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

ML322, A Small Molecule Inhibitor of Dimerization of the Core Protein of Hepatitis C Virus (HCV)

, , , , , , , , , and .

Author Information and Affiliations

Received: ; Last Update: May 13, 2014.

Over three million Americans are infected by the Hepatits C Virus (HCV) virus 2 which is particularly lethal for Acquired Immunodeficiency Syndrome (AIDS) patients, of whom increasing numbers are co-infected with Human Immunodeficiency Virus infection (HIV) and HCV. There is no vaccine, and the only available treatment, based on a combination of interferon and ribavirin, cures less than half of the patients. One of the important structural proteins of HCV is the core protein. HCV core has many reported functions in the host cells, but its main purpose for the virus is to enclose and thereby protect the HCV Ribonucleic acid (RNA) genome during disease transmission. The conserved nature of HCV and absence of a vaccine, along with studies demonstrating that core contributes to host cell oncogenesis, apoptosis inhibition, and suppression of host T cell responses, support a role for core as a major pathogenic component of HCV. As a result, the identification of specific inhibitors of HCV core dimerization may provide tools for inhibiting HCV assembly without host cell effects. Several studies identify HCV capsid assembly as an appropriate target for chemotherapeutic intervention of HCV infections. Thus, this High Throughput Screen (HTS) program was initiated to identify small molecules that inhibit HCV core protein dimerization. Following an HTS effort and several rounds of medicinal chemistry, we have identified compound CID 49800087 as inhibitor probe (ML322) belonging to the [1,2,4]triazolo[1,5-a]pyrimidin-2-yl scaffold. The probe inhibits dimerization of core proteins in biochemical Alphascreen assays (IC50 = 8.4 μM; 51% inhibition at 15 μM). Using quantitative polymerase chain reactions (QPCR) the probe was shown to reduce levels of total HCV RNA in infected cells (IC50 = 1.8 μM) and significantly reduces levels of infectious HCV RNA in cells treated with HCV-containing culture supernatants (IC50 = 2 nM). Profiling revealed that probe ML322 lacks activity against a panel of 63 receptors, transporters, and ion channels (Ricerca). Modest activity was detected at muscarinic M1-3, adrenergic alpha, and dopamine transporter (DAT). Critically, probe ML322 is not cytotoxic against the Huh7.5 liver cell line used for the infectivity assays (CC50 = 33 μM). This novel probe represents an exciting tool for use in basic HCV research to elucidate the mechanisms by which core-core dimerization contributes to HCV nucleocapsid formation, HCV infection, and liver cancers associated with HCV.

Assigned Assay Grant #: 1-X01-MH085709-01

Screening Center Name & PI: The Scripps Research Institute Molecular Screening Center (SRIMSC), Hugh Rosen

Chemistry Center Name & PI: SRIMSC, Hugh Rosen

Assay Submitter & Institution: A. Donny Strosberg, Scripps Florida

PubChem Summary Bioassay Identifier: AID 1911

Probe Structure & Characteristics

Probe ML322.

Probe ML322

CID
ML#
SIDs
Target NameIC50 [SID, AID]Anti-TargetIC50 [SID, AID]Fold SelectiveAdditional Assay(s) Name:
[SID, AID]
CID 49800087
ML322; SR-03000001966

SID 103771539 (synth) Probe;

SID 125311267 (re-synth)
HCV Core InfectivityTCID50 = 2 nM
[SID 103771539, AID 651575]
Liver Cell CytotoxicityCC50 =33.79 μM
[SID 103771539, AID 651583]
>16,000-foldT1/T2 QPCR IC50 =1.8 μM
[SID 103771539, AID 651574]

IC50 = 8.4 μM
[SID 103771539, AID 651577]
(Alphascreen)

Ricerca Selectivity Profiling [SID 103771539, AID 624406]
Inactive at 63 Receptors, Transporters, and Channels.
Active at 4: Muscarinic M3, Adrenergic a1D & a1B, and Dopamine Transporter (DAT).

Fold-selectivity was calculated as IC50 for anti-target/IC50 for the target, both cell-based assays

1. Introduction

The Hepatitis C virus (HCV) is a major cause of liver failure and hepatocellular cancer, with about 170 million people infected worldwide [1]. In the United States over three million individuals are infected with the HCV virus 2, which is particularly lethal for AIDS patients, of whom increasing numbers are co-infected with HIV and HCV3 [2]. There is currently no vaccine available [3, 4], and the only available treatment, based on a combination of pegylated interferon and ribavirin, cures less than half of the patients.

The HCV has a small RNA genome that is directly translated by the infected host cell into a single precursor polyprotein that is processed by enzymatic cleavage into 10 proteins of diverse function. The most N-terminal 21kDa protein of this HCV polyprotein is the HCV core (C) protein, which is a highly basic, RNA-binding structural protein essential for assembly and packaging of the viral genome [5]. Core protein is cleaved by a host peptidase and anchored to the host cell endoplasmic reticulum, where it undergoes further processing into its mature form [6]. The N terminal portion of this mature C protein mediates viral assembly through homodimerization and formation of higher order complexes with viral RNA to form the nucleocapsid, while the hydrophobic C terminal interacts with envelope glycoproteins to form the infectious particle [7].

Several drugs are under clinical investigation (Phase 2 or 3) as anti-viral therapy for HCV. These include inhibitors of NS5B polymerase, NS3/NS4A protease, and NS5A [3]. However, potent, low nanomolar inhibitors of HCV infection based on the inhibition of core protein dimerization are unavailable. Thus, any small molecule probe identified will serve as an important step in the development of lead compounds for novel HCV therapies. The novel probe reported here, ML322, exhibits an IC50 of 1.8 μM (T2 QPCR) for blocking the levels of total HCV RNA in cells. In addition the probe potently reduces the levels of infections HCV RNA in cells (TCID50 = 2 nM). All sets of biochemical and cellular assay data were highly reproducible between batches and runs. Importantly, ML322 also consistently lacks activity in the cytotoxicity counterscreen (CC50 >30 μM), demonstrating that it does not significantly affect liver cell viability. Probe ML322 lacks significant inhibitory activity effects against the majority of therapeutic targets tested (Ricerca screening). Together these results demonstrate that probe ML322 far surpasses the probe criteria set forth in the chemical probe development plan.

2. Materials and Methods

All chemical reagents and solvents were acquired from commercial vendors. All assay protocols are reported in the relevant PubChem AIDs, provided in Table 1. Solubility, stability, and glutathione reactivity analyses were conducted in accordance with NIH guidelines. CYP450 inhibition and microsome stability analyses were performed as previously described [8].

Table 1. HCV Core Inhibitors Project (see also Summary AID 1911).

Table 1

HCV Core Inhibitors Project (see also Summary AID 1911).

2.1. Assays

Table 1 lists all PubChem AIDs for the HCV Core Inhibitor project. Descriptions of the assays follow the table.

Primary Assay

2.1a. HCV Core Primary Assays (see AID 1899 (1X%INH), AID 2152 (3X %INH), AID 2159(IC50), and AID 2488 (powders, assay provider’s lab)

The purpose of this biochemical assay is to determine the ability of test compounds to prevent dimerization of the hydrophilic N-terminal domain (residues 1–106; core106) of the HCV core protein (9). In this assay, test compounds are incubated with N-terminally tagged GST-core106 and Flag-core106 peptides, followed by addition of a Europium cryptate-tagged anti-GST antibody and a XL-665-tagged anti-Flag antibody. Dimerization of the core106 peptides and subsequent antibody binding brings the antibody tags into close proximity, allowing FRET from the Europium donor to the XL-665 acceptor, resulting in an increase in well FRET. As designed, compounds that inhibit core106 dimerization will prevent the interaction of the tagged antibodies, blocking the transfer of energy from Europium to XL-665, and thus inhibiting well FRET. Test compounds were assayed in singlicate (AID 1899) and triplicate (AID 2152) at a final nominal concentration of 5 micromolar, in quadruplicate in a 10-point 1:3 dilution series starting at a nominal test concentration of 50 micromolar (Round 0: AID 2159, Round 1: AID 2488).

