Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Aug 10.
Published in final edited form as: J Med Chem. 2023 Jul 28;66(15):10746–10760. doi: 10.1021/acs.jmedchem.3c00924

Quinazolinone Compounds Have Potent Antiviral Activity against Zika and Dengue Virus

Md Ashraf-Uz-Zaman 1,§, Xin Li 1,§, Yuan Yao 1,§, Chandra Bhushan Mishra 1, Bala Krishna Moku 1, Yongcheng Song 1,2,*
PMCID: PMC10463567  NIHMSID: NIHMS1926884  PMID: 37506506

Abstract

Dengue (DENV) and Zika (ZIKV) virus are important human pathogens, causing ~100 million symptomatic infections each year. These infections carry 20-fold increased incidence of serious neurological diseases, such as microcephaly in newborns (for ZIKV) and Guillain-Barré syndrome. Moreover, DENV can develop serious and possibly life-threatening dengue hemorrhagic fever in certain patients. Patients recovered from one of the 4 serotypes of DENV are still susceptible to other serotypes with a higher likelihood of serious disease because of antibody-dependent enhancement. Except for mosquito control, there have been no antiviral drugs to prevent and treat ZIKV/DENV infections. Phenotypic screening found 2,3,6-trisubstituted quinazolinone compounds are novel inhibitors of ZIKV replication. Fifty-four analogs were synthesized and their structure-activity relationships are discussed. Additional testing shows that compounds 22, 27 and 47 exhibited broad and potent activities against ZIKV and DENV with EC50 values as low as 86 nM with no significant cytotoxicity to mammalian cells.

Keywords: Zika virus, Dengue virus, Structure-activity relationship, Antiviral agents

Graphical Abstract

graphic file with name nihms-1926884-f0001.jpg

Introduction

Zika virus (ZIKV) and closely related dengue virus (DENV) belong to the genus Flavivirus of the virus family Flaviviridae, which also includes other major human pathogenic viruses such as yellow fever, Japanese encephalitis and West Nile viruses. ZIKV and DENV are transmitted by Aedes mosquitoes in the tropical and subtropical regions, where approximately 3 billion people or 40% of world population live and are at risk of these infections 1, 2.

ZIKV, discovered and isolated in 1947 3, has caused three major outbreaks in the Yap Island (~7,000 cases in 2007), the French Polynesia (~28,000 cases in 2013) in the Pacific ocean, and Brazil and other American countries (2015–2016) with significantly broader impact and damages 4, 5. Several millions of people in 48 Pan-American countries and territories have been infected, showing symptoms including fever, rashes and conjunctivitis. Although most people recovered in several days, ZIKV infection has been found to cause 20-fold increased incidence of serious neurological diseases, such as Guillain-Barré syndrome 68 and >4,000 cases of microcephaly (small brain/head) and other neurological defects in newborns 911. The prognosis of these infants to have normal brain functions is low and their life expectancy is shorter. WHO announced ZIKV is a “Public Health Emergency”. Moreover, ZIKV can be transmitted through sex or body fluids even when infected people have no symptoms 12, 13. This secondary transmission route renders it more difficult to contain ZIKV infection.

DENV has been a major human pathogen for the past several decades. It is estimated that DENV infects ~400 million people per year with 100 million developing symptoms including fever, headache, rash, conjunctivitis and pain in muscle and joints, which are usually self-healing in 3–10 days 1. However, ~500,000 cases/year develop serious and possibly life-threatening dengue hemorrhagic fever and shock syndrome. Approximately 22,000 people (mostly children) die of the disease per year 2. More problematic is that there are 4 serotypes of DENV (DENV-1–4) with significantly different genomes 14, 15. Patients who have recovered from one DENV serotype are still susceptible to other DENV serotypes and there is an increased likelihood to develop a more serious disease 16, because of existing antibodies against the previous DENV serotype 14, 17.

Except for mosquito control, there have been no antiviral drugs to prevent and treat ZIKV and DENV infections. The only licensed DENV vaccine, Dengvaxia, has been found to have safety and efficacy concerns for DENV-naïve individuals in clinical trials 18. A limited number of compounds have been reported to have potent antiviral activities against ZIKV or DENV 1930. Most of these compounds are natural products with complex chemical structures that are not amenable for medicinal chemistry optimization. Only a few compounds showed broad activities against both ZIKV and DENV 1923. There is therefore a pressing need to find effective antiviral agents against ZIKV and DENV. Here, we report discovery, synthesis and structure-activity relationships (SAR) of a series of tri-substituted quinazolinone compounds that exhibit potent antiviral activities against both ZIKV and DENV. Interestingly, a quinazolinone compound was reported to exhibit modest activities (16.7 μM) against Venezuelan equine encephalitis virus 31, which is a mosquito-borne Alphavirus causing flu-like symptoms in humans.

RESULTS and DISCUSSION

Discovery of novel anti-Zika compounds

A cell-based, phenotypic anti-ZIKV activity assay was developed. Rapid replication of ZIKV (FLR strain, Colombia, 2015 32) in Vero (monkey kidney cells that lack interferon-mediated antiviral defense 33) cells causes significant cytopathic effects (CPE) and eventually cell lysis, which can be clearly observed under the microscope. However, a compound that inhibits ZIKV replication can protect cells from CPE and death. If needed, an MTT [3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide] assay may further quantitate the number of viable cells. 0.1 multiplicity of infection (MOI, the number of infectious viral particles per cell) of ZIKV was added to a monolayer of cells in 96-well plates with culturing media containing a compound at 10 μM in duplicate for 72h. Observing CPE followed by MTT assay enabled us to quickly screen compounds to identify anti-ZIKV compounds. Moreover, cytotoxic compounds (at 10 μM), showing similar phenotypes (CPE and/or cell death) in the assay, were also eliminated.

Next, activities of these hits to reduce the viral titers were further determined, using an end-point dilution assay as described in our previous publications 3436. Half-log (0.32×) serial dilutions of the supernatant containing newly generated ZIKV particles were added to Vero cells in quadruplicate. Upon incubation for ~5 days, ZIKV infection in each sample was determined with CPE. TCID50 (tissue culture infective dose) was calculated based on the highest dilution in which ≥50% of the quadruplicate samples were infected with ZIKV. As compared to TCID50 of the untreated samples, ability of a compound to reduce ZIKV replication can be determined.

