Abstract
Human cytomegalovirus (CMV) is a ubiquitous β-herpesvirus that establishes latent asymptomatic infections in healthy individuals but can cause serious infections in immunocompromised people, resulting in increased risk of morbidity and mortality. The current FDA-approved CMV drugs target late stages of the CMV life-cycle. While these drugs are effective in most cases, they have serious drawbacks, including poor oral bioavailability, dose-limiting toxicity, and a low barrier to resistance. Given the clinical relevance of CMV-associated diseases, novel therapies are needed. Thus, a novel class of compounds that inhibits the early stages of the CMV life-cycle was identified and found to block infection of different strains in physiologically relevant cell types. This class of compounds, N-arylpyrimidinamine (NAPA), demonstrated potent anti-CMV activity against ganciclovir-sensitive and -resistant strains in in vitro replication assays, a selectivity index >30, and favorable in vitro ADME properties. Mechanism of action studies demonstrated that NAPA compounds inhibit an early step of virus infection. NAPA compounds are specific inhibitors of cytomegaloviruses and do not exhibit anti-viral activity against other herpesviruses. Collectively, we have identified a novel class of CMV inhibitor that effectively limits viral infection and proliferation.
Keywords: Human cytomegalovirus, early-stage infection inhibitors, NAPA compounds, congenital CMV and transplant recipients, high-throughput screening
1. Introduction
Human cytomegalovirus (CMV) is a ubiquitous pathogen with high seroprevalence (~83%) worldwide(Adane and Getawa, 2021). CMV can spread through bodily fluids, during organ transplants, and across the placenta from mother to fetus. Virus proliferation in the immunocompromised leads to increased morbidity and mortality in newborns, organ transplant recipients, AIDS patients, and the elderly. CMV is the leading infectious cause of birth defects, affecting ~0.5-2% of newborns worldwide, with up to 40,000 new cases/year of congenital CMV infection in the United States(Damato and Winnen, 2002; Goderis et al., 2014; Ssentongo et al., 2021). CMV can also exacerbate cardiovascular diseases associated with atherosclerosis (Ji et al., 2012; Lee et al., 2014) and cardiac allograft vasculopathy (Petrakopoulou et al., 2004; Simmonds et al., 2008). Given the patient populations at risk for CMV-associated diseases and the estimated cost-of-illness is $0.4-1.3billion/year for congenital CMV(Grosse et al., 2021)) and ~$4.4billion/year overall(Dove, 2006)), CMV is a significant health challenge requiring the development of a multi-faceted therapeutic strategies.
While several drugs have been approved by the FDA for treatment of CMV, including ganciclovir (GCV), foscarnet (PFA), cidofovir (CDV) and letermovir (LTV), these drugs exhibit high frequencies of drug resistance(Chou, 2008, 2015; Jabs et al., 1998; Weinberg et al., 2003) and can elicit severe side effects(Kenneson and Cannon, 2007). Recently, maribavir (LIVTENCITY™) has been approved for post-transplant CMV disease in patients refractory to ganciclovir, valganciclovir, cidofovir, or foscarnet(Kang, 2022). Approved anti-CMV therapies target the DNA polymerase UL54 (GCV, PFA, and CDV) and the terminase protein UL56 (LTV), both of which are essential in later phases of the CMV life-cycle(Lurain and Chou, 2010). The CMV genome encodes many unexploited potential anti-viral targets for novel therapies. In particular, early stages of the CMV life-cycle mediate several essential functions, such as binding and entry into host cells, membrane fusion, transfer of the genome to the nucleus, and expression of immediate early genes, which are carried out by cellular and viral proteins.
To explore the discovery of novel drugs that act on unexploited targets, we developed a high-content screening (HCS) assay to identify inhibitors of the early stages of the CMV life-cycle, which is driven by proteins not targeted by existing drugs. The assay is comprised of a CMV reporter virus (AD169IE2/YFP) that expresses an IE2:yellow-fluorescent protein (YFP) fusion protein(Gardner et al., 2015). We screened >112,000 compounds using AD169IE2/YFP-infected fibroblasts(Gardner et al., 2015) and found several distinct compounds that effectively limit CMV infection. One of these, a N-arylpyrimidinamine (NAPA), exhibited potent anti-CMV activity with limited cytotoxicity. The current study describes NAPA compounds as effective inhibitors of in infection and dissemination.
2. Materials and Methods
2.1. Cells and viruses
HFF (ATCC#SCRC-1041), MRC-5 (ATCC #CCL-171), NHDF (ATCC #PSC-201-010) fibroblast cells and ARPE-19 human retinal epithelial cells (ATCC #CRL-2302) were cultured as described(Parsons et al., 2022). CMV strains AD169 (ATCC:VR-538), AD169IE2-YFP (gift from Dr. Leor Weinberger, Gladstone Inst/UCSF), AD169 (BADrUL131-C4) containing the UL131 open reading frame of the CMV strain TR that expresses EGFP (AD169R)(Wang and Shenk, 2005) (gift from Dr. Thomas Shenk, Princeton U.), and TB40/E and TB40/EUL32-EGFP (Sampaio et al., 2005) (gifts from Dr. Christian Sinzger, Ulm U.) were propagated and titered as described (Gardner et al., 2013).