Prior to the start of the assay, 5 microliters of Assay Buffer (100 mM HEPES, 0.5 mM EDTA, 2.5 mM DTT, 200 mM Potassium Fluoride, 0.05% CHAPS, 0.05% BSA, pH 7.45, filtered at 0.22 micrometer) were dispensed into columns 1 and 2 of 1536-well assay plates. The remaining 46 columns were filled with 2 microliters of Assay Buffer supplemented with 225 ng/mL of Eu(K)-anti GST antibody and 42.5 nM of Flag-Core106. Next, 1 microliter of inhibition controls were dispensed into column 3 (12.5 micromolar unlabeled core106 protein, 100% inhibition), column 46 (200 nM core106 protein, 50% inhibition), and columns 4 to 45, 47 and 48 (Assay Buffer alone). Twenty-five nL of 10-points serial dilutions of test compounds or DMSO alone (0.5% final concentration) were then added to the appropriate wells. Next, 2 microliters of Assay Buffer supplemented with 2.5 micrograms/mL XL665-anti FLAG antibody and 33.75 nM GST-Core106 were dispensed to columns 3 to 48. The assay plates were centrifuged for 30 seconds at 300g and incubated for 4 hours at 22.5 degrees Celsius. At the end of the incubation time, TR-FRET was measured by exciting the plates at 340nm, and monitoring well fluorescence at 617 nm (Eu) and 671 nm (XL665) with the ViewLux microplate reader (PerkinElmer). All wells had a final volume of 5 microliters. The final reagent concentrations were 90 ng/mL Eu(K)-anti GST antibody, 1 microgram/mL XL665-anti FLAG, 17 nM Flag-Core106 and 13.5 nM GST-Core106. Final concentrations of competing, untagged Core 106 proteins were 2.5 micromolar and 40 nM for the 100% and 50% inhibition controls, respectively.

Secondary Assays

2.1b. Alphascreen Assays (see AID 463085, AID 651576, and AID 651577)

The purpose of this biochemical assay is to determine whether powder samples of compounds identified as possible HCV core probe candidates can disrupt the dimerization of HCV Core molecules (9). This assay employs the AlphaScreen technology, a secondary Amplified Luminescent Proximity Homogeneous Assay. This assay was run by the assay provider. Compounds were tested in duplicate at 15 micromolar (AID 651576) and in triplicate in a 10-point series starting at a nominal test concentration of 50 micromolar (AID 463085 and AID 651577).

Figure 1. Alphascreen Technology.

Figure 1Alphascreen Technology

AlphaScreen is based on the use of photoactive donor and acceptor beads that recognize specific tags on interacting proteins (10). Core106 dimerization was confirmed using AlphaScreen technology in which a core106 protein domain was tagged with either Glutathione-S-transferase (GST) tag or a Flag peptide tag. The untagged core106 protein domain was used as a model competitor in the assay. The proteins were diluted to working concentrations in ‘protein buffer’ (100 mM HEPES pH 7.5, 1 mM EDTA, 5 mM DTT, 0.1% CHAPS, 10% glycerol). The donor and acceptor beads were diluted to working concentrations in ‘bead buffer’ (20 mM HEPES pH7.5, 125 mM NaCl, 0.1% BSA, 0.1% CHAPS). GST-tagged core106 (150 nM) was incubated with 150 nM of Flag-tagged core106 for 1 hour at room temperature. Anti-Flag acceptor beads were added to the proteins at a final concentration of 20 μg/ml and incubated for 1 hour at room temperature. Then Glutathione donor beads were added to the proteins at a final concentration of 20 μg/ml and incubated for 1 hour. The assays were executed in a white 384 well Packard optiplate and were read on Perkin Elmer Envision.

2.1c. Cytotoxicity Assays (see AID 485280 and AID 651583)

The purpose of this assay is to determine whether powder samples of compounds identified as possible HCV core probe candidates are cytotoxic to Huh-7.5 cells. The assay utilizes compound XTT (2,3-bis[2-Methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxyanilide inner salt) with addition of an electron coupling agent phenazine methosulfate (PMS) to measure mitochondrial dehydrogenases activity in living cells. Dehydrogenases in mitochondria of living cells will cleave the tetrazolium ring of XTT changing the color of the solution from clear to orange. As designed, compounds that reduce cell viability will reduce activity of the mitochondrial dehydrogenases, thereby reducing the orange color in solution and well absorbance. Compounds were tested in triplicate in a 7-point 1:3 dilution series starting at a nominal test concentration of 320 μM (AID 485280 and AID 651583).

The Huh-7.5 cell line was routinely cultured in 15-cm dishes at 37 C and 95% relative humidity (RH). The growth media consisted of DMEM supplemented with 10% v/v certified fetal bovine serum, 4 mM L-glutamine, 1 mM sodium pyruvate, 1X antibiotic mix (penicillin, streptomycin, and glutamine), and 1X non-essential amino acids.

Prior to the start of the assay 8000 cells in 100 μL volume of colorless growth media were dispensed into each well of 96-well tissue culture-treated microtiter plates and incubated overnight at 37 C (5% CO2, 95% RH) to allow cells to adhere to the plate. The next day, 100 μL of test compound in DMSO (1.25 % final DMSO concentration) and DMSO only were added to wells. Wells with only test compound in media without cells were used to normalize the data (to account for the color a test compound may have). Next, the plates were incubated for 72 hours at 37 C (5% CO2, 95% RH). Solution of XTT-PMS was prepared in 1X PBS and added to the cells in 50 μL volume to each well and incubated for 4 hours at 37 C (5% CO2, 95% RH). After equilibrating the plates to room temperature for 10 minutes, optical density at 450 nm and 650 nm was measured on Biotek plate reader.

2.1d. Follow-up Assays to determine Selectivity and Cell-based Efficacy

Additional assays were outsourced to Ricerca or were performed by the assay provider in order to elucidate the selectivity and cell-based efficacy of the probe candidates. These assays are explained in subsequent sections of this report. See Sections 3.5 & 3.6 for details.

2.2. Probe Chemical Characterization

The chemical structure of the probe was verified by analysis of its 400 MHz 1H NMR spectra obtained on a Brüker 400 MHz instrument; data are summarized in Section 2.3 (Probe Preparation). The chemical structure was also corroborated by its LC/MS molecular ion (calc form/z (M+1): 416.2454.4 and (m/z (M+Na)): 476.3, using an Agilent 1200/6140 multimode quadrupole rapid resolve system) as well as its expected fragmentation patterns. Purity was measured at >95% (LC/MS analysis, confirmed by analytical HLPC analysis). HPLC data was obtained using an Agilent 1200 analytical HPLC with an Agilent Eclipse XDB-C18 column, 2.1×50mm. The HPLC solvents used were acetonitrile and water (gradient 0–100% acetonitrile in 5 min) with 0.1% formic acid added to each mobile phase as the pH modifier. The retention time found was 1.23 min.

As shown in Table 2, the solubility of probe ML322 in PBS at pH 7.4 was determined to be ≥100 μM. Solubility was ≥100 μM in conditions reflective of a cell-based assay environment (DMEM, 10% FBS). Importantly, its solubility is fully adequate to provide the high potency seen in cell-based infectivity assays (0.1–10 μM) and is also adequate for broad use as a biological probe to be used in a variety of aqueous-based media.

Table 2. Probe Properties.

Table 2

Probe Properties.

The probe has a half-life of >48 hours in PBS at room temperature when tested at 10 μM. Disappearance of the LC peak for the ML322 is not altered by addition of excess glutathione, indicating that ML322 is not thiol-reactive under physiologically-relevant conditions.