Through screening of ~1,000 compounds in our in-house compound library, which were synthesized for medicinal chemistry studies targeting H3K4 demethylase LSD1, histone acetyltransferase p300 and transcription cofactor AF9/ENL 3739, 2,3,6-trisubstituted quinazolinone compound 1 (Chart 1) was found to be a potent anti-ZIKV agent, which can inhibit the viral replication by 99.9% (3-log reduction) at 10 μM. Interestingly, di-substituted quinazolinone compounds 2-4 can also inhibit ZIKV replication by 68%–90% (0.5–1-log reduction). Known anti-ZIKV compounds chloroquine and SYC-1307 (developed by us) 34, 35 were used as positive controls and exhibited expected antiviral activities (90% inhibition by chloroquine at 10 μM and 68% by SYC-1307 at 1 μM). These results show certain quinazolinone compounds are novel, potent antiviral agents against ZIKV and medicinal chemistry optimization based on these compounds are needed.

Chart 1.

Chart 1.

Structures and anti-ZIKV (FLV strain) activities in Vero cells (at 10 μM) of compounds 1–4.

Chemical synthesis

Synthesis of quinazolinone compounds 1-54 for structure activity relationship studies is shown in Scheme 1. For compounds 2–21 and 43, isatoic anhydride compound 55 was condensed with an aniline derivative to give a benzamide compound 56, which was subjected to a cyclization reaction at 110 °C to produce 2-methyl-3-phenylquinazolinone 57. The methyl group of compound 57 was reacted with an arylaldehyde to yield target compounds 2–21 and 43.

Scheme 1:

Scheme 1:

Synthesis of compounds 1–54.a

aReagents and conditions: (i) an aniline, 1,4-dioxane, 90 °C, 12 h, 45–81%; (ii) 1,1,1-triethoxyethane, 110 °C, overnight, 32–94%; (iii) aryl-CHO, AcONa, AcOH, reflux, overnight, 35–91%; (iv) an amine, tris(dibenzylideneacetone)dipalladium(0), 2-dicyclohexylphosphino-2’,6’-dimethoxybiphenyl, sodium tert-butoxide, toluene, 110 °C, 24 h, 30–81%; (v) Pd/C, H2, MeOH, 25 °C, overnight, 80–88%; (vi) tetrakis(triphenylphosphine)palladium(0), 1,4-dioxane, H2O, 110 °C, 24 h, 45–80%.

For synthesis of compounds 1, 22-28, 35–42 and 44–52, 6-chloro-quinazolinone 20 was subjected to a Buchwald-Hartwig amination reaction with an amine to give the target compounds. Compound 25 was prepared from compound 24 by deprotection of its tert-butoxycarbonyl (BOC) group. Saturated compounds 53 and 54 were synthesized from compounds 22 and 27, respectively, by Pd/C catalyzed hydrogenation. A Suzuki coupling reaction with 6-bromo-quinazolinone 58 gave compounds 29-34.

Structure-activity relationships for inhibiting ZIKV replication

Disubstituted quinazolinone compounds 5-11 (Table 1) with a variety of R3 groups were found to be inactive at 10 μM in the compound screen. Lack of anti-ZIKV activities of these compounds, together with the high activity of compound 1, show the 3-tert-butyl R3 group in 1-3 seems to be favored. Thus, compounds 12-19 bearing a 3-tert-butyl R3 group and various R2 groups were synthesized and tested for their activities to inhibit ZIKV replication. As shown in Table 1, none of these compounds exhibited significantly improved activity. SAR analysis suggested an aromatic R2 group seems not to significantly affect the anti-ZIKV activity. For example, while an electron-deficient pyridin-4-yl or pyrimidine-containing R2 group in compounds 13 and 14 (68% inhibition) appears to be more favorable than a phenyl in 15 (inactive), compound 12 with a pyridin-3-yl R2 group was inactive. With both compounds having 90% ZIKV inhibitory activity at 10 μM, the pyridinyl R2 group in compound 3 does not exhibit a superior activity to the phenyl in 18. Interestingly, the para-CF3 group in compounds 3 and 18 appears to provide a moderate activity improvement, compared to unsubstituted or methoxy-substituted compounds 2, 11-17. A bicyclic naphthyl R2 group in compound 19 (68% inhibition) does not significantly enhance anti-ZIKV activity.

Table 1:

Structures and anti-ZIKV (FLV strain) activities of compounds 2–19 in Vero cells.

graphic file with name nihms-1926884-t0008.jpg
Cpd # R2 R3 % inhibition of ZIKV at 10 μM
5 graphic file with name nihms-1926884-t0009.jpg H 0
6 same as above 2-CH3 0
7 same as above 3-CH3 0
4 same as above 3-OCH3 68.4%
8 same as above 4-t-Bu 0
9 graphic file with name nihms-1926884-t0010.jpg same as above 0
10 graphic file with name nihms-1926884-t0011.jpg same as above 0
2 graphic file with name nihms-1926884-t0012.jpg 3-t-Bu 68.4%
3 graphic file with name nihms-1926884-t0013.jpg same as above 90.0%
11 graphic file with name nihms-1926884-t0014.jpg same as above 0
12 graphic file with name nihms-1926884-t0015.jpg same as above 0
13 graphic file with name nihms-1926884-t0016.jpg same as above 68.4%
14 graphic file with name nihms-1926884-t0017.jpg same as above 68.4%
15 graphic file with name nihms-1926884-t0018.jpg same as above 0
16 graphic file with name nihms-1926884-t0019.jpg same as above 0
17 graphic file with name nihms-1926884-t0020.jpg same as above 0
18 graphic file with name nihms-1926884-t0021.jpg same as above 90.0%
19 graphic file with name nihms-1926884-t0022.jpg same as above 68.4%

Next, we focused on optimization of the R6 group, since compared to unsubstituted compound 11, the diethylamino group in compound 1 (99.9% inhibition at 10 μM) renders ≥1,000-fold activity enhancement to inhibit ZIKV replication. Further testing showed compound 1 can inhibit ZIKV replication in Vero cells by 68% (0.5-log reduction) at 1 μM (Table 2). With the same 3-tert-butyl R3 and 6-methoxy-pyridin-2-yl R2 group, compounds 20-34 having various R6 groups were synthesized and their anti-ZIKV activities at 10 and 1 μM shown in Table 2. While compounds 20 and 21 with a -Cl and -OMe R6 group were inactive, compound 22 with a piperidin-1-yl group inhibited ZIKV replication by ≥99.9% at 10 μM and 90% at 1 μM, showing superior activities to compound 1 (68% inhibition at 1 μM). However, compound 23 with a 5-membered pyrolidin-1-yl R6 group is only a modest anti-ZIKV agent (68% inhibition at 10 μM). A larger, 4-BOC protected piperazin-1-yl R6 group in compound 24 (99% inhibition at 10 μM) is tolerable, although it is considerably less active than 1 and 22. Deprotection of BOC yielded compound 25 with a piperazin-1-yl R6 group, which was found to exhibit significant cytotoxicity to Vero cells, which is undesirable for this study. Similarly, compound 26 with a 4-methyl-piperazin-1-yl group is also cytotoxic, suggesting a basic N atom at the 4-position of the piperazine ring (in 25 and 26) is attributed to the toxicity, as compared to 22 and 24. Indeed, compound 27 having a morpholin-4-yl R6 substituent, as well as compound 28 with a larger 4-(morpholin-4-yl)piperazin-1-yl R6 group, is one of our most potent anti-ZIKV agents without cytotoxicity. Both compounds can inhibit ZIKV replication by 99.9% at 10 μM and 90% at 1 μM.