2.2. High-content screening
The optimized screening assay was performed using 384-well assay plates (Corning #COR-3712) seeded with 3000 HFF cells (40μL DMEM) for 24 hours. The next day, 0.2μL compound (stock: 2.5mM, final: 10μM) from screening libraries was added to each well in columns 3-22 using a Caliper robot with a pin tool array. DMSO (0.2μl) was added to control columns 1-2 and 23-24. After incubation at 37°C for 1 hour, 10μL diluted AD169IE2-YFP (multiplicity of infection, MOI=1) was added to wells in columns 1-22 and spin-infected (2000 rpm for 30min at room temperature (RT)) to achieve 30-50% infection. Columns 23-24 were uninfected controls. After 17 hours post-infection (hpi) at 37°C, cells were fixed (7.4% formaldehyde final in PBS) for 30 min at RT, then washed with PBS using a BioTek Elisa plate washer prior to adding Hoechst 33342 (30μL, 25μg/mL). Cells were washed with PBS, and 30μL PBS was added to each well prior to imaging with a Cytation 5 Cell Imaging Multi-Mode plate reader (BioTek) to quantify YFP+ and H33342-stained nuclei in 5 separate fields of view. The numbers of YFP+ and total nuclei (H33342 stained) were normalized to average counts of DMSO-treated controls (columns 1-2). Compounds that reduced YFP+ nuclei by ≥70% but had a minimal effect (≤25% reduction) on the number of total nuclei (H33342 stained) were scored as “primary hits.” Primary hits were confirmed (four technical replicates) using the screening assay, and the average reduction of YFP+ and total nuclei as compared to negative controls was determined. We screened a total of 112,197 small molecules from commercially available compound collections, comprised of compounds purchased from MicroSource Discovery (Gaylordsville, CT), Chembridge (San Diego, CA), Biomol GmbH (Hamburg, Germany), Life Chemicals (Niagara-on-the-Lake, Canada), ChemDiv (San Diego, CA), and TimTec (Newark, DE).
2.3. Dose response assay
The half-maximal inhibitory concentration (IC50) of primary hits and analogs was determined by measuring infection with a two-fold compound dilution series (0-100 μM) using the screening assay protocol. Dose-response data was normalized to untreated cells, and the IC50 was determined using a four-parameter curve-fitting algorithm (GraphPad Prism). Each assay was performed over three replicates and repeated at least once. The reported value is the average of at least six technical replicates.
2.4. Compound evaluation/assay funnel
NAPA analogs were evaluated for solubility, cytotoxicity, and predicted ADME properties using the following assays. Each assay was performed with three replicates repeated at least once. The reported value is the average of at least six technical replicates. Solubility. The maximum aqueous solubility of analogs was determined by nephelometry using published methods(Bevan and Lloyd, 2000). Cytotoxicity. We determined the concentration of each compound that produces a half-maximal decrease in cytotoxicity (CC50) of Hela cells exposed to inhibitor (0.4-300μM) using the vital stain MTS (Abcam, ab197010). The dose-response data was normalized to untreated cells, and the CC50 was determined using a four-parameter curve-fitting algorithm (GraphPad Prism) (Butler et al., 2007). A CellTiter Glo Luminescent Assay (Promega Inc, Madison, WI) was performed on MBX-4992-treated NHDF cells based on manufacture’s instructions. Mouse liver microsome stability. To examine the potential for first-pass metabolism of analogs in the liver, the half-life (T1/2) was assessed in the presence of mouse liver microsome preparations (XenoTech) and of NADPH (Kuhnz and Gieschen, 1998). Cytochrome 450 (CYP450 3A4) inhibition. Inhibition of CYP450 3A4 by NAPA analogs was measured using commercially available kits for CYP3A4 activity (BD Gentest Corp, Woburn, MA).
MBX-4992 compound synthesis.
To a solution of furan-2-carboxylic acid (1.0eq), N1-(2-methyl-6-(pyrrolidin-1-yl)pyrimidin-4-yl)benzene-1,4-diamine (1.2eq), and HBTU (1.1eq) in DMF (0.3mM) was added diisopropylethylamine (1.3eq) and stirred at 25°C for 72hr; this mixture was purified on prep-HPLC and freeze-dried to light yellow solid. Compounds were resuspended in DMSO (final: 50mM).
2.5. Analysis of inhibitory properties of the NAPA compounds
Cells (104/well, 96-well plates (Greiner) plated overnight were pretreated for 1 hour at 37°C with the NAPA compounds or DMSO in triplicate prior to virus infection (MOI that yields ~20-30% infection). Virus infection was evaluated at 24hpi using a rabbit anti-IE1 antibody (Tortorella lab) as described (Stein et al., 2019). The relative percent infection of NAPA-treated cells was determined using DMSO-treated cells as 100% infection. The half-maximal inhibitory concentration (IC50) for each compound was calculated using a nonlinear four-parameter curve fitting algorithm (GraphPad Prism). The immunoblot assay was performed as described(Stein et al., 2019). The mechanistic studies and herpes virus inhibition (Keith et al., 2018; Hartline et al., 2018) protocols are described in Supplemental Information.
3. Results
3.1. High-content screen (HCS) for early-stage CMV replication inhibitors
We utilized the reporter virus AD169IE2-YFP(Gardner et al., 2015) to screen a library of discreet synthetic small molecules for compounds that reduce expression of the IE2/yellow fluorescent protein (YFP) (IE2-YFP) chimera. The IE1 and IE2 CMV gene products are translated within 3-6hpi and are key markers of the early stages of CMV replication(Cherrington and Mocarski, 1989; Meier and Stinski, 1997). The IE2-YFP chimera exhibits expression kinetics and nuclear localization like that of IE2 (Teng et al., 2012). Expression of the nucleus-localized IE2-YFP can be readily quantified using a confocal fluorescence microscopic plate reader. The AD169IE2-YFP reporter virus thus provides the basis for the HCS assay for identification of inhibitors to the early stages of CMV’s life cycle.