ML322 is stable in DMSO solution at room temperature (no erosion of peak intensity after 7 days) and is also stable as a free base dry powder.

Table 3 lists the samples and quantities of probe ML322 and analogs submitted to the MLSMR.

Table 3. Probe and Analogs submitted to the MLSMR.

Table 3

Probe and Analogs submitted to the MLSMR.

2.3. Probe Preparation (SR-1966; ML322)

Figure 2. Synthesis of probe ML322.

Figure 2Synthesis of probe ML322

Synthesis of diketone 3[9, 10]

To a solution of 2,4-pentadione 1 (1.0 g, 10 mmol) in acetone (10 mL) were successively added potassium carbonate (1.38 g, 10 mmol) and 4-methylbenzyl bromide 2 (1.85 g, 10 mmol). The mixture was stirred overnight at reflux and was quenched with water (10 mL). The aqueous phase was extracted with AcOEt and the combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. Purification of the crude product by flash chromatography on silica gel (hexanes/AcOEt: 4/1) afforded the known1,2 diketone 3 (1.82 g, 89%). 1H NMR (300 MHz, CDCl3) δ 16.81–16.83 (br. s, 0.5H), 6.97–7.15 (m, 4H), 3.99 (t, 0.5H, J= 7.3 Hz), 3.61 (s, 1H), 3.10 (d, 1H, J= 7.3 Hz), 2.30–2.32 (s, 3H), 2.12 (s, 3H), 2.06 (s, 3H).

Synthesis of ester 6 [11]

To a solution of NaOH (880 mg, 22 mmol) in water (50 mL) was added the commercially available 5-amino-3-mercapto-1,2,4-triazole (4) (2.32 g, 20 mmol). The mixture was stirred for 15 min at room temperature, then a solution of ethyl bromoacetate (3.34g, 20 mmol) in MeOH (5 mL) was added. The mixture was stirred for 2 h at room temperature. The solid in suspension was filtered and recrystallized from EtOH providing the known3 ester 6 (2.0 g, 50%). 1H NMR (400 MHz, DMSO-d6) δ 11.87 (s, 1H), 6.04 (s, 2H), 4.05 (q, J = 7.2 Hz, 2H), 3.82 (s, 2H), 1.15 (t, J = 7.2 Hz, 3H).

Synthesis of ester 7

To a solution of ester 6 (202 mg, 1 mmol) in acetic acid (5 mL) was added diketone 3 (204 mg, 1 mmol). The reaction was heated at 100°C for 24 h, then was concentrated under reduced pressure. Purification of the crude product by flash chromatography on silica gel (hexanes/AcOEt : 1/1) afforded ester 7 (160 mg, 43%). 1H (400 MHz, CDCl3) δ 7.10 (d, J = 8.0 Hz, 2H), 6.89 (d, J = 8.0 Hz, 2H), 4.23 (q, J = 7.2 Hz, 2H), 4.12 (s, 2H), 4.07 (s, 2H), 2.73 (s, 3H), 2.55 (s, 3H), 2.32 (s, 3H), 1.29 (t, J = 7.2 Hz, 3H).

Synthesis of carboxylic acid 8

To a 3N solution of NaOH (2 mL, 6 mmol) in THF (2 mL) was added ester 7 (160 mg, 0.43 mmol). The reaction mixture was stirred 5 h at room temperature, then was diluted with water (5 mL). The aqueous phase was extracted with EtOAc, then was acidified until pH 5 and was extracted twice with EtOAc. The combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give acid 8 (143 mg, 97%). 1H (400 MHz, CDCl3) δ 7.11 (d, J = 8.0 Hz, 2H), 6.89 (d, J = 8.0 Hz, 2H), 4.11 (s, 2H), 3.89 (s, 2H), 2.78 (s, 3H), 2.60 (s, 3H), 2.32 (s, 3H).

Synthesis of amide 10

To a solution of acid 8 (50 mg, 0.15 mmol) and triethylamine (0.1 mL, 0.75 mmol) in CH2Cl2 (1 mL) were successively added the hydrochloride salt of EDC (43 mg, 0.22 mmol), HOBt (30 mg, 0.22 mmol), and commercially available amine 9 (103 mg, 0.45 mmol). The reaction mixture was stirred at room temperature for 24 h and was directly subjected to column chromatography on silica gel (CH2Cl2/MeOH: 9/1). This provided amide 10 (75 mg, 90%). 1H (400 MHz, CDCl3) δ 7.69 (broad s, 1H), 7.11 (d, J = 8.0 Hz, 2H), 6.89 (d, J = 8.0 Hz, 2H ), 4.08 (s, 2H), 3.91 (s, 2H), 3.35 (q, J = 5.8 Hz, 2H), 3.27 (m, 4H), 2.75 (s, 3H), 2.56 (s, 3H), 2.44 (t, J = 6.3 Hz, 2H), 2.34-2.27 (m, 7H), 1.43 (s, 9H).

Synthesis of SR-1966 (Probe ML322)

To a solution of amide 10 (75 mg, 0.13 mmol) in CH2Cl2 (3 mL) was added trifluoroacetic acid (0.5 mL). The reaction mixture was stirred overnight at room temperature, then was concentrated under reduced pressure. Purification of the crude product by flash column chromatography using a Biotage SNAP cartridge KP-NH (CH2Cl2/MeOH : 9/1) afforded SR-1966 (56 mg, 95%). 1H (400 MHz, CDCl3) δ 7.59 (t, J = 5.1 Hz, 1H), 7.1 (d, J = 8.1 Hz, 2H), 6.9 (d, J = 8.1 Hz, 2H), 4.09 (broad s, 2H), 3.93 (broad s, 2H), 3.35 (q, J = 5.9 Hz, 2H), 2.76 (s, 3H), 2.69 (dd, J = 4.49, 5.25 Hz, 4H), 2.58 (s, 3H), 2.42 (t, J = 6.2 Hz, 2H), 2.34-2.28 (m, 7H); 13C (100 MHz, CDCl3) δ 168.8, 166.0, 164.8, 153.7, 144.6, 136.6, 133.8, 129.5 (2C), 127.3 (2C), 119.6, 56.8, 52.9 (2C), 45.2 (2C), 36.3, 34.4, 32.9, 24.0, 20.9, 14.2; IR (neat) 3293, 2943, 2816, 1663, 1613, 1511, 1377, 1367, 1341, 1295, 1158 cm−1. MS (ES−) m/z = 452 (found for C23H31N7OS-H+).

3. Results

The flow chart (Figure 3) outlines the results of the uHTS screening campaign for HCV Core dimerization inhibitors at the SRIMSC. Following primary HTS in singlicate to identify HCV Core dimerization inhibitors using a TR-FRET-based assay (AID 1899), available MLSMR compounds were selected for confirmation testing in triplicate (AID 2152). Available compounds that confirmed activity and were deemed not to be fluorescent artifacts were tested in dose response assays in triplicate (AID 2159). Of the 128 compounds ordered, 24 were removed as being artifact, and 12 were not available from the MLSMR. The dose response identified 92 compounds as possible quenchers or fluorescent artifiacts. As a result the top 9 available non-optically active compounds were ordered for testing as powders for confirmation of activity.

Figure 3. HCV Core dimerization HTS campaign overview.

Figure 3

HCV Core dimerization HTS campaign overview.

The compounds with the highest potency in the HTS HCV Core FRET-based dose response assay (AID 2159) are shown in Table 4.

Table 4. Most potent scaffolds from the HTS campaign.

Table 4

Most potent scaffolds from the HTS campaign.

Following cheminformatic analyses of the 92 compounds tested in HCV core titration HTS assays, fluorescence quenchers and fluorescent artifacts (non-optically active) were removed, resulting in 9 compounds with promising activity. Two compounds did not recapitulate activity.