Table 2:

Structures and anti-ZIKV (FLV strain) activities of compounds 1, 20–43 in Vero cells.

graphic file with name nihms-1926884-t0023.jpg
Cpd # R2 R6 % inhibition of ZIKV at 10 μM % inhibition of ZIKV at 1 μM
1 graphic file with name nihms-1926884-t0024.jpg graphic file with name nihms-1926884-t0025.jpg 99.9% 68.4%
20 same as above -Cl 0 NT a
21 same as above -OCH3 0 NT a
22 same as above graphic file with name nihms-1926884-t0026.jpg 99.9% 90.0%
23 same as above graphic file with name nihms-1926884-t0027.jpg 68.4% NT a
24 same as above graphic file with name nihms-1926884-t0028.jpg 99.0% NT a
25 same as above graphic file with name nihms-1926884-t0029.jpg Cytotoxic NT a
26 same as above graphic file with name nihms-1926884-t0030.jpg Cytotoxic NT a
27 same as above graphic file with name nihms-1926884-t0031.jpg 99.9% 90.0%
28 same as above graphic file with name nihms-1926884-t0032.jpg 99.9% 90.0%
29 same as above graphic file with name nihms-1926884-t0033.jpg 0 NT a
30 same as above graphic file with name nihms-1926884-t0034.jpg 0 NT a
31 same as above graphic file with name nihms-1926884-t0035.jpg 99.0% NT a
32 same as above graphic file with name nihms-1926884-t0036.jpg 96.8% NT a
33 same as above graphic file with name nihms-1926884-t0037.jpg 96.8% NT a
34 same as above graphic file with name nihms-1926884-t0038.jpg 96.8% NT a
35 graphic file with name nihms-1926884-t0039.jpg graphic file with name nihms-1926884-t0040.jpg 90.0% NT a
36 graphic file with name nihms-1926884-t0041.jpg same as above 99.0% NT a
37 graphic file with name nihms-1926884-t0042.jpg same as above 68.4% NT a
38 graphic file with name nihms-1926884-t0043.jpg same as above 99.7% NT a
39 graphic file with name nihms-1926884-t0044.jpg same as above 96.8% NT a
40 same as above graphic file with name nihms-1926884-t0045.jpg Cytotoxic NT a
41 graphic file with name nihms-1926884-t0046.jpg graphic file with name nihms-1926884-t0047.jpg 99.9% 68.4%
42 same as above graphic file with name nihms-1926884-t0048.jpg 99.0% NT a
43 same as above -OCH3 0 NT a
a

Not tested.

Several compounds with an aromatic R6 group were investigated. While compounds 29 and 30 with a pyridine-4-yl and 3-methoxyphenyl are inactive, compounds 31-33 with a para-F, -dimethylamino and -pyrolidine substituted phenyl R6 group were moderately active with 99–96.8% ZIKV inhibition at 10 μM (Table 2). Compound 34 with a partially unsaturated tetrahydropyran-4-yl substituent exhibited moderate anti-ZIKV activity (96.8% inhibition at 10 μM). Collectively, SAR studies for the R6 show that 1) a N-atom directly attaching to the 6-position of the quinazolinone core provides a significant activity enhancement (e.g., 27 vs. 34); 2) 6-membered piperidin-1-yl or morpholin-4-yl (in 22, 27 and 28) is the most favored, while smaller 5-membered pyrrolidine ring loses most activity; and 3) a second, basic N-atom at the 4-position of the 6-membered ring is cytotoxic.

With the same 3-tert-butyl R3 and a piperidine or related R6 group, compounds 35-43 (Table 2) were synthesized to optimize the R2 substituent. Compounds 35 and 36 (99% and 90% inhibition at 10 μM), which contain a para-CF3-substituted pyridine and phenyl R2 group showing good activities in Table 1, are less active than compound 22, indicating the 6-methoxy-pyridin-2-yl R2 group in 22 is more favored. Compound 37 with a pyridine-4-yl R2 substituent is a modest compound (68% inhibition at 10 μM). Compound 38 having a 2-naphthyl R2 substituent exhibited a strong anti-ZIKV activity of 99.7% at 10 μM, while compound 39 bearing a 3,5-dimethoxyphenyl R2 moiety is less potent (96.8% ZIKV inhibition). Its analog 40 with a 4-methyl-piperazin-1-yl R6 group is also cytotoxic, consistent with the results of compounds 26 and 25. With a 3-methoxyphenyl R2 substituent, compound 41 is also a potent anti-ZIKV agent with 99.9% and 68% inhibition at 10 and 1 μM, but it is slightly weaker than compound 22 (90% inhibition at 1 μM), showing a pyridine-containing R2 substituent is more favorable than a phenyl. Similarly, compounds 42 and 43 with a 3-methoxyphenyl R2 moiety exhibited comparable anti-ZIKV activities to their pyridine-containing analogs 24 and 21. These results show the 6-methoxy-pyridin-2-yl R2 group (in 22) is the most favored for this series of compounds.

Compounds 44-52 (Table 3) with the most favored R2 and R6 groups were synthesized to further optimize the R3 substituent. 3-methyl group in compounds 44-46 (no inhibition at 1 μM) were found to be considerably less active than their 3-tert-butyl analogs 22, 27 and 28. 3-Methoxy R3 substituent in compounds 47-49 gives potent anti-ZIKV activities, but it is slightly less favored than 3-tert-butyl R3 group, when combined with a morpholine-containing R6 (e.g., compounds 48/49 vs. 27/28). Compounds 50-52 with an electron-withdrawing 3-CF3 R3 substituent are considerably less active, as compared to 22, 27 and 28. These results show 3-tert-butyl or 3-OMe R3 group is more favorable.

Table 3:

Structures and anti-ZIKV (FLV strain) activities of compounds 44–52 in Vero cells.

graphic file with name nihms-1926884-t0049.jpg
Cpd # R3 R6 % inhibition of ZIKV at 10 μM % inhibition of ZIKV at 1 μM
44 3-CH3 graphic file with name nihms-1926884-t0050.jpg 99.9% 0%
45 same as above graphic file with name nihms-1926884-t0051.jpg 90.0% NT a
46 same as above graphic file with name nihms-1926884-t0052.jpg 99.9% 0%
47 3-OCH3 graphic file with name nihms-1926884-t0053.jpg 99.9% 90.0%
48 same as above graphic file with name nihms-1926884-t0054.jpg 99.0% NT a
49 same as above graphic file with name nihms-1926884-t0055.jpg 99.9% 68.4%
50 3-CF3 graphic file with name nihms-1926884-t0056.jpg 99.0% NT a
51 same as above graphic file with name nihms-1926884-t0057.jpg 0% NT a
52 same as above graphic file with name nihms-1926884-t0058.jpg 68.4% NT a
a

Not tested.