After optimization of the AD169IE2-YFP reporter assay (average Z’ factor ~0.5), we screened a library of 112,197 small molecules from commercially-available compound collections and quantified the percentage of nuclei containing YFP (YFP+) as compared to total nuclei (H33342-stained) using a confocal microscopic plate reader. Compounds that reduced YFP+ nuclei by ≥75% but had a minimal effect (≤25% reduction) on the number of total nuclei (H33342 stained) were scored as “primary hits.” The workflow for screening and evaluation of primary hits is summarized in Fig. 1A. We identified 300 primary hits, which were further evaluated using an assay funnel designed to prioritize compounds based on antiviral activity, specificity, and drug-like properties, ultimately uncovering two potent, selective, drug-like candidates for hit-to-lead optimization that were not Pan Assay Interference compounds (PAINS)(Baell and Holloway, 2010; Capuzzi et al., 2017; Dahlin and Walters, 2016). The primary hit-to-lead series selected was an N-arylpyrimidinamine (NAPA) compound referred to as MBXC-4302 (Fig. 1B), in which secondary validation assays demonstrated that this compound exhibited potent anti-CMV activity (IC50=3.2μM) with limited cytotoxicity (CC50 > 100μM).
Figure 1: Screening strategy to identify early stage CMV inhibitors.

(A) The schematic and criteria of the high-content screen to identify inhibitors of CMV infection using AD169IE2-YFP/fibroblast infection. (B) Structure of MBXC-4302.
To evaluate the potency of MBXC-4302 in blocking CMV infection of physiologically relevant cell types, infectivity assays were performed in human fibroblasts (NHDF) and epithelial (ARPE-19) cells. AD169R and TB40/E (MOI=0.5)-infected ARPE-19 and NHDF cells treated with MBXC-4302 were analyzed for virus infection at 24hpi (Materials and Methods). MBXC-4302 inhibited AD169R-infection of NHDF and ARPE-19 cells with IC50 values of 3 μM and 5.8 μM respectively (Fig. 2A and C). The MBXC-4302 IC50 values for TB40/E-infected NHDF and ARPE-19 cells were ~4.5 μM for both cell types (Fig. 2B and D). These data demonstrate that MBXC-4302 broadly inhibits CMV infection of physiologically relevant cell types.
Figure 2. MBXC-4302 inhibits CMV infection.

CMV strains AD169R (A and C) and TB40/E (B and D) infection (MOI=0.2) of fibroblasts (A-B) and ARPE-19 cells (C-D) pretreated with 0, 1, 5, and 20 μM MBXC-4302 were assessed 24hpi based on GFP fluorescence (A,C) or IE1 (B,D) expression by an anti-IE1 immunostain. Relative % infection was determined using DMSO-treated cells as 100% infection. The data points are the averages of three replicates and the error bars are the standard deviation. Statistical tests were performed using one-way ANOVA with multiple comparisons to DMSO-treated cells as a control and a Dunnett’s post-test; ****, p<0.0001.
3.2. Preliminary structure activity relationships of the NAPA series
Preliminary structure activity relationship (SAR) studies of the NAPA series demonstrated responsiveness in various biological activities to structural changes within the five zones of MBXC-4302 defined in Table 1. Modulations in Zone 1 were found to alter compounds’ potency and selectivity. Switching from an heteroaromatic pyrazole in Zone 1 of MBXC-4302 (entry 7) to an aromatic secondary amine (entries 8-10) led to a 10fold increase in potency, highlighting a potential hydrophobic binding pocket. This is corroborated by the decrease in potency when the aryl ring was replaced by a polar acyclic ethylamine (entry 1), resulting in potency similar to MBXC-4302. Increasing lipophilicity (entries 2-5) also improved potency of inhibition, which was restored to sub-micromolar levels with the piperidine substitution (entry 5). Zone 1 analogs varied widely in CC50 values, with a selectivity index ranging from 7 to >100, consistent with a specific antiviral mechanism independent of any cytotoxic pathway. Similarly, tertiary amines (entries 2-6) were highly selective relative to the phenyl analogs. With regards to analogs with modifications in Zone 2 (entries 8-10), MBXC-4297 had a selectivity index of >50, while the other two isomeric pyrimidine ligands had index values of <20, suggesting the electronics of the pyrimidine moiety plays a role in selectivity. In Zone 4, addition of a sulfone linker (entry 11) was not tolerated, but a urea linker (entry 12) improved potency, suggesting a potential hydrogen-bonding site in this binding pocket. Finally, alterations in Zone 5 from a furan to a thiophene or a phenyl ring (entries 13, 14) resulted in complete loss of activity, suggesting the need for further optimization with regards to this Zone.
Table 1.
Structure Activity Relationships of the NAPA Series
|
HeLa cell lines;
Maximum solubility in water determined by nephlometry;
Murine liver microsomal stability;
% remaining after 60 min at 37 °C in the presence of NADPH.
Analogs were further evaluated for solubility and in vitro ADME properties. The basic amines (entries 1-6) are highly soluble compared to other analogs, and the majority of compounds were stable in the presence of mouse liver microsomes, supporting the analysis of effective analogs in future in vivo studies. While some compounds exhibited CYP3A4 inhibition, modulation of Zone 1 provides a potential solution to overcome or avoid drug-drug interactions. Based on the data gathered from in vitro assays and a cytotoxicity study in NHDFs (Fig. S1A), MBX-4992 (entry 4) was found to exhibit favorable parameters, and was therefore selected for further characterization as a novel CMV inhibitor.