Compounds identified as clean hits included SID 49716510 (CID 22422906/ SR-01000819396), SID 56317368 (CID 24979772), SID 56323176 (CID 9426403), SID 47193664 (CID 5280489), SID 56316484 (CID 24979394) (see Figure 6). Powder samples of 9 promising compounds were ordered by the SRIMSC and tested in the same FRET-based biochemical HCV core dimerization assay to confirm activity (Table 5, AID 2488). Only CID 22422906 was active (IC50 = 3.66 μM), and its potency was similar to that of the earlier (liquid) batch provided by the MLSMR (IC50 = 9.86 μM, AID 2159).

Figure 6. Structures of prior art compounds SL201 and SL209.

Figure 6

Structures of prior art compounds SL201 and SL209.

Table 5. The top Round 0 analog results.

Table 5

The top Round 0 analog results.

Samples of SID 56317368 and SID 56323176 that were synthesized in our laboratory had activity far less than the HTS and were not pursued further. SID 56316484 (CID 24979394) was not pursued owing to the weak activity from HTS (IC50 = 48 μM), plus the presence of a labile ester bond was a concern. SID 47193664 (CID 5280489) was not pursued owing to the lack of a clear path for SAR. Finally, SID 855627 (CID 1780) was identified as a “frequent hitter” active in numerous PubChem assays and was removed from further consideration.

Based on recapitulation of CID 22422906, this compound and purchased analogs were submitted to the assay provider for testing in Alphascreen assays at 15 micromolar and liver cell cytotoxicity assays. As shown in the first row of Table 5, only CID 22422906 met the criteria of a probe candidate: non-toxic to Huh 7.5 liver cells (AID 485280, CC50 = 123 μM), active in a secondary amplified luminescent proximity homogeneous assay (Alphascreen AID 463085, IC50 = 5 μM), and able to inhibit the levels of HCV RNA in infected cells treated with viral culture supernatants (QPCR assay, AID 485271). CID 22422906 is active in only 15 of 288 PubChem Bioassays tested (5.2%); however, several of the assays are fluorescence- or FRET-based. As a result, further SAR was pursued. This compound belongs to the 1,2,4]triazolo[1,5-a]pyrimidin-2-yl scaffold.

Medicinal Chemistry to explore SAR of CID 22422906

Due to the promising activity and lack of liver cell cytotoxicity of CID 22422906, the SRISMC purchased and synthesized additional analogs to explore the SAR. These analogs were submitted to the Strosberg lab for testing using an orthogonal Alphascreen-based biochemical assay at 15 micromolar and to determine potency (Table 6).

Table 6. Round 1 Analogs: Alphascreen results.

Table 6

Round 1 Analogs: Alphascreen results. SL274 was ultimately claimed as probe ML322.

3.1. Summary of Screening Results

The HTS campaign identified a number of potential probe precursors (HTS Figure 3). The series of efforts used to determine probe candidates and ultimately the probe ML322 is summarized in the hit to probe optimization flow chart (Figure 5). Data from primary, confirmatory and concentration response for the HCV Core screen were subjected to cheminformatics analysis to determine what scaffolds have near-neighbor active and inactive analogs, indicating the potential for SAR.

Figure 5. Probe development overview (post-HTS).

Figure 5

Probe development overview (post-HTS).

The most promising hits in HCV Core active scaffolds were then validated by assay data obtained using independent batches of test samples, purchased or synthesized in-house. Certain hits gave variable assay results upon re-supply. Others validated upon re-supply, with HCV Core IC50 values within 2-fold of the HTS value. These were the hits of highest interest that progressed to SAR studies. Compounds were further prioritized as candidates for probe development based upon the SAR of purchased or synthesized analogs, based also upon the finding that certain compounds gave only low levels of HCV Core dimerization inhibition relative to other compounds (Figure 5).

As discussed in Section 3, only CID 22422906 survived as a viable starting point for probe development. To explore this series, the Strosberg lab pursued additional counterscreening and cell-based efficacy testing (Table 7). Also included in Table 7 are comparative data for two compounds, SL201 and SL209 (Figure 6), derived from prior work by the assay provider on the development of small molecule inhibitors of the hepatitis C virus by inhibition of Core dimerization [12]. As shown by these comparative data, SL201 and SL209 are far less effective at inhibiting production of infectious HCV particles (IC50 = 0.286 μM for SL209) than the probe compound (SID 103771539; SR-1966, SL274) that has IC50 = 2 nM in this assay.

Table 7. Secondary assay results (Strosberg lab).

Table 7

Secondary assay results (Strosberg lab). SL274 was the most potent in cell-based HCV infectivity assays and was selected to be probe ML322.

3.2. Dose Response Curves for Probe

As shown in Table 7, ML322 has robust on-target activity in biochemical and cell-based target assays. The probe is not cytotoxic to Huh7.5 liver cells, and is able to block HCV production in these same cells. The dose response curves for the probe in the HCV biochemical and cell-based assays are shown (Figure 7).

Figure 7. Dose response curves for Probe ML322 in the Alphascreen (A), QPCR T1/T2 (B), and TCID50 assays (C).

Figure 7

Dose response curves for Probe ML322 in the Alphascreen (A), QPCR T1/T2 (B), and TCID50 assays (C).

3.3. Scaffold/Moiety Chemical Liabilities

As discussed in the original Chemical Probe Development Plan (CPDP) for this project, hits were prioritized based on their SAR profile determined from HTS, follow-up assays and results of SAR by purchase. The potency, selectivity and chemical tractability of the hit series summarized in Sections 3.0 and 3.1 were considered. Hits or scaffold series that possess potential chemical instability or toxicological problems were eliminated (e.g., SID 47193664 and SID 855627), as clear strategies could not be devised to address these issues for these two hits. The chemical series selected for optimization (e.g., SID 49716510) has the potential to address lesions associated with existing probes, to meet the criteria for a successful probe as laid out in the CPDP, and to improve the state of the art in the field.

Synthetic tractability, chemical stability, stability in biological systems, drug-likeness [13, 14], lead likeness [15], lack of structural alerts [16] and anticipated physical properties such as solubility are considered in the selection of hits for probe development.

The post-HTS analysis led to the identification of six compounds as the top hits (Figure 4). Of these, all but SID 49716510 were eliminated from further consideration owing to the limited opportunities for SAR expansion or the weakness of the original hit (see reasons summarized in red for each structure in Figure 4). SID855627 has also been eliminated from further consideration owing to its promiscuous nature in PubChem assays, and concerns about the chemical/biochemical stability of the polyacetylene unit.

Figure 4. Selection of SID 49716510 as the starting point for SAR hit optimization.

Figure 4

Selection of SID 49716510 as the starting point for SAR hit optimization.

SID 49716510 was confirmed using a powder sample purchased from a commercial source. One round of SAR by purchase was performed (9 analogs were available), and several of the compounds were as active as, if not more active as SID 49716510. Based on these results, the first round of analog synthesis was completed, and an additional ca. 20 SID 49716510 analogs were provided to the Strosberg lab. After HCV core dimerization inhibition data with those new analogs, a decision was made to resource the project for additional analog synthesis. Subsequent testing of synthesized analogs by the Strosberg lab revealed that CID 49800087 was the most promising probe candidate (Figure 8).

Figure 8. Comparison of the structures of the original HTS lead and final Probe ML322.

Figure 8

Comparison of the structures of the original HTS lead and final Probe ML322.

Because several individual compounds within this [1,2,4]triazolo[1,5-a]pyrimidin-2-yl inhibitor series showed the biochemical and cell-based potency and lack of cytotoxicity necessary to be high quality probes (Table 7), we subjected samples of the top compounds to CYP450 analysis (Table 8) and microsomal stability (Table 9) to aid in probe selection. The results of these studies revealed that probe ML322 does not significantly inhibit cytochrome P450 enzymes. The probe is highly stable to liver microsomes in mouse and human, with a measured half-life of 40 min and 27 min, respectively.