Finally, the trans-C=C double bond in the most potent compounds 22 and 27 was hydrogenated to generate the corresponding saturated compounds 53 and 54 (Chart 2). Both compounds were found to be inactive (at 10 μM) against ZIKV replication in Vero cells. To test whether the double bond is chemically reactive in cells, which could cause covalent binding to a protein, compound 27 was tested in culture media with added glutathione (GSH), a nucleophilic thiol commonly present in cells. As shown in Supporting Information Figure S1, anti-ZIKV activity of compound 27 (as well as ZIKV replication) in Vero cells was not affected in the presence of 1 mM of GSH. These results show that the rigid, trans-C=C bond at the 2-position of the quinazolinone core is critical to the anti-ZIKV activity of this series of compounds.

Chart 2.

Chart 2.

Structures and anti-ZIKV (FLV strain) activities of compounds 53 and 54 in Vero cells.

Antiviral activity evaluation

The most potent compounds 22, 27 and 47 in the anti-ZIKV screen were selected for dose-dependent testing against several ZIKV and DENV strains. The representative results are shown in Figure 1 and the antiviral EC50 values summarized in Table 4. Compound 22 inhibited replication of the ZIKV FLR strain in Vero cells by 0%, 90% and >99.9% at 0.3, 1, 3 and 10 μM (Figure 1A) with a calculated EC50 value of 900 nM (Table 4). Compound 27 exhibited ~4× more potent antiviral activity with an EC50 of 180 nM, suppressing the ZIKV-FLR replication in Vero cells by 68%, 90% and >99.9% at 0.3, 1 and ≥3 μM. Compound 47 (EC50 = 210 nM) was found to have similar activities to 27. Moreover, as shown in Figure 2A, treatment of ZIKV-infected Vero cells with compounds 22 and 27 can dose-dependently reduce the ZIKV RNA copies in the supernatant. Although ~1/104 of these RNAs represent infectious viruses 32, the quantitative PCR results confirm the antiviral activities of these compounds. Treatment with compound 27 can also significantly reduce the cellular levels of viral NS5, NS3 and capsid proteins (Figure 2B). In addition, these three compounds inhibited the ZIKV-FLR replication in human glioblastoma U87 cells with comparable potencies, with compound 27 showing the most potent antiviral activity. It inhibited the viral replication by 90%, 97%, 99% and >99.9% at 0.3, 1, 3 and 10 μM (Figure 1B) with an EC50 of 100 nM. Besides the two mammalian cells, ZIKV-FLR replication in mosquito C6/36 cells was also significantly blocked by these three compounds with EC50s of 230–770 nM (Table 4 and Figure 1C).

Figure 1.

Figure 1.

Representative antiviral activities of compounds 22, 27, and 47 in cells. (A-C, E) Treatment of Vero (A, E), U87 (B) and mosquito C6/36 cells (C) with compounds 22, 27, and 47 caused dose-dependent reduction of infectious ZIKV-FLR (A-C) and DENV-2 (E); (D, F) Treatment of Vero cells with compound 27 inhibited generation of infectious ZIKV-HN16 (D) and DENV-3 (F).

Table 4:

Antiviral activity (EC50 in μM) against ZIKV and DENV.

ZIKV-FLR in Vero cells ZIKV-FLR in U87 cells ZIKV-FLR in C6/36 cells ZIKV-HN16 in Vero cells DENV-2 in Vero cells DENV-3 in Vero cells CC50 (μM) in U87 cells

22 0.90 ± 0.32 0.82 ± 0.09 0.75 ± 0.40 NT a 0.16 ± 0.04 NT a >20
27 0.18 ± 0.02 0.10 ± 0.022 0.23 ± 0.01 0.086 ± 0.057 0.21 ± 0.02 0.12 ± 0.07 >20
47 0.21 ± 0.01 0.69 ± 0.47 0.77 ± 0.41 NT a 0.56 ± 0.02 NT a >20
a

Not tested.

Figure 2.

Figure 2.

Treatment of Vero cells with compounds 27 and/or 22 dose-dependently reduced (A) the ZIKV RNA copies in the cell supernatant and (B) cellular viral NS5, NS3 and capsid proteins.

Next, the most potent compound 27 was tested against replication of the HN16 strain of ZIKV (Honduras, 2016 16) in Vero cells. The compound can potently inhibit ZIKV-HN16 replication in a dose-dependent manner with an EC50 of 86 nM (Table 4 and Figure 1D). Moreover, these three compounds did not significantly inhibit proliferation of human U87 cells at 20 μM, showing they are non-cytotoxic with a very high selectivity against ZIKV replication.

We next tested antiviral activity of these compounds against replication of DENV, using a similar end-point dilution assay 34. We were pleased to find that compounds 22, 27 and 47 strongly inhibited replication of DENV serotype 2 virus (DENV-2, strain K0049, Thailand, 1995) in Vero cells with EC50 values of 160, 210 and 560 nM, respectively (Table 4 and Figure 1E). In addition, replication of DENV serotype 3 virus (DENV-3, strain 7431, Sri Lanka, 1989) was also potently suppressed by compound 27 with an EC50 of 120 nM (Table 4 and Figure 1F).

We also performed experiments to see whether delayed addition of compound 27 affects its anti-ZIKV activity in Vero cells. As shown in Figure 3A, adding compound 27 (1 μM) together with the initial virus into the cell culture media and incubating for 2 hours (2h-pretreatment), which is the regular compound treatment procedure for all of the above experiments, provided significantly higher anti-ZIKV activity, consistently reducing the viral titers by ≥90%. However, adding compound 27 (1 μM) immediately (0h-posttreatment) or 2 hours (2h-posttreatment) after removal of the inoculated virus showed reduced antiviral activity with 68% of viral titer reduction. Chloroquine, a known entry inhibitor of ZIKV, showed similar anti-ZIKV activities for these experiments (Figure 3B). These results suggest that compound 27 inhibits the attachment and/or entry of ZIKV to the cells.

Figure 3.

Figure 3.

Anti-ZIKV (FLV strain) activities of (A) compound 27 and (B) chloroquine when added into the cell culture media at −2, 0 and 2 hours before removal of the inoculated virus (designated as 2h-pretreatment, 0h-, and 2h-posttreatment, respectively).

