates) for the probe compound tested at a concentration of 10 µM.

Table A3. Results for ML370 for the Eurofins Panlabs LeadProfilingScreen.

Table A3Results for ML370 for the Eurofins Panlabs LeadProfilingScreen

Appendix I. Merck Millipore Kinase Report for ML370

A sample of the probe compound ML370 (CID 70680248, SID 163686507, KUC112612N-02, KSC-329-108-1) was submitted to Merck Millipore for screening across a panel of 59 kinases.

The following text is adapted from the Merck Millipore report that accompanied the kinase profiling data.

Assay Formats

Protein kinases are tested in a radiometric assay format and the raw data are measured by scintillation counting (in cpm). Lipid kinases are measured in an HTRF® format, with fluorescence measured at two wavelengths and raw data subsequently expressed as HTRF ratio.

Controls and Blanks

Positive control wells contain all components of the reaction except the compound of interest; however, DMSO is included in these wells to control for solvent effects. For protein kinase assays, blank wells contain all components of the reaction with a reference inhibitor replacing the compound of interest. This abolishes kinase activity and establishes the base line (0% kinase activity remaining). For lipid kinase assays, the 0% activity base

% Activity Values

In the graph, results are expressed as a percentage of the mean kinase activity in the positive control samples. The positive control value is considered to be 100%, and all test samples are measured in relation to this value. For example, a result of 42% means that, in comparison to the positive control, 42% kinase activity remains in the presence of the test compound. Expressed another way, the test compound inhibits the kinase activity by 58%.

Table A4. Merck Millipore Kinase Report for ML370.

Table A4Merck Millipore Kinase Report for ML370

Appendix J. Compounds submitted to Evotec

Table A5Probe and Analog Information

BRD/KU IDSIDCIDP/AMLSIDML
BRD-K33563922-001-01-6

KUC112612N
16065570570680248PMLS004820389ML370
BRD-K03587580-001-01-4

KUC112617N
16065571070680250AMLS004820392-
BRD-K71786710-001-01-2

KUC112615N
16065570870680254AMLS004820391-
BRD-K08510569-001-01-6

KUC112613N
16065570670680252AMLS004820390-
BRD-K64513287-001-01-2

KUC112614N
16065570770680251AMLS004820394-
BRD-K86942225-001-01-2
KUC112619N
16065571270680247AMLS004820393-

A = analog; P = probe

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this page (2.8M)

Related information

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...