Table 8. Probe ML322 is not a strong inhibitor of cytochrome P450 enzymes.

Table 8

Probe ML322 is not a strong inhibitor of cytochrome P450 enzymes.

Table 9. Hepatic Microsome Stability of Selected HCV Core Inhibitors.

Table 9

Hepatic Microsome Stability of Selected HCV Core Inhibitors.

Results in Tables 79 show that several compounds were generally suitable as probes. We chose CID 49800087 (SR-1966 or SL274) as the probe (designated ML322) due to its superior activity in inhibiting formation of infectious HCV particles (Table 7), its lack of significant inhibition of human CYPs, and the presence of a basic amine to aid solubility and ease of formulation. ML322 has no serious structural liabilities. Minor issues are the relatively poor stability of ML322 analogs CID 22422908 and CID 9550395 in mouse microsomes.

The probe contains a [1,2,4]triazolo[1,5-a]pyrimidin-2-yl scaffold (Figure 7). It contains one amide bond which may pose a liability to protease degradation and a thioether that is potentially prone to metabolic oxidation or reduction. The probe and analogs do not react with glutathione and are chemically stable so long as they are not used in highly acidic media (pH<2). The relative ease of synthesis of the probe molecule and analogs by the described method is a strong asset for this scaffold series and this probe. Scale-up synthesis, if needed for extensive long-term biological studies, will be highly feasible.

3.4. SAR Tables

SAR Table 1Lead scaffolds from the HTS campaign

SR NumberSIDCIDVendorStructureAID 2488
HTRF IC50 (uM)
OutcomeAID 463085
Alphascreen IC50 (uM)
OutcomeAID 485271
HCV QPCR T1 IC50 (uM)
T2 IC50 (uM)OutcomeAID 485280
Cytox CC50
Outcome
SR-01000819396-28721921122422906ChemDiv
Image ml322fu37.jpg
3.666Active5.0Active6.612Active123Inactive
SR-01000763803-4872192085280489Sigma Chemical
Image ml322fu38.jpg
>49.751Inactive34.8InactiveNot tested due to lack of Alphascreen activity171.9Inactive
SR-01000839920-28721921324979772Enamine
Image ml322fu39.jpg
20.49Inactive29.8Inactive140.7Inactive
SR-01000000036-3872192071780Sigma Chemical
Image ml322fu40.jpg
>49.751InactiveNot tested due to lack of HTRF activityNot tested due to lack of HTRF activity178.6Inactive
SR-01000812654-287219209694792ChemBridge
Image ml322fu41.jpg
>49.751InactiveNot tested due to lack of HTRF activityNot tested due to lack of HTRF activity142.9Inactive
SR-01000812946-2872192101557339Pharmeks
Image ml322fu42.jpg
46.17Inactive24.7Inactive
SR-01000839796-28721921224981182Enamine
Image ml322fu43.jpg
>49.751Inactive27.4Inactive
SR-01000840236-28721921424979394Enamine
Image ml322fu44.jpg
40.43Inactive148.4Inactive
SR-01000849831-2872192159426403Enamine
Image ml322fu45.jpg
21.17Inactive163.3Inactive

SAR Table 2Probe and lead analogs

SR NumberSIDCIDVendorStructureAID 651576
%INH at 15 μM
%INH OutcomeIC50 Core Dimerization (μM)IC50 OutcomeAssay OUTCOMEAID 651574
T1 IC50 (μM)
OutcomeT2 IC50 (μM)OutcomeOverall OutcomeAID 624575
TCID50 (uM)
OutcomeAID 624583
Cytox
Outcome
SR-03000001966-110377153949800087SRIMSC-synth
Image ml322fu46.jpg
51Active8.4ActiveActive: PROBE ML322IC50 not determined at this timepoint1.8ActiveActive (PROBE ML322)0.002Active (PROBE ML322)33.79Inactive (Probe ML322)
SR-03000001539-39930921122422908SRIMSC-synth
Image ml322fu47.jpg
82Active1.9ActiveActive8.5Active13.7ActiveActive0.191Active42.6Inactive
SR-01000668805-21037715389550395SRIMSC-synth
Image ml322fu48.jpg
60Active1.0ActiveActive11.96ActiveNot tested due to probe’s lower TCID50 valueActive0.312Active> 12Inactive
SR-03000002010-110417972349849980SRIMSC-synth
Image ml322fu49.jpg
66Active3.0ActiveActive0.099ActiveActive0.032Active180.4Inactive
SR-03000002013-110417972649849987SRIMSC-synth
Image ml322fu50.jpg
87Active1.3ActiveActiveNot tested due to probe’s lower cytotoxicity> 4Inactive
SR-03000002015-110417972849849986SRIMSC-synth
Image ml322fu51.jpg
65Active5.7ActiveActive3.067ActiveNot tested due to probe’s lower TCID50 valueActive0.294Active> 36Inactive
SR-03000002037-110422309749852628SRIMSC-synth
Image ml322fu52.jpg
79Active2.6ActiveActive2.7Active7.2ActiveActive0.004Active13.83Active
SR-03000002053-110423365349859551SRIMSC-synth
Image ml322fu53.jpg
62.4Active10.2ActiveActive5.3Active12.5ActiveActiveNot tested due to probe’s lower TCID50 value37.64Inactive