Conclusion

DENV and ZIKV are important human pathogens, causing ~100 million symptomatic infections each year. More significantly, these viral infections can further cause serious and life-threatening diseases, such as microcephaly (in newborns), Guillain-Barré syndrome, and dengue hemorrhagic fever and shock syndrome. In addition, patients recovered from one of 4 serotypes of DENV infection are still susceptible to other serotypes with an increased likelihood of serious disease because of antibody-dependent enhancement. Except for mosquito control, there have been no antiviral drugs to prevent and treat ZIKV/DENV infections. There is therefore a pressing need for such antiviral agents. Phenotype-based compound screening found several 2,3,6-trisubstituted quinazolinone compounds are novel inhibitors of ZIKV replication. Fifty-four analogs were synthesized and tested with their structure-activity relationships discussed above. Additional characterization shows that the most potent compounds 22, 27 and 47 are non-cytotoxic and exhibited potent activities against ZIKV and DENV with EC50 values as low as 86 nM in a variety of mammalian and mosquito cells. Their structures are amenable for further medicinal chemistry optimization. Although the protein or nucleic acid target(s) that these compounds inhibit in mammalian and mosquito cells is unknown and is the subject of future investigation, our results suggest they inhibit ZIKV attachment or entry to the cell. Nevertheless, given the scarcity of potent and broad anti-Flavivirus agents, these compounds are novel pharmacological leads for drug development against ZIKV and DENV infections.

Experimental Section

All the chemicals used for synthesis were purchased from Aldrich (Milwaukee, WI) or Alfa Aesar (Ward Hill, MA). Unless otherwise stated, all solvents and reagents were used as received. All reactions were conducted with the use of a Teflon-coated magnetic stir bar at the indicated temperature and were performed under an inert atmosphere when stated. The identity of the synthesized compounds was characterized by 1H and 13C NMR on a Varian (Palo Alto, CA) 400-MR spectrometer and mass spectrometer (Shimadzu LCMS-2020). Chemical shifts were reported in parts per million (ppm, δ) downfield from tetramethylsilane. Proton coupling patterns are described as singlet (s), doublet (d), triplet (t), quartet (q), multiplet (m), and broad (br). The identity of the potent inhibitors was confirmed with high resolution mass spectra (HRMS) using an Agilent 6550 iFunnel quadrupole-time-of-flight (Q-TOF) mass spectrometer with electrospray ionization (ESI). The purities of the final compounds were determined to be >95% with a Shimadzu Prominence HPLC using a Zorbax C18 (or C8) column (4.6 × 250 mm) at 0.7 mL/min flow rate (water:acetonitrile with 0.1% formic acid, 90:10 – 5:95 in 3 min) monitored by UV at 254 nm.

Chemical synthesis.

General synthetic methods for compounds 2–21 and 43:

A mixture of isatoic anhydride 55 (3.07 mmol, 1 eq) and an aniline (3.37 mmol, 1.1 eq) was heated in 1,4-dioxane (10 mL) at 90 °C for 12 h. The solvent was evaporated, and the crude product purified with column chromatography (silica gel, hexane/ethyl acetate, 6/1) to give compound 56 as a white or pale-yellow powder in 45–89% yield.

A mixture of 2-amino-N-phenyl-benzamide 56 (0.565 mmol, 1 eq) and 1,1,1-triethoxy-ethane (0.28 g, 1.696 mmol, 3 eq) were stirred at 110 °C for overnight. The reaction mixture was quenched with water (10 mL) and the product extracted with ethyl acetate (3 × 20 mL). The combined organic layers were washed with brine and dried over Na2SO4. Upon removal of the solvent, the residue was purified with column chromatography (silica gel, hexane/ethyl acetate, 10/1) to provide compound 57 as a white or pale-yellow powder in 32–94% yield.

A mixture of 2-methyl-3-phenyl-4(3H)-quinazolinone 57 (0.14 mmol, 1 eq), an aldehyde (0.18 mmol, 1.3 eq) and NaOAc (6.8 mg, 0.08 mmol, 0.6 eq) in AcOH (1 ml) was heated at 110 °C for overnight. The reaction mixture was cooled down and the product extracted with ethyl acetate (3 × 20 mL). The combined organic layers were washed with brine and dried over Na2SO4. Upon removal of the solvent, the residue was purified with column chromatography (silica gel, hexane/ethyl acetate, 5/1) to provide compounds 2–21 and 43 as a white or pale-yellow powder in 35–91% yield.

3-(3-tert-Butylphenyl)-2-[2-(2-methoxyl-3-pyridinyl)ethenyl]-4(3H)-quinazolinone (2)

1H NMR (CDCl3, 400 MHz) δ 8.31 (d, J = 8.3 Hz, 1H), 8.07 (dd, J = 4.9, 1.7 Hz, 1H), 7.96 (d, J = 15.6 Hz, 1H), 7.82 – 7.73 (m, 2H), 7.60 – 7.40 (m, 4H), 7.29 (d, J = 1.7 Hz, 1H), 7.18 – 7.11 (m, 1H), 6.85 (dd, J = 7.3, 4.9 Hz, 1H), 6.74 (d, J = 15.6 Hz, 1H), 3.79 (s, 3H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.5, 161.9, 153.4, 152.4, 147.9, 147.2, 138.7, 137.1, 134.6, 134.3, 129.5, 127.5, 127.3, 126.7, 126.2, 125.8, 123.4, 121.1, 119.1, 117.1, 53.3, 35.0, 31.4. MS (ESI) calculated for (C26H26N3O2)+ [M+H]+ 412.2, found 412.2.

3-(3-tert-Butylphenyl)-2-[2-(4-trifluoromethyl-3-pyridinyl)ethenyl]-4(3H)-quinazolinone (3)

1H NMR (CDCl3, 400 MHz) δ 8.64 (s, 1H), 8.33 (d, J = 7.7 Hz, 1H), 7.95 (d, J = 15.7 Hz, 1H), 7.84 – 7.76 (m, 2H), 7.71 (d, J = 8.0 Hz, 1H), 7.56 (ddd, J = 15.5, 13.8, 8.0 Hz, 4H), 7.29 (s, 1H), 7.15 (dd, J = 7.6, 0.9 Hz, 1H), 6.52 (d, J = 15.7 Hz, 1H), 1.34 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.1, 153.8, 150.7, 149.0, 147.6, 136.4, 135.5, 134.9, 134.0, 129.8, 127.7, 127.4, 126.7, 125.8, 125.7, 124.7, 121.4, 120.6, 110.1, 35.1, 31.4. MS (ESI) calculated for (C26H23F3N3O)+ [M+H]+ 450.2, found 450.2.