3.3. MBX-4992 broadly inhibits CMV infection
To investigate the broad inhibitory potential of the NAPA analog MBX-4992, NHDF and ARPE-19 cells were treated with MBX-4992 (1, 5, and 10μM) followed by infection with AD169R and TB40/E strains due to their wide tropism. Cells were then examined for infection at 24hpi, revealing IC50 values ranging from 1.9-4.8 μM (Figs. 3A–D). To confirm that MBX-4992 reduces expression of the CMV IE1 protein during infection, we examined IE1 protein levels in AD169R-infected NHDFs treated with MBX-4992 (Fig. 3E). A decrease in IE1 expression was observed upon treatment with MBX-4992 (Fig. 3E, lanes 1-4). As expected, treatment with ganciclovir (GCV) did not impact IE1 expression, while convallatoxin (CVT), a compound targeting the Na+/K+ ATPase(Cohen et al., 2016) inhibited IE1 expression (Fig. 3E, lanes 5-6). Analysis of IE1 expression at 2 days post-infection further validated that MBX-4992 prevents virus infection (Fig. S1B). These findings demonstrate that MBX-4992 inhibits CMV infection.
Figure 3. NAPA compound MBX-4992 significantly limits CMV infection in vitro.

CMV strains AD169R (A,C) and TB40/E (B,D) infection (MOI=0.2 or 0.4, respectively) of fibroblasts (A-B) and ARPE-19 cells (C-D) pre-treated with 0, 1, 5, and 20 μM MBX-4992 were assessed 24hpi based on GFP fluorescence (A,C) or anti-IE1 immunostain (B,D). Relative % infection was determined using DMSO-treated cells as 100% infection. The data points are the averages of three replicates with error bars indicating standard deviation. Statistical tests were performed using one-way ANOVA with multiple comparisons to DMSO-treated cells as a control and a Dunnett’s post-test; ***, p<0.001; ****, p<0.0001. (E) Total cell lysates from AD169R-infected (MOI=0.2) NHDF cells pretreated with DMSO, MBX-4992 (1-10 μM), ganciclovir (5 μM) or convallotoxin (0.01 μM) collected at 24hpi were subjected to Western blot analysis using a rabbit anti-IE1 antibody (Lanes 1-6) and an anti-GAPDH antibody (Lanes 7-12) as a loading control. The molecular weight markers and respective proteins are indicated.
3.4. MBX-4992 inhibits several herpes viruses
To determine the spectrum of antiviral activity of MBX-4992, MBX-4992 was evaluated for inhibition of a panel of herpes viruses (Table 2). MBX-4992 exhibited potent antiviral activity against human CMV (IC50=1.7μM) that was comparable to that of GCV (IC50=0.9μM). MBX-4992 was also effective against a GCV-resistant strain of human CMV (IC50=1.8μM). Interestingly, the compound exhibited modest activity against murine CMV and was highly active against HHV-8. However, MBX-4992 did not exhibit activity against any additional herpes viruses or other viral pathogens, such as hepatitis B, dengue, influenza A H1N1, respiratory syncytial, yellow fever, or Zika virus (data not shown). Thus, antiviral activity of MBX-4992 is the most sensitive against cytomegaloviruses.
Table 2.
Spectrum of anti-viral activity of MBX-4992 against herpes viruses.
|
|
|||||||
|---|---|---|---|---|---|---|---|
| MBX-4992 (μM) | GCV (μM) | ||||||
|
| |||||||
| Virus | Strain | Cell line | Assay | IC50 | CC50 | SI | IC50* |
| EBV | Akata | Akata | qPCR | 30.9 | >100 | >3 | ND |
| GpCMV | 22122 | GP Lung | qPCR | >30 | 118.8 | <4 | ND |
| HCMV | AD169 | HFF | CPE | 1.7 | >150 | >88 | 0.9 |
| HCMV | GDGr K17 (GCVR) | HFF | CPE | 1.8 | >151 | >83 | 10.4 |
| HHV-6B | Z29 | MOLT-3 | qPCR | 43.7 | >100 | >2 | ND |
| HHV-8 | BCBL-1 | BCBL-1 | qPCR | 10 | >100 | >10 | ND |
| HSV-1 | E-377 | HFF | CPE | >30 | 60.6 | <2 | ND |
| HSV-2 | G | HFF | CPE | >150 | >150 | 1 | ND |
| MCMV | Smith | MEF | qPCR | 4.5 | >150 | >33 | 0.2 |
| VZV | Ellen | HFF | CPE | >30 | 139.2 | <5 | ND |
Abbreviations: EBV, Epstein-Barr virus; GpCMV, Guinea pig cytomegalovirus; HCMV, Human cytomegalovirus; HHV-6, Human herpes virus 6B; HHV-8, Human herpes virus 8; HSV-1, Herpes simplex virus 1; HSV-2, Herpes simplex virus 2; MCMV, Murine cytomegalovirus; VZV, Varicella-Zoster virus; CPE, cytopathic effects; qPCR, quantitative PCR; HFF, Human foreskin fibroblasts; MEF, mouse embryonic fibroblasts.