SAR Table 3Analogs

SR NumberSIDCIDVendorStructureAID 651576
%INH of Core dimerization at 15 μM
OutcomeIC50 Core
Dimerization (μM)
Outcome (Active, Inactive, or Not Tested)OVERALL OUTCOMEAID 651574
T1 IC50 (μM)
OutcomeT2 IC50 (μM)OutcomeOverall OutcomeAID 624575
TCID50 (uM)
OutcomeAID 624583
Cytox
Outcome
SR-03000002229-112305059452914829SRIMSC-synth
Image ml322fu54.jpg
85%Active9.9ActiveActive7.7ActiveT2 result not determinedActive0.286Active22.11Active
SR-03000002054-110423365449859550SRIMSC-synth
Image ml322fu55.jpg
53%Active5.1ActiveActive18.9Active24.3InactiveInactive1.241Active193.2Inactive
SR-03000002055-110423365549859547SRIMSC-synth
Image ml322fu56.jpg
83%Active1.6ActiveActive3.2Active2.2ActiveActive0.665Active9.51Active
SR-03000001539-19602235222422908ChemNavigator
Image ml322fu57.jpg
97%Active1.9ActiveActive8.5Active13.7ActiveActiveNot tested due to probe’s lower TCID50 value
SR-01000819396-39602235722422906ChemNavigator
Image ml322fu58.jpg
16%InactiveLow %INH value so IC50 not pursuedInactive6.7Active12ActiveActive
SR-03000001523-19602233422422894ChemNavigator
Image ml322fu59.jpg
59%ActiveActiveNot tested due to probe’s lower TCID50 value
SR-01000550123-2960223484060916ChemNavigator
Image ml322fu60.jpg
40%InactiveInactive
SR-03000002051-110423365149859548SRIMSC-synth
Image ml322fu61.jpg
< 50%InactiveInactive
SR NumberSIDCIDVendorStructureAID 651576
%INH of Core dimerization at 15 μM
OutcomeIC50 Core
Dimerization (μM)
Outcome (Active, Inactive, or Not Tested)
SR-03000002052-110423365249859549SRIMSC-synth
Image ml322fu62.jpg
< 50%InactiveNot tested because Probe
ML322 found to be more potent.
SR-03000001537-19602234915997098ChemNavigator
Image ml322fu63.jpg
71%Active
SR-01000550140-2960223504144944ChemNavigator
Image ml322fu64.jpg
30%Inactive
SR-03000001538-19602235115998711ChemNavigator
Image ml322fu65.jpg
38%Inactive
SR-01000550124-2960223544060917ChemNavigator
Image ml322fu66.jpg
22%Inactive
SR-01000550112-2960223554060781ChemNavigator
Image ml322fu67.jpg
52%Active
SR-03000001540-19602235622422895ChemNavigator
Image ml322fu68.jpg
65%Active
SR-03000001541-1960223586621239ChemNavigator
Image ml322fu69.jpg
33%Inactive
SR-03000001542-19602235915987932ChemNavigator
Image ml322fu70.jpg
43%Inactive
SR-03000001581-19609979646238463SRIMSC-synth
Image ml322fu71.jpg
52%Active
SR-03000001587-19730216346245561SRIMSC-synth
Image ml322fu72.jpg
87%Active
SR-03000001588-19920583746829287SRIMSC-synth
Image ml322fu73.jpg
88%Active
SR-03000001650-19923365746846222SRIMSC-synth
Image ml322fu74.jpg
96%Active
SR-03000001651-19923365846846224SRIMSC-synth
Image ml322fu75.jpg
88%Active
SR-03000001652-19923365946846226SRIMSC-synth
Image ml322fu76.jpg
78%Active
SR-03000001653-19923366046846218SRIMSC-synth
Image ml322fu77.jpg
80%Active
SR-03000001654-19923366146846216SRIMSC-synth
Image ml322fu78.jpg
67%Active
SR-03000001655-19923366246846225SRIMSC-synth
Image ml322fu79.jpg
85%Active
SR-03000001656-19923366346846228SRIMSC-synth
Image ml322fu80.jpg
89%Active
SR-03000001657-19923366446846219SRIMSC-synth
Image ml322fu81.jpg
90%Active
SR-03000001658-19923366546846231SRIMSC-synth
Image ml322fu82.jpg
92%Active
SR-03000001659-19923366646846217SRIMSC-synth
Image ml322fu83.jpg
98%Active
SR-03000001660-19923366746846232SRIMSC-synth
Image ml322fu84.jpg
89%Active
SR-03000001661-19923366846846221SRIMSC-synth
Image ml322fu85.jpg
80%Active
SR-03000001668-19923367046846220SRIMSC-synth
Image ml322fu86.jpg
93%Active
SR-03000001692-19923945746850834SRIMSC-synth
Image ml322fu87.jpg
67%Active
SR-03000001693-19923945846850820SRIMSC-synth
Image ml322fu88.jpg
80%Active
SR-03000001963-110377153649800086SRIMSC-synth
Image ml322fu89.jpg
< 50%Inactive
SR-03000001964-110377153749800089SRIMSC-synth
Image ml322fu90.jpg
< 50%Inactive
SR-03000001982-110394720449837841SRIMSC-synth
Image ml322fu91.jpg
< 50%Inactive
SR-03000001983-110394720549837838SRIMSC-synth
Image ml322fu92.jpg
< 50%Inactive
SR-03000002009-110417972249849981SRIMSC-synth
Image ml322fu93.jpg
< 50%Inactive
SR-03000002011-110417972449849983SRIMSC-synth
Image ml322fu94.jpg
66%Active52Inactive
SR-03000002012-110417972549849979SRIMSC-synth
Image ml322fu95.jpg
< 50%InactiveNot tested because Probe
ML322 found to be more potent.
SR-03000002014-110417972749849975SRIMSC-synth
Image ml322fu96.jpg
< 50%Inactive
SR-03000002029-110422308949852629SRIMSC-synth
Image ml322fu97.jpg
< 50%Inactive
SR-03000002029-110422308949852629SRIMSC-synth
Image ml322fu97.jpg
<50%Inactive
SR-03000002030-110422309049852642SRIMSC-synth
Image ml322fu98.jpg
< 50%Inactive
SR-03000002030-110422309049852642SRIMSC-synth
Image ml322fu98.jpg
<50%Inactive
SR-03000002031-110422309149852630SRIMSC-synth
Image ml322fu99.jpg
<50%Inactive
SR-03000002031-110422309149852630SRIMSC-synth
Image ml322fu99.jpg
< 50%Inactive
SR-03000002032-110422309249852633SRIMSC-synth
Image ml322fu100.jpg
< 50%Inactive
SR-03000002033-110422309349852646SRIMSC-synth
Image ml322fu101.jpg
< 50%Inactive
SR-03000002034-110422309449852637SRIMSC-synth
Image ml322fu102.jpg
< 50%Inactive
SR-03000002035-110422309549852649SRIMSC-synth
Image ml322fu103.jpg
<50%Inactive
SR-03000002036-110422309649852644SRIMSC-synth
Image ml322fu104.jpg
<50%Inactive
SR-03000002128-111427961250930769SRIMSC-synth
Image ml322fu105.jpg
<50%Inactive
SR-03000002169-111693361450985833SRIMSC-synth
Image ml322fu106.jpg
<50%Inactive
SR-03000002186-111769595251035445SRIMSC-synth
Image ml322fu107.jpg
−11%Inactive
SR-03000002228-112305059352914830SRIMSC-synth
Image ml322fu108.jpg
44%Inactive
SR-03000002230-112305059552914838SRIMSC-synth
Image ml322fu109.jpg
46%Inactive
SR-03000002231-112305059652914841SRIMSC-synth
Image ml322fu110.jpg
87%Inactive
SR-03000002232-112305059752914840SRIMSC-synth
Image ml322fu111.jpg
41%Inactive
SR-01000120796-212305059822422913SRIMSC-synth
Image ml322fu112.jpg
13%Inactive
SR-03000002233-112305059952914831SRIMSC-synth
Image ml322fu113.jpg
37%Inactive
SR-03000002234-112305060052914835SRIMSC-synth
Image ml322fu114.jpg
63%Active
SR-03000002252-112305498152918352SRIMSC-synth
Image ml322fu115.jpg
79%,Active
SR-03000002253-112305498252918345SRIMSC-synth
Image ml322fu116.jpg
11%Inactive
SR-03000002256-112305498452918341SRIMSC-synth
Image ml322fu117.jpg
51%Active
SR-03000002294-112433863053230259SRIMSC-synth
Image ml322fu118.jpg
42%Inactive
SR-03000002295-112433863153230283SRIMSC-synth
Image ml322fu119.jpg
25%Inactive
SR-03000002340-112434934653239851SRIMSC-synth
Image ml322fu120.jpg
12%Inactive
SR-03000002341-112434934753239825SRIMSC-synth
Image ml322fu121.jpg
10%Inactive
SR-03000002342-112434934853239847SRIMSC-synth
Image ml322fu122.jpg
44%Inactive
SR-03000002343-112434934953239839SRIMSC-synth
Image ml322fu123.jpg
43%Inactive
SR-03000002344-112434935053239842SRIMSC-synth
Image ml322fu124.jpg
14%Inactive
SR-03000002345-112434935153239843SRIMSC-synth
Image ml322fu125.jpg
41%Inactive
SR-03000001965-113564960756946976SRIMSC-synth
Image ml322fu126.jpg
51%Active29Inactive

3.5. Cellular Activity

HCV Infectivity Assays

The next set of assays performed were biological assays used to determine the ability of compounds to inhibit production of J6/JFH1, 2a strain virus in Huh-7.5 liver cells (Figure 9). HCV infectivity is measured using real-time RT-PCR to monitor changes in expression of HCV 2a J6/JFH-1 RNA. Cells are incubated with test compound in the presence of HCV, followed by isolation of RNA, conversion to cDNA, and Taqman-based QPCR. As designed, a compound that inhibits HCV infectivity will reduce HCV RNA expression, leading to decreased production of the PCR amplicon, thereby reducing fluorescence, and increasing Ct. Compounds were tested in triplicate using a 6-point 1:10 dilution series starting at a nominal test concentration of 100 μM. Note that the TCID50 values (AID 624575) are significantly lower than the T1/T2 QPCR values (AID 485271 Rounds 0–1; AID 651574 (Round 2). These assays have been previously described [12, 17].