3-(3-Methoxylphenyl)-2-[2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone (4)

1H NMR (CDCl3, 400 MHz) δ 8.58 (s, 1H), 8.49 (d, J = 4.1 Hz, 1H), 8.27 (d, J = 7.9 Hz, 1H), 7.94 (d, J = 15.6 Hz, 1H), 7.77 (d, J = 3.6 Hz, 2H), 7.56 (d, J = 8.0 Hz, 1H), 7.46 (td, J = 8.2, 5.0 Hz, 2H), 7.21 (dd, J = 7.9, 4.8 Hz, 1H), 7.07 (dd, J = 8.2, 2.2 Hz, 1H), 6.91 – 6.80 (m, 2H), 6.48 (d, J = 15.6 Hz, 1H), 3.82 (s, 3H). 13C NMR (CDCl3, 100 MHz) δ 162.1, 160.8, 151.0, 150.3, 149.4, 147.6, 137.9, 136.1, 134.7, 134.1, 131.1, 130.8, 127.5, 127.2, 127.0, 123.7, 121.9, 121.1, 120.8, 115.4, 114.4, 55.6. MS (ESI) calculated for (C22H18N3O2)+ [M+H]+ 356.1, found 356.2.

3-Phenyl-2-[2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone (5)

1H NMR (CDCl3, 400 MHz) δ 8.58 (s, 1H), 8.50 (s, 1H), 8.30 (d, J = 7.8 Hz, 1H), 7.94 (d, J = 15.6 Hz, 1H), 7.80 (s, 2H), 7.57 (t, J = 8.3 Hz, 4H), 7.49 (s, 1H), 7.32 (d, J = 7.0 Hz, 2H), 7.24 (dd, J = 11.8, 6.8 Hz, 1H), 6.45 (d, J = 15.5 Hz, 1H). 13C NMR (CDCl3, 100 MHz) δ 162.3, 151.1, 150.4, 149.5, 147.7, 136.9, 136.2, 134.8, 134.1, 131.2, 130.1, 129.7, 128.8, 127.6, 127.3, 127.1, 123.7, 122.0, 121.2. MS (ESI) calculated for (C21H16N3O)+ [M+H]+ 326.1, found 326.1.

3-(2-Methylphenyl)-2-[2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone (6)

1H NMR (CDCl3, 400 MHz) δ 8.57 (s, 1H), 8.50 (d, J = 4.3 Hz, 1H), 8.32 (d, J = 7.9 Hz, 1H), 7.97 (d, J = 15.6 Hz, 1H), 7.81 (d, J = 4.2 Hz, 2H), 7.55 (d, J = 7.9 Hz, 1H), 7.51 – 7.34 (m, 4H), 7.25 – 7.18 (m, 2H), 6.38 (d, J = 15.6 Hz, 1H), 2.12 (s, 3H). 13C NMR (CDCl3, 100 MHz) δ 161.7, 151.1, 150.4, 149.5, 147.9, 136.6, 136.2, 136.0, 134.8, 134.1, 131.7, 131.2, 130.0, 128.7, 127.8, 127.6, 127.4, 127.0, 123.8, 121.3, 121.2, 17.7. MS (ESI) calculated for (C22H18N3O)+ [M+H]+ 340.1, found 340.1

3-(3-Methylphenyl)-2-[2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone (7)

1H NMR (CDCl3, 400 MHz) δ 8.59 (s, 1H), 8.51 (d, J = 3.9 Hz, 1H), 8.30 (d, J = 7.8 Hz, 1H), 7.95 (d, J = 15.6 Hz, 1H), 7.79 (d, J = 3.2 Hz, 2H), 7.59 (d, J = 7.6 Hz, 1H), 7.51 – 7.41 (m, 2H), 7.36 (d, J = 7.3 Hz, 1H), 7.24 (dd, J = 7.4, 4.7 Hz, 1H), 7.15 – 7.04 (m, 2H), 6.48 (d, J = 15.6 Hz, 1H), 2.44 (s, 3H). 13C NMR (CDCl3, 100 MHz) δ 162.3, 151.1, 150.2, 149.3, 147.7, 140.4, 136.7, 136.0, 134.8, 134.3, 131.3, 130.5, 129.9, 129.2, 127.5, 127.3, 127.0, 125.6, 123.8, 122.2, 121.2, 21.5.

134.1, 131.1, 130.8, 127.5, 127.2, 127.0, 123.7, 121.9, 121.1, 120.8, 115.4, 114.4, 55.6. MS (ESI) calculated for (C22H18N3O)+ [M+H]+ 340.1, found 340.1.

3-(4-tert-Butylphenyl)-2-[2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone (8)

1H NMR (CDCl3, 400 MHz) δ 8.59 (s, 1H), 8.51 (s, 1H), 8.30 (d, J = 7.6 Hz, 1H), 7.94 (d, J = 15.5 Hz, 1H), 7.79 (s, 2H), 7.59 (d, J = 7.3 Hz, 3H), 7.49 (s, 1H), 7.23 (d, J = 7.3 Hz, 3H), 6.49 (d, J = 15.6 Hz, 1H), 1.40 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.4, 152.8, 151.4, 150.3, 149.4, 147.7, 136.1, 134.8, 134.3, 134.1, 131.3, 128.2, 127.6, 127.4, 127.1, 127.0, 123.8, 122.3, 121.2, 35.1, 31.5. MS (ESI) calculated for (C25H24N3O)+ [M+H]+ 382.2, found 382.2.

3-(4-tert-Butylphenyl)-2-[2-(5-bromo-3-pyridinyl)ethenyl]-4(3H)-quinazolinone (9)

1H NMR (CDCl3, 400 MHz) δ 8.56 (d, J = 2.1 Hz, 1H), 8.46 (d, J = 1.7 Hz, 1H), 8.35 – 8.27 (m, 1H), 7.84 (d, J = 15.6 Hz, 1H), 7.80 (dd, J = 5.8, 1.2 Hz, 2H), 7.66 (t, J = 1.8 Hz, 1H), 7.54 (dddd, J = 11.9, 8.2, 4.3, 1.8 Hz, 3H), 7.28 (t, J = 1.8 Hz, 1H), 7.19 – 7.13 (m, 1H), 6.43 (d, J = 15.6 Hz, 1H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.1, 153.6, 151.1, 150.8, 147.6, 147.1, 136.5, 136.4, 134.8, 134.1, 132.9, 129.8, 127.6, 127.3, 127.2, 126.7, 125.7, 123.7, 121.3, 121.1, 35.1, 31.4. MS (ESI) calculated for (C25H23BrN3O)+ [M+H]+ 460.1, found 460.1

3-(4-tert-Butylphenyl)-2-[2-(5-pyrimidyl)ethenyl]-4(3H)-quinazolinone (10)