3.5. MBX-4992 targets an early stage of infection
A time-of-addition assay was performed to elucidate the step of CMV infection targeted by MBX-4992 (Fig. 4A). MBX-4992 (10 μM) was added at times relative to infection (−60, 0, 15, 30, 60, and 90 mpi). MBX-4992 was most effective at preventing infection when introduced between −60 min prior to infection to 30 mpi. The inhibitory activity of MBX-4992 began to diminish at 60 mpi and further at 90 mpi. A similar study in ARPE-19 cells demonstrated similar kinetics to loss of inhibition at 60 and 90 mpi (Fig. S2A). MBX-4992 likely inhibits an early-step of infection based on CMV expression of IE1/2 and localization of pp65 within 2hpi(Bodaghi et al., 1999; Stenberg, 1996)}. Further, virus infection was evaluated using a TB40/E variant expresses the chimeric capsid protein UL32 with EGFP (TB40/EUL32-EGFP)(Sampaio et al., 2005) (Fig. S2B). We observed a dramatic decrease in TB40/EUL32-EGFP particles after 2hpi in MBX-4992 treated cells compared to no treatment. Also, the GFP signal from the virus particles appears to localize at the cell membrane. The controls anti-gH neutralizing mAb 4E7(Parsons et al., 2022) and heparin(Kapoor et al., 2020) that block virus entry also displayed limited the virus particles in the cells. These studies are consistent with the paradigm that MBX-4992 is targeting an early-step of virus infection.
Figure 4. NAPA compounds target an early, post-attachment step.

(A) MBX-4992 (10 μM) was added along a time course following infection with AD169 virus (MOI=0.2) from −60 mins prior to infection to 90 mpi on NHDF cells. Infection at 24 hours was quantified with anti-IE1 immunostain and untreated virus-infected cells was normalized to 100%. (B) NHDFs were preincubated with DMSO, MBX-4992, or heparin for 30 mins at 37°C followed by the addition of AD169R (MOI=0.2) for 30 minutes at 4°C. Cell media was either unchanged (No Wash) or removed (Wash) and replaced with media containing DMSO, MBX-4992, or heparin. (C) AD169R (MOI=0.2) pre-incubated with DMSO, MBX-4992 (1 μM), or heparin (50μg/ml) for 30 mins at 37°C was added to fibroblasts for 30 min at 4°C. Cells were not washed (No Wash) or media was removed (Wash) and replaced with media containing either DMSO, MBX-4992, or heparin shifting cells to 37°C. Virus infection was quantified at 24hpi by GFP fluorescence with DMSO-treated virus normalized to 100%. The data points are the averages of three replicates with error bars. Statistical tests were performed using ordinary one-way ANOVA with multiple comparisons to DMSO-treated cells as a control and a Dunnett’s post-test: *, p<0.05; ****, p<0.0001.
Does pretreating cells with MBX-4922 limit infection? To address this question, cells were pre-treated with MBX-4992 followed by the addition of AD169R 30 mins at 4°C allowing the virus to bind. The cells were not washed or washed/replaced with media containing DMSO or MBX-4992 (10μM) followed by analysis of virus infection at 24hpi in which DMSO-treated cells was defined as 100% (Fig. 4B). Heparin treatment (50μg/ml) was used an inhibition control. MBX-4992 and heparin significantly inhibited virus during infection. Yet, pre-incubation of cells with MBX-4992 and its subsequent removal did not significantly virus infection suggesting MBX-4992 requires to be present at time of infection. We next addressed whether MBX-4992 irreversibly targets the virion (Fig. 4C). MBX-4992 (10μM) pre-incubated with CMV at 37°C for 30 min and then added to cells for 30 min at 4°C. The cells were not washed or washed/replaced with media containing DMSO or MBX-4992 (10μM). Virus infection was evaluated at 24hpi in which DMSO treated cells was normalized to 100% (Fig. 4C). Heparin treatment (50μg/ml) was used an inhibition control. Consistent with previous results, MBX-4992 or heparin treatment significantly decreased infection by >75% under all conditions. Interestingly, virus pre-incubated with MBX-4992 and then washed decreased virus infection by ~25% suggesting MBX-4992 has a slight impact on the virus binding. Together, the data connote that MBX-4992 is the most effective when incubated with virus and cells during infection.
4. Discussion
The complex life-cycle of CMV is reflected in the size of its genome (~235 kb), containing at least ~180 ORFs, 45 of which are essential for replication in fibroblasts (Dunn et al., 2003). Accordingly, there are many unexploited essential viral targets for anti-CMV drug discovery. We identified a novel series of N-arylpyrimidinamine (NAPA) compounds that broadly inhibit infection of diverse strains (Fig. 2). Further, NAPA compounds limited virus proliferation in vitro indicating that the compounds are effective inhibitors of virus dissemination (Table 2). Importantly, the NAPA compounds target early steps of the CMV life-cycle, likely impacting a post-binding step (Fig. 5). These inhibitors represent novel CMV inhibitors that target a step of the CMV life-cycle that differs from approved late-stage drug inhibitors of CMV.
The NAPA inhibitor MBX-4992 is specific for CMV replication based on its IC50 values for HCMV (~1.8 μM) and MCMV (4.5 μM). While also demonstrating activity against HHV-8, MBX-4992 showed weak or no inhibition for EBV, GpCMV, HHV-6B, HSV-1, HSV-2, and VZV replication, supporting its relative specificity for cytomegaloviruses. Interestingly, the NAPA compounds have some structural similarities to previously identified benzyl-N’-arylthiourea inhibitors of CMV(Bloom et al., 2003; Bloom et al., 2004; Jones et al., 2004) with regards to Zones 3 and 4 of the NAPA compounds. The arylthiourea compounds target the HCMV envelope protein gB based on the generation of drug-resistant mutants of gB(Jones et al., 2004) and a structure of gB complexed with the arylthiourea analog WAY-174865(Liu et al., 2021). However, several critical gB residues of human CMV (Y155 and D714) that interact with the arylthiourea compound are not conserved in mouse CMV, a virus sensitive to the NAPA compound (Table 2). Despite these differences, the structural similarities of the arylthiourea compounds and the NAPA compounds are quite intriguing, indicating that NAPA compounds may indeed target an early step of infection.