Figure 9. HCV Infectivity assay method.

Figure 9

HCV Infectivity assay method.

Cells were plated the day before the assay. After allowing the cells to adhere overnight, test compound was prepared in HCV supernatant by making 1:10 serial dilutions from 100 μM down to 0.001 μM. Doses of test compound in virus were added to cells and incubated for 24 hours. The next day, cell culture media was removed from each well and replaced with the same dilutions of compound in complete media. This was added to cells and incubated for another 48 hours for the T1 timepoint. Next, the cells were lysed and RNA was isolated using the RNeasy kit (Qiagen, Valencia, CA). Supernatant from T1 (early stage infection) was transferred to naive cell cultures and incubated for 24 hours. After 24 hours the culture medium was removed from the cells and replaced with complete media and the cells were incubated for 48 hours for the T2 (late stage) timepoint. Cells for the T2 timepoint were lysed and RNA was isolated as was done for the T1 timepoint. cDNA was generated using the Taqman reverse transcription kit (Applied Biosystems, Foster City, CA). Quantitative real-time polymerase chain reaction (QPCR) was performed in triplicate using the LightCycler RNA Amplification Kit HybProbe master mix (Roche) with Taqman MGB Probe 6FAM-TATGAGTGTCGTGCAGCCTC-MGBNFQ on a model LightCycler480 real time PCR system (Roche). Data are expressed as the mean fold change in mRNA levels, plus or minus the SE of 3 replicates normalized to 100 μg total RNA. Primers used were forward CTTCACGCAGAAAGCGTCTA and reverse CAAGCACCCTATCAGGCAGT. The range of activity was normalized based on measurement of total RNA. Compounds with an IC50 value greater than 20 μM were considered inactive. Compounds with an IC50 value equal to or less than 20 μM were considered active. All compounds tested were active in this assay. However, one compound (SR-03000001966-1/ SID 103771539/ CID 49800087) exhibited significantly greater potency than all others compounds tested: TCID50 = 2 nM. As a result this compound was claimed as the probe ML322 (Table 10, next page).

Table 10. TCID50 Results (see AID 624575).

Table 10

TCID50 Results (see AID 624575).

3.6. Profiling Assays

Ricerca Receptor Profiling

The cell-based assays used in screening and in support of our lead optimization efforts define a clear mode of action for ML322: disruption of the dimerization of HCV Core proteins. To support the selectivity for this mode of action hypothesis, we submitted the probe for ligand binding competition studies against a panel of protein targets of therapeutic and/or toxicological interest including GPCRs, transporters, ion channels, and receptors. This assay was performed in duplicate by Ricerca, LLC at 10 micromolar See company website and PubChem AID 624406 for details.

Methods employed in this study 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. To be considered “Active” the probe must have inhibited ligand binding to the target by greater than 50% (this value was set by Ricerca). Negative inhibition values represent a stimulation of binding. Interestingly, the top 6 targets (%INH > 40%) are restricted to the adrenergic (alpha), muscarinic (M1–M3), and dopamine transporter families. Note that the probe did not significantly inhibit the majority of targets tested (%INH < 20%). Further, the probe did not inhibit ligand binding to any target by more than 64%. While future efforts may be pursued to elucidate the mechanism by which ML322 affects adrenergic (alpha), muscarinic (M1-3), and dopaminergic ligand binding, the general lack of activity in this assay and in the liver cell cytotoxicity counterscreens provides evidence for Core target specificity.

Table 11Ricerca profiling of probe ML322: SID 103771539 /CID49800087

Ricerca CatTargetSpecies% INH (10 uM)Outcome
203200Adrenergic α1Brat63Active
220320Transporter, Dopamine (DAT)hum62Active
203400Adrenergic α1Dhum60Active
252810Muscarinic M3hum54Active
252710Muscarinic M2hum49Inactive
252610Muscarinic M1hum42Inactive
241000Imidazoline I2, Centralrat31Inactive
203100Adrenergic α1Arat27Inactive
226500GABAA, Muscimol, Centralrat26Inactive
239610Histamine H1hum24Inactive
255520Tachykinin NK1hum24Inactive
271700Serotonin (5-Hydroxytryptamine) 5-HT2Bhum21Inactive
226600GABAA, Flunitrazepam, Centralrat19Inactive
260210Opiate κ(OP2, KOP)hum18Inactive
232030Glucocorticoidhum17Inactive
204410Transporter, Norepinephrine (NET)hum17Inactive
203620Adrenergic α2Ahum16Inactive
232700Glutamate, Kainaterat16Inactive
274030Transporter, Serotonin (5-Hydroxytryptamine)hum16Inactive
271110Serotonin (5-Hydroxytryptamine) 5-HT1Ahum15Inactive
204110Adrenergic β2hum14Inactive
243520Interleukin IL-1mouse13Inactive
265900Potassium Channel hERGhum13Inactive
233000Glutamate, NMDA, Phencyclidinerat12Inactive
278110Sigma σ1hum12Inactive
214510Calcium Channel L-Type, Benzothiazepinerat11Inactive
260410Opiate μ(OP3, MOP)hum11Inactive
232910Glutamate, NMDA, Glycinerat10Inactive
200510Adenosine A1hum9Inactive
217030Cannabinoid CB1hum9Inactive
239820Histamine H3hum9Inactive
285010Androgen (Testosterone) ARrat7Inactive
225510Epidermal Growth Factor (EGF)hum7Inactive
228610GABAB1Ahum7Inactive
219500Dopamine D1hum6Inactive
239710Histamine H2hum6Inactive
268700Purinergic P2Xrabbit6Inactive
212510Bradykinin B1hum5Inactive
216000Calcium Channel N-Typerat5Inactive
219700Dopamine D2Shum5Inactive
285900Thyroid Hormonerat5Inactive
214600Calcium Channel L-Type, Dihydropyridinerat3Inactive
232810Glutamate, NMDA, Agonismrat3Inactive
251600Melatonin MT1hum3Inactive
265010Platelet Activating Factor (PAF)hum3Inactive
268810Purinergic P2Yrat3Inactive
200720Adenosine A3hum2Inactive
226400Transporter, GABArat2Inactive
258590Nicotinic Acetylcholinehum1Inactive
258700Nicotinic Acetylcholine α, Bungarotoxinhum1Inactive
268420Prostanoid EP4hum1Inactive
270000Rolipramrat1Inactive
219800Dopamine D3hum0Inactive
257010Neuropeptide Y Y1hum0Inactive
219900Dopamine D4.2hum−1Inactive
224110Endothelin ETBhum−1Inactive
264500Phorbol Estermouse−1Inactive
200610Adenosine A2Ahum−2Inactive
204010Adrenergic β1hum−2Inactive
257110Neuropeptide Y Y2hum−3Inactive
260130Opiate δ1 (OP1, DOP)hum−3Inactive
212620Bradykinin B2hum−4Inactive
226010Estrogen ERαhum−4Inactive
271910Serotonin (5-Hydroxytryptamine) 5-HT3hum−4Inactive
224010Endothelin ETAhum−5Inactive
250460Leukotriene, Cysteinyl CysLT1hum−7Inactive
265600Potassium Channel [KATP]hamster−11Inactive

4. Discussion

Core is a structural protein of the HCV virion and is essential for viral assembly and infectious virus production. ML322 is a potent and selective inhibitor of the core protein-protein dimerization. The probe has an IC50 of 8.4 μM in the absorbance-based biochemical Amplified Luminescent Proximity Homogeneous Assay (Alphascreen), indicating it can block the interaction between HCV core proteins. Because core-core dimerization is thought to be important for mediating viral assembly and formation of the viral nucleocapsid cell-based infectivity assays were performed to determine whether the probe could block infectivity using a cell culture system. The probe exhibits an IC50 of 1.8 μM (T2 QPCR) for blocking the levels of total HCV RNA in cells. In addition the probe potently reduces the levels of infections HCV RNA in cells (TCID50 = 2 nM). The significantly lower IC50 value in the TCID50 assay, relative to the T2 QPCR assay, reflects the fact that one needs to interfere with only a few of the potential core-core protein interactions to prevent formation an infectious HCV particle (TCID50 assay), whereas the T2 QPCR data reflects the concentration required to inhibit one-half of all possible core-core dimer interactions in the in vivo experiment. All sets of biochemical and cellular assay data were highly reproducible between batches and runs. Importantly, ML322 also consistently lacks activity in the hepatocyte cytotoxicity counterscreen (CC50 >30 μM), demonstrating that it does not significantly affect liver cell viability. Probe ML322 lacks significant inhibitory activity effects against the majority of therapeutic targets tested (Ricerca screening). Together these results demonstrate that probe ML322 far surpasses the probe criteria set forth in the chemical probe development plan.