1H NMR (CDCl3, 400 MHz) δ 9.10 (s, 1H), 8.67 (s, 2H), 8.31 (d, J = 7.9 Hz, 1H), 7.88 (d, J = 15.7 Hz, 1H), 7.79 (s, 2H), 7.59 (d, J = 8.1 Hz, 2H), 7.51 (t, J = 5.7 Hz, 1H), 7.24 (t, J = 7.8 Hz, 2H), 6.57 (d, J = 15.7 Hz, 1H), 1.40 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.2, 158.6, 155.4, 153.0, 150.7, 147.5, 134.9, 133.8, 132.1, 129.4, 128.1, 127.6, 127.4, 127.2, 124.1, 121.3, 35.1, 31.5. MS (ESI) calculated for (C24H23N4O)+ [M+H]+ 383.2, found 383.2

3-(3-tert-Butylphenyl)-2-[2-(3-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (11)

1H NMR (CDCl3, 400 MHz) δ 8.32 (d, J = 7.7 Hz, 1H), 7.81 (d, J = 8.6 Hz, 3H), 7.60 – 7.43 (m, 4H), 7.32 (s, 1H), 7.14 (d, J = 7.2 Hz, 1H), 7.04 (d, J = 14.8 Hz, 1H), 6.89 (d, J = 7.0 Hz, 1H), 6.62 (d, J = 8.2 Hz, 1H), 3.59 (s, 3H), 1.33 (s, 9H).13C NMR (CDCl3, 100 MHz) δ 163.3, 162.4, 153.5, 152.0, 150.8, 148.0, 139.1, 137.7, 137.1, 134.6, 129.5, 127.5, 127.3, 126.8, 126.2, 125.8, 125.6, 124.1, 121.3, 118.2, 112.2, 52.7, 35.0, 31.4. MS (ESI) calculated for (C26H26N3O2)+ [M+H]+ 412.2, found 412.2

3-(3-tert-Butylphenyl)-2-[2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone (12)

1H NMR (CDCl3, 400 MHz) δ 8.57 (s, 1H), 8.51 (d, J = 4.5 Hz, 1H), 8.32 (d, J = 7.8 Hz, 1H), 7.92 (d, J = 15.6 Hz, 1H), 7.80 (d, J = 3.6 Hz, 2H), 7.59 – 7.45 (m, 4H), 7.29 (s, 1H), 7.26 – 7.19 (m, 1H), 7.15 (d, J = 7.5 Hz, 1H), 6.44 (d, J = 15.6 Hz, 1H), 1.34 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.3, 153.6, 151.3, 150.3, 149.3, 147.7, 136.6, 135.9, 134.8, 134.1, 131.3, 129.7, 127.6, 127.3, 127.1, 126.6, 125.8, 125.7, 123.8, 122.3, 121.3, 35.1, 31.4. MS (ESI) calculated for (C25H26N3O)+ [M+H]+ 382.2, found 382.2

3-(3-tert-Butylphenyl)-2-[2-(4-pyridinyl)ethenyl]-4(3H)-quinazolinone (13)

1H NMR (CDCl3, 400 MHz) δ 8.55 (d, J = 4.5 Hz, 2H), 8.33 (d, J = 7.6 Hz, 1H), 7.86–7.80 (m, 3H), 7.62 – 7.47 (m, 3H), 7.27 (d, J = 7.9 Hz, 1H), 7.14 (dd, J = 19.1, 5.6 Hz, 3H), 6.54 (d, J = 15.6 Hz, 1H), 1.34 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.2, 153.6, 151.0, 150.5, 147.6, 142.6, 136.6, 136.5, 134.8, 129.7, 127.7, 127.4, 127.3, 126.6, 125.8, 125.7, 124.7, 121.6, 121.4, 35.1, 31.4. MS (ESI) calculated for (C25H26N3O)+ [M+H]+ 382.2, found 382.2

3-(3-tert-Butylphenyl)-2-[2-(5-pyrimidyl)ethenyl]-4(3H)-quinazolinone (14)

1H NMR (CDCl3, 400 MHz) δ 9.09 (s, 1H), 8.64 (s, 2H), 8.32 (d, J = 7.7 Hz, 1H), 7.92 – 7.75 (m, 3H), 7.54 (dt, J = 11.4, 7.8 Hz, 3H), 7.28 (s, 1H), 7.14 (d, J = 7.6 Hz, 1H), 6.50 (d, J = 15.7 Hz, 1H), 1.34 (s, 9H).13C NMR (CDCl3, 100 MHz) δ 162.1, 158.6, 155.3, 153.8, 150.6, 147.5, 136.4, 134.9, 131.9, 129.8, 129.4, 127.7, 127.4, 126.7, 125.7, 125.7, 124.1, 121.4, 35.1, 31.4. MS (ESI) calculated for (C24H23N4O)+ [M+H]+ 383.2, found 383.2.

3-(3-tert-Butylphenyl)-2-(2-phenylethenyl)-4(3H)-quinazolinone (15)

1H NMR (CDCl3, 400 MHz) δ 8.32 (d, J = 7.8 Hz, 1H), 7.95 (d, J = 15.5 Hz, 1H), 7.82 – 7.78 (m, 2H), 7.60 – 7.44 (m, 3H), 7.30 (s, 6H), 7.16 (dd, J = 7.8, 1.6 Hz, 1H), 6.38 (d, J = 15.5 Hz, 1H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.4, 153.5, 152.1, 147.9, 139.8, 136.8, 135.5, 134.7, 129.7, 129.6, 128.9, 127.8, 127.5, 127.3, 126.7, 126.4, 125.9, 125.8, 121.2, 120.3, 35.1, 31.4. MS (ESI) calculated for (C26H25N2O)+ [M+H]+ 381.2, found 381.2.

3-(3-tert-Butylphenyl)-2-[2-(2-methoxylphenyl)ethenyl]-4(3H)-quinazolinone (16)

1H NMR (CDCl3, 400 MHz) δ 8.31 (d, J = 7.8 Hz, 1H), 8.18 (d, J = 16.0 Hz, 1H), 7.87 – 7.73 (m, 2H), 7.60 – 7.42 (m, 4H), 7.29 (dd, J = 6.3, 1.6 Hz, 1H), 7.24 (s, 1H), 7.15 (d, J = 7.5 Hz, 1H), 6.91 – 6.79 (m, 2H), 6.63 (d, J = 15.6 Hz, 1H), 3.71 (s, 3H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.5, 158.5, 153.4, 137.1, 134.6, 130.8, 130.0, 129.5, 127.3, 126.5, 126.2, 125.9, 124.5, 120.8, 111.1, 55.2, 35.1, 31.4. MS (ESI) calculated for (C27H27N2O2)+ [M+H]+ 411.2, found 411.2

3-(3-tert-Butylphenyl)-2-[2-(3-methoxylphenyl)ethenyl]-4(3H)-quinazolinone (17)

1H NMR (CDCl3, 400 MHz) δ 8.32 (d, J = 8.0 Hz, 1H), 7.92 (d, J = 15.5 Hz, 1H), 7.79 (dd, J = 3.6, 1.7 Hz, 2H), 7.60 – 7.43 (m, 3H), 7.30 (s, 1H), 7.24 – 7.13 (m, 2H), 6.93 – 6.79 (m, 3H), 6.36 (d, J = 15.5 Hz, 1H), 3.76 (s, 3H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.4, 159.9, 153.5, 152.0, 147.9, 139.7, 136.9, 136.8, 134.7, 129.9, 129.6, 127.4, 127.3, 126.7, 126.4, 125.8, 121.1, 120.6, 120.3, 115.4, 113.0, 55.3, 35.0, 31.4. MS (ESI) calculated for (C27H27N2O2)+ [M+H]+ 411.2, found 411.2.