The development of alternative CMV therapies is being pursued by modification of current drugs, drug repurposing, and screening of compound libraries for inhibitors that target different steps of the virus life-cycle(Bogner et al., 2021; Mercorelli et al., 2011). Potential CMV inhibitors include the antimalarial drugs artemisinins(Arav-Boger et al., 2010; Kaptein et al., 2006), which bind to the filament protein vimentin to prevent HCMV replication(Roy et al., 2020). In fact, an artemisinin derivative, artemisone, demonstrated antiviral activity against the early phase of virus replication(Oiknine-Djian et al., 2018). Other compounds that impact early steps of the life-cycle are valspodar(Parsons et al., 2021), dispirotripiperazines, tricyclic molecules with two quaternary positively-charged nitrogen atoms (or “spiro atoms”), modified lectins that block entry of herpes viruses(Egorova et al., 2021; Zoepfl et al., 2021;Lloyd et al., 2022)), and a furan-based molecule(Kapoor et al., 2020). In addition, marine sulfated glycans limited CMV and adenovirus infection by targeting viral attachment and entry(Zoepfl et al., 2021). These studies exemplify the diversity of inhibitors that target different cellular and viral entry factors and show promise as novel CMV therapeutics.
5. Conclusions
Human cytomegalovirus (CMV) has a high seroprevalence rates of 60-90% within the population. Virus proliferation significantly increases the morbidity and mortality of immunocompromised individuals, such as newborns and organ transplant recipients. The development of novel therapeutics that target different steps of the viral life cycle to limit virus propagation and dissemination would provide therapeutic for treating CMV-related diseases. A high-content screening assay using a CMV reporter assay identified an N-arylpyrimidinamine (NAPA) compound with anti-CMV infection activity and limited cytotoxicity. Thus, NAPA compounds represent novel CMV early-stage infection inhibitors that can potentially be developed into effective CMV therapeutics.
Supplementary Material
Highlights.
NAPA compounds target an early step of human cytomegalovirus infection
NAPA compounds inhibit cytomegalovirus infection and proliferation
NAPA compounds represent a novel class of cytomegalovirus inhibitors
Acknowledgements
This work was supported in part by NIH grants AI139258, AI147632, AI113971, AI139258, AG059319, and T32AI007647. We thank Drs. Weinberger (Gladstone Inst/UCSF), Shenk Princeton U.), and Sinzger (Ulm U.) for viruses. The evaluation of antiviral activity against a diverse panel of herpes viruses was funded by NIAID Preclinical Services. and by federal funds from the NIAID, NIH, Department of Health and Human Services (HHSN272201100016I (MNP)).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- Adane T, Getawa S, 2021. Cytomegalovirus seroprevalence among blood donors: a systematic review and meta-analysis. J Int Med Res 49, 3000605211034656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arav-Boger R, He R, Chiou CJ, Liu J, Woodard L, Rosenthal A, Jones-Brando L, Forman M, Posner G, 2010. Artemisinin-derived dimers have greatly improved anti-cytomegalovirus activity compared to artemisinin monomers. PLoS One 5, e10370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baell JB, Holloway GA, 2010. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J Med Chem 53, 2719–2740. [DOI] [PubMed] [Google Scholar]
- Bevan CD, Lloyd RS, 2000. A high-throughput screening method for the determination of aqueous drug solubility using laser nephelometry in microtiter plates. Anal Chem 72, 1781–1787. [DOI] [PubMed] [Google Scholar]
- Bloom JD, DiGrandi MJ, Dushin RG, Curran KJ, Ross AA, Norton EB, Terefenko E, Jones TR, Feld B, Lang SA, 2003. Thiourea inhibitors of herpes viruses. Part 1: bis-(aryl)thiourea inhibitors of CMV. Bioorg Med Chem Lett 13, 2929–2932. [DOI] [PubMed] [Google Scholar]
- Bloom JD, Dushin RG, Curran KJ, Donahue F, Norton EB, Terefenko E, Jones TR, Ross AA, Feld B, Lang SA, DiGrandi MJ, 2004. Thiourea inhibitors of herpes viruses. Part 2: N-Benzyl-N’-arylthiourea inhibitors of CMV. Bioorg Med Chem Lett 14, 3401–3406. [DOI] [PubMed] [Google Scholar]
- Bodaghi B, Slobbe-van Drunen ME, Topilko A, Perret E, Vossen RC, van Dam-Mieras MC, Zipeto D, Virelizier JL, LeHoang P, Bruggeman CA, Michelson S, 1999. Entry of human cytomegalovirus into retinal pigment epithelial and endothelial cells by endocytosis. Invest Ophthalmol Vis Sci 40, 2598–2607. [PubMed] [Google Scholar]
- Bogner E, Egorova A, Makarov V, 2021. Small Molecules-Prospective Novel HCMV Inhibitors. Viruses 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butler MM, Lamarr WA, Foster KA, Barnes MH, Skow DJ, Lyden PT, Kustigian LM, Zhi C, Brown NC, Wright GE, Bowlin TL, 2007. Antibacterial activity and mechanism of action of a novel anilinouracil-fluoroquinolone hybrid compound. Antimicrob Agents Chemother 51, 119–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capuzzi SJ, Muratov EN, Tropsha A, 2017. Phantom PAINS: Problems with the Utility of Alerts for Pan-Assay INterference CompoundS. J Chem Inf Model 57, 417–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cherrington JM, Mocarski ES, 1989. Human cytomegalovirus ie1 transactivates the alpha promoter-enhancer via an 18-base-pair repeat element. Journal of virology 63, 1435–1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chou S, 2008. Cytomegalovirus UL97 mutations in the era of ganciclovir and maribavir. Rev Med Virol 18, 233–246. [DOI] [PubMed] [Google Scholar]