It must be noted that protein-protein interactions are notoriously difficult to efficiently and selectivity disrupt using small molecule ligands and that micromolar activity for disrupting a protein-protein interaction using a rule-of-five compliant small molecule is noteworthy. In fact, certain clinically useful anticancer compounds acting by disrupting protein-protein interactions are less potent against their targeted proteins [18]. The finding that probe ML322 is not cytotoxic to liver cells (Huh7.5) indicates that the probe may be particularly useful to researchers in testing the hypothesis that targeting interactions of core proteins can thwart mechanisms used by HCV to avoid inhibition by other immune enhancing drugs such as pegylated interferon or ribavirin, treatments currently in use for hepatitis with significant off-target effects.

The probe lacks general reactivity (is nonreactive with glutathione), is not a significant inhibitor of major CYP450 isoforms, and is readily synthesized in high purity without chromatography in a single chemical step. Probe ML322 also has chemical stability, with useful levels of water solubility (>100 μM in PBS, >100 μM in simulated assay conditions). Moreover, the probe has favorable properties associated with drug-likeness, with a moderate molecular weight (454), acceptable parameters for LogP (2.76), tPSA (84.7), and lack of groups associated with inherent toxicity. Due to these chemical and biological properties, ML322 should prove useful in in vitro and perhaps in in vivo studies aimed at understanding how HCV core dimerization regulates HCV packaging, host cell oncogenesis, and suppression of host T cell responses.

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

At the onset of the HTS campaign no published prior art small molecules were known to selectively disrupt the dimerization of HCV core. However, during the HTS and chemistry effort that led to discovery of ML322, the assay provider in collaboration with Boston University discovered several molecules with activity in the HCV Core dimerization Alphascreen and cell-based infectivity assays (Table 7). While these compounds represent exciting and promising scaffolds, none exhibits the cell-based potency as high as that of ML322 (TCID50 = 2nM). Additional studies are needed to elucidate the specific mechanisms by which these compounds inhibit core dimerization. However, 4 of the 5 compounds in Table 12 (ML322 included) do not exhibit liver cell toxicity, suggesting their action is targeted to the viral core protein.

Table 12. Comparison of the prior art with HCV Core probe ML322.

Table 12

Comparison of the prior art with HCV Core probe ML322.

4.2. Mechanism of Action Studies

Because our screening and hit follow-up protocols include relevant targets and anti-targets in both biochemical assays and cell-based assays, we were able to establish a HCV core dimerization specific mode of action. ML322 has robust on-target activity in biochemical and cell-based assays and lacks toxicity in the same Huh7.5 liver cell line used for the cell-based infectivity assays. It has clear advantages over prior art in selectivity and defined mode of action (Table 12).

4.3. Planned Future Studies

Sadly, the assay provider Dr. Donny Strosberg suddenly passed away during the probe development effort. As a result there are no further efforts planned by the SRIMSC for this probe. However, the compound and analogs have been made available to the MLSMR for other researchers to use as tools for inhibiting HCV core dimerization and HCV infectivity in cells.

5. References

1.
Hoofnagle JH. Course and outcome of hepatitis C. Hepatology. 2002;36(5 Suppl 1):s21–s29. [PubMed: 12407573]
2.
Soriano V, et al. Challenges and opportunities for hepatitis C drug development in HIV-hepatitis C virus-co-infected patients. AIDS. 2011;25(18):2197–208. [PubMed: 21866039]
3.
Dore GJ. The changing therapeutic landscape for hepatitis C. Med J Aust. 2012;196(10):629–32. [PubMed: 22676877]
4.
Hoofnagle JH. Course and outcome of hepatitis C. hepatology. 2002;36(5 Suppl 1):S21–9. [PubMed: 12407573]
5.
Lin C, et al. Processing in the hepatitis C virus E2-NS2 region: identification of p7 and two distinct E2-specific products with different C termini. J Virol. 1994;68(8):5063–73. [PMC free article: PMC236449] [PubMed: 7518529]
6.
Moradpour D, Blum HE. A primer on the molecular virology of hepatitis C. Liver Int. 2004;24(6):519–25. [PubMed: 15566499]
7.
Majeau N, et al. The N-terminal half of the core protein of hepatitis C virus is sufficient for nucleocapsid formation. J Gen Virol. 2004;85(Pt 4):971–81. [PubMed: 15039539]
8.
Brothers SP, et al. Selective and brain penetrant neuropeptide y y2 receptor antagonists discovered by whole-cell high-throughput screening. Mol Pharmacol. 2010;77(1):46–57. [PMC free article: PMC2802430] [PubMed: 19837904]
9.
Terent’ev AO, et al. Facile and selective procedure for the synthesis of bridged 1,2,4,5-tetraoxanes; strong acids as cosolvents and catalysts for addition of hydrogen peroxide to beta-diketones. J Org Chem. 2009;74(9):3335–40. [PubMed: 19298073]
10.
Sakai T, et al. One-Pot C-Arylmethylation of Active Methylene Compounds with Aromatic Aldehydes Induced by a Me3SiCl–NaI–MeCN Reagent. Bull. Chem. Soc. Jpn. 1989;62(12):4072–4074.
11.
Chen Q, et al. Synthesis, antifungal activity and CoMFA analysis of novel 1,2,4-triazolo[1,5-a]pyrimidine derivatives. Eur J Med Chem. 2008;43(3):595–603. [PubMed: 17618711]
12.
Wei W, et al. New small molecule inhibitors of hepatitis C virus. Bioorg Med Chem Lett. 2009;19(24):6926–30. [PMC free article: PMC2783740] [PubMed: 19896376]
13.
Lipinski CA, et al. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46(1–3):3–26. [PubMed: 11259830]
14.
Walters WP. Going further than Lipinski's rule in drug design. Expert Opin Drug Discov. 2012;7(2):99–107. [PubMed: 22468912]
15.
Rishton GM. Nonleadlikeness and leadlikeness in biochemical screening. Drug Discov Today. 2003;8(2):86–96. [PubMed: 12565011]
16.
Benigni R, Bossa C. Mechanisms of chemical carcinogenicity and mutagenicity: a review with implications for predictive toxicology. Chem Rev. 2011;111(4):2507–36. [PubMed: 21265518]
17.
Kota S, et al. A time-resolved fluorescence-resonance energy transfer assay for identifying inhibitors of hepatitis C virus core dimerization. Assay Drug Dev Technol. 2010;8(1):96–105. [PMC free article: PMC2971647] [PubMed: 20035614]
18.
Verdine GL, Walensky LD. The challenge of drugging undruggable targets in cancer: lessons learned from targeting BCL-2 family members. Clin Cancer Res. 2007;13(24):7264–70. [PubMed: 18094406]

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this page (1.8M)

Related information

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...