- Chou S, 2015. Rapid In Vitro Evolution of Human Cytomegalovirus UL56 Mutations That Confer Letermovir Resistance. Antimicrob Agents Chemother 59, 6588–6593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen T, Williams JD, Opperman TJ, Sanchez R, Lurain NS, Tortorella D, 2016. Convallatoxin-Induced Reduction of Methionine Import Effectively Inhibits Human Cytomegalovirus Infection and Replication. J Virol 90, 10715–10727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dahlin JL, Walters MA, 2016. How to Triage PAINS-Full Research. Assay Drug Dev Technol 14, 168–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Damato EG, Winnen CW, 2002. Cytomegalovirus infection: perinatal implications. J Obstet Gynecol Neonatal Nurs 31, 86–92. [DOI] [PubMed] [Google Scholar]
- Dove A, 2006. A long shot on cytomegalovirus, The Scientist. Richard Gallagher, Philadelphia, PA, pp. 40–45. [Google Scholar]
- Dunn C, Chalupny NJ, Sutherland CL, Dosch S, Sivakumar PV, Johnson DC, Cosman D, 2003. Human cytomegalovirus glycoprotein UL16 causes intracellular sequestration of NKG2D ligands, protecting against natural killer cell cytotoxicity. J Exp Med 197, 1427–1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Egorova A, Bogner E, Novoselova E, Zorn KM, Ekins S, Makarov V, 2021. Dispirotripiperazine-core compounds, their biological activity with a focus on broad antiviral property, and perspectives in drug design (mini-review). Eur J Med Chem 211, 113014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardner TJ, Bolovan-Fritts C, Teng MW, Redmann V, Kraus TA, Sperling R, Moran T, Britt W, Weinberger LS, Tortorella D, 2013. Development of a high-throughput assay to measure the neutralization capability of anti-cytomegalovirus antibodies. Clinical and vaccine immunology : CVI 20, 540–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardner TJ, Cohen T, Redmann V, Lau Z, Felsenfeld D, Tortorella D, 2015. Development of a high-content screen for the identification of inhibitors directed against the early steps of the cytomegalovirus infectious cycle. Antiviral Res 113, 49–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goderis J, De Leenheer E, Smets K, Van Hoecke H, Keymeulen A, Dhooge I, 2014. Hearing loss and congenital CMV infection: a systematic review. Pediatrics 134, 972–982. [DOI] [PubMed] [Google Scholar]
- Grosse SD, Dollard SC, Ortega-Sanchez IR, 2021. Economic assessments of the burden of congenital cytomegalovirus infection and the cost-effectiveness of prevention strategies. Semin Perinatol 45, 151393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hartline CB, Keith KA, Eagar J, Harden EA, Bowlin TL, Prichard MN, 2018. A standardized approach to the evaluation of antivirals against DNA viruses: Orthopox-, adeno-, and herpesviruses. Antiviral Res 159, 104–112. [DOI] [PubMed] [Google Scholar]
- Jabs DA, Enger C, Dunn JP, Forman M, 1998. Cytomegalovirus retinitis and viral resistance: ganciclovir resistance. CMV Retinitis and Viral Resistance Study Group. J Infect Dis 177, 770–773. [DOI] [PubMed] [Google Scholar]
- Ji YN, An L, Zhan P, Chen XH, 2012. Cytomegalovirus infection and coronary heart disease risk: a meta-analysis. Mol Biol Rep 39, 6537–6546. [DOI] [PubMed] [Google Scholar]
- Jones TR, Lee SW, Johann SV, Razinkov V, Visalli RJ, Feld B, Bloom JD, O’Connell J, 2004. Specific inhibition of human cytomegalovirus glycoprotein B-mediated fusion by a novel thiourea small molecule. J Virol 78, 1289–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang C, 2022. Maribavir: First Approval. Drugs 82, 335–340. [DOI] [PubMed] [Google Scholar]
- Kapoor A, Ghosh AK, Forman M, Hu X, Ye W, Southall N, Marugan J, Keyes RF, Smith BC, Meyers DJ, Ferrer M, Arav-Boger R, 2020. Validation and Characterization of Five Distinct Novel Inhibitors of Human Cytomegalovirus. J Med Chem 63, 3896–3907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaptein SJ, Efferth T, Leis M, Rechter S, Auerochs S, Kalmer M, Bruggeman CA, Vink C, Stamminger T, Marschall M, 2006. The anti-malaria drug artesunate inhibits replication of cytomegalovirus in vitro and in vivo. Antiviral Res 69, 60–69. [DOI] [PubMed] [Google Scholar]
- Keith KA, Hartline CB, Bowlin TL, Prichard MN, 2018. A standardized approach to the evaluation of antivirals against DNA viruses: Polyomaviruses and lymphotropic herpesviruses. Antiviral Res 159, 122–129. [DOI] [PubMed] [Google Scholar]
- Kenneson A, Cannon MJ, 2007. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol 17, 253–276. [DOI] [PubMed] [Google Scholar]
- Kuhnz W, Gieschen H, 1998. Predicting the oral bioavailability of 19-nortestosterone progestins in vivo from their metabolic stability in human liver microsomal preparations in vitro. Drug Metab Dispos 26, 1120–1127. [PubMed] [Google Scholar]
- Lee YL, Liu CE, Cho WL, Kuo CL, Cheng WL, Huang CS, Liu CS, 2014. Presence of cytomegalovirus DNA in leucocytes is associated with increased oxidative stress and subclinical atherosclerosis in healthy adults. Biomarkers 19, 109–113. [DOI] [PubMed] [Google Scholar]
- Liu Y, Heim KP, Che Y, Chi X, Qiu X, Han S, Dormitzer PR, Yang X, 2021. Prefusion structure of human cytomegalovirus glycoprotein B and structural basis for membrane fusion. Sci Adv 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lloyd MG, Liu D, Legendre M, Markovitz DM, Moffat JF, 2022. H84T BanLec has broad spectrum antiviral activity against human herpesviruses in cells, skin, and mice. Sci Rep 12, 1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lurain NS, Chou S, 2010. Antiviral drug resistance of human cytomegalovirus. Clinical microbiology reviews 23, 689–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meier JL, Stinski MF, 1997. Effect of a modulator deletion on transcription of the human cytomegalovirus major immediate-early genes in infected undifferentiated and differentiated cells. Journal of virology 71, 1246–1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mercorelli B, Lembo D, Palu G, Loregian A, 2011. Early inhibitors of human cytomegalovirus: state-of-art and therapeutic perspectives. Pharmacology & therapeutics 131, 309–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oiknine-Djian E, Weisblum Y, Panet A, Wong HN, Haynes RK, Wolf DG, 2018. The Artemisinin Derivative Artemisone Is a Potent Inhibitor of Human Cytomegalovirus Replication. Antimicrob Agents Chemother 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parsons AJ, Cohen T, Schwarz TM, Stein KR, Ophir SI, Casado JP, Tortorella D, 2021. Valspodar limits human cytomegalovirus infection and dissemination. Antiviral Res 193, 105124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parsons AJ, Ophir SI, Duty JA, Kraus TA, Stein KR, Moran TM, Tortorella D, 2022. Development of broadly neutralizing antibodies targeting the cytomegalovirus subdominant antigen gH. Commun Biol 5, 387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrakopoulou P, Kubrich M, Pehlivanli S, Meiser B, Reichart B, von Scheidt W, Weis M, 2004. Cytomegalovirus infection in heart transplant recipients is associated with impaired endothelial function. Circulation 110, II207–212. [DOI] [PubMed] [Google Scholar]
- Roy S, Kapoor A, Zhu F, Mukhopadhyay R, Ghosh AK, Lee H, Mazzone J, Posner GH, Arav-Boger R, 2020. Artemisinins target the intermediate filament protein vimentin for human cytomegalovirus inhibition. J Biol Chem 295, 15013–15028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampaio KL, Cavignac Y, Stierhof YD, Sinzger C, 2005. Human cytomegalovirus labeled with green fluorescent protein for live analysis of intracellular particle movements. J Virol 79, 2754–2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simmonds J, Fenton M, Dewar C, Ellins E, Storry C, Cubitt D, Deanfield J, Klein N, Halcox J, Burch M, 2008. Endothelial dysfunction and cytomegalovirus replication in pediatric heart transplantation. Circulation 117, 2657–2661. [DOI] [PubMed] [Google Scholar]
- Ssentongo P, Hehnly C, Birungi P, Roach MA, Spady J, Fronterre C, Wang M, Murray-Kolb LE, Al-Shaar L, Chinchilli VM, Broach JR, Ericson JE, Schiff SJ, 2021. Congenital Cytomegalovirus Infection Burden and Epidemiologic Risk Factors in Countries With Universal Screening: A Systematic Review and Meta-analysis. JAMA Netw Open 4, e2120736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stein KR, Gardner TJ, Hernandez RE, Kraus TA, Duty JA, Ubarretxena-Belandia I, Moran TM, Tortorella D, 2019. CD46 facilitates entry and dissemination of human cytomegalovirus. Nat Commun 10, 2699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stenberg RM, 1996. The human cytomegalovirus major immediate-early gene. Intervirology 39, 343–349. [DOI] [PubMed] [Google Scholar]
- Teng MW, Bolovan-Fritts C, Dar RD, Womack A, Simpson ML, Shenk T, Weinberger LS, 2012. An endogenous accelerator for viral gene expression confers a fitness advantage. Cell 151, 1569–1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang D, Shenk T, 2005. Human cytomegalovirus virion protein complex required for epithelial and endothelial cell tropism. Proceedings of the National Academy of Sciences of the United States of America 102, 18153–18158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinberg A, Jabs DA, Chou S, Martin BK, Lurain NS, Forman MS, Crumpacker C, Cytomegalovirus R, Viral Resistance Study G, Adult ACTGCL, 2003. Mutations conferring foscarnet resistance in a cohort of patients with acquired immunodeficiency syndrome and cytomegalovirus retinitis. J Infect Dis 187, 777–784. [DOI] [PubMed] [Google Scholar]
- Zoepfl M, Dwivedi R, Taylor MC, Pomin VH, McVoy MA, 2021. Antiviral activities of four marine sulfated glycans against adenovirus and human cytomegalovirus. Antiviral Res 190, 105077. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