3-(3-tert-Butylphenyl)-2-[2-(4-trifluoromethylphenyl)ethenyl]-4(3H)-quinazolinone (18)

1H NMR (CDCl3, 400 MHz) δ 8.33 (d, J = 8.0 Hz, 1H), 7.95 (d, J = 15.6 Hz, 1H), 7.81 (d, J = 3.7 Hz, 2H), 7.61 – 7.46 (m, 5H), 7.38 (d, J = 8.0 Hz, 2H), 7.30 (s, 1H), 7.17 (d, J = 7.5 Hz, 1H), 6.45 (d, J = 15.6 Hz, 1H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.2, 153.6, 151.5, 147.6, 138.9, 137.9, 136.6, 134.8, 129.7, 127.9, 127.5, 127.4, 127.1, 126.6, 125.9, 125.9, 125.8, 125.8, 122.6, 121.3, 35.1, 31.4. MS (ESI) calculated for (C27H24F3N2O)+ [M+H]+ 449.2, found 449.2.

3-(3-tert-Butylphenyl)-2-[2-(1-naphthyl)ethenyl]-4(3H)-quinazolinone (19)

1H NMR (CDCl3, 400 MHz) δ 8.77 (d, J = 15.3 Hz, 1H), 8.34 (d, J = 7.9 Hz, 1H), 8.25 (d, J = 8.3 Hz, 1H), 7.93 – 7.74 (m, 4H), 7.59 – 7.46 (m, 5H), 7.39 – 7.30 (m, 3H), 7.19 (d, J = 7.5 Hz, 1H), 6.48 (d, J = 15.3 Hz, 1H), 1.35 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 153.6, 136.9, 134.8, 133.8, 133.1, 131.6, 130.1, 129.7, 128.9, 127.5, 127.3, 126.9, 126.5, 126.3, 125.9, 125.8, 125.5, 124.8, 123.9, 122.9, 121.2, 35.1, 31.4. MS (ESI) calculated for (C30H27N2O)+ [M+H]+ 431.2, found 431.2.

6-Chloro-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (20)

1H NMR (CDCl3, 400 MHz) δ 8.25 (d, J = 1.5 Hz, 1H), 7.79 (d, J = 14.8 Hz, 1H), 7.75 – 7.66 (m, 2H), 7.51 (ddd, J = 13.0, 12.3, 6.7 Hz, 3H), 7.31 (s, 1H), 7.14 (d, J = 7.5 Hz, 1H), 7.01 (d, J = 14.8 Hz, 1H), 6.88 (d, J = 7.1 Hz, 1H), 6.62 (d, J = 8.3 Hz, 1H), 3.58 (s, 3H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.3, 161.4, 153.6, 152.2, 150.6, 146.4, 139.0, 138.1, 136.8, 135.0, 132.4, 129.6, 129.2, 126.6, 126.3, 125.7, 125.5, 123.7, 122.2, 118.2, 112.3, 52.7, 35.0, 31.4. MS (ESI) calculated for (C26H25ClN3O2)+ [M+H]+ 446.2, found 446.1.

6-Methoxy-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (21)

1H NMR (CDCl3, 400 MHz) δ 7.80 – 7.71 (m, 2H), 7.69 (d, J = 3.0 Hz, 1H), 7.56 – 7.44 (m, 3H), 7.39 (dd, J = 8.9, 3.0 Hz, 1H), 7.30 (d, J = 2.0 Hz, 1H), 7.12 (ddd, J = 7.5, 2.2, 1.3 Hz, 1H), 7.01 (d, J = 14.8 Hz, 1H), 6.87 (d, J = 7.2 Hz, 1H), 6.60 (dd, J = 8.2, 0.9 Hz, 1H), 3.92 (s, 3H), 3.58 (s, 3H), 1.32 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 158.5, 153.3, 150.8, 149.8, 138.9, 137.0, 129.3, 128.9, 125.9, 125.6, 125.5, 124.8, 117.8, 111.8, 106.5, 55.8, 52.5, 34.8, 31.2. MS (ESI) calculated for (C28H28N3O3)+ [M+H]+ 442.2, found 442.2.

6-Methoxy-3-(3-tert-butylphenyl)-2-(3-methoxystyryl)-4(3H)-quinazolinone (43)

1H NMR (CDCl3, 400 MHz) δ 7.84 (d, J = 15.5 Hz, 1H), 7.73 (d, J = 8.9 Hz, 1H), 7.67 (d, J = 2.9 Hz, 1H), 7.55 (ddd, J = 8.0, 1.9, 1.2 Hz, 1H), 7.53 – 7.47 (m, 1H), 7.38 (dd, J = 9.0, 3.0 Hz, 1H), 7.28 (t, J = 1.9 Hz, 1H), 7.19 (t, J = 7.9 Hz, 1H), 7.13 (ddd, J = 7.6, 2.1, 1.2 Hz, 1H), 6.86 (ddd, J = 7.6, 1.7, 0.9 Hz, 1H), 6.84 – 6.76 (m, 2H), 6.33 (d, J = 15.5 Hz, 1H), 3.91 (s, 3H), 3.74 (s, 3H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.0, 159.7, 158.4, 153.2, 149.8, 136.9, 136.7, 129.6, 129.4, 128.9, 126.1, 125.6, 125.6, 124.9, 121.7, 120.0, 115.0, 112.8, 106.5, 55.8, 55.1, 34.8, 31.2. MS (ESI) calculated for (C28H29N3O2)+ [M+H]+ 441.2, found 441.2.

General synthetic methods for compounds 1, 22–28, 35–42, 44–52:

A mixture of a compound 20 (35 mg, 0.07 mmol), an amine (0.08 mmol), tris(dibenzylideneacetone)dipalladium(0) (4 mg, 0.004 mol), 2-dicyclohexylphosphino-2’,6’-dimethoxybiphenyl (3 mg, 0.008 mmol), sodium tert-butoxide (10 mg, 0.10 mmol) in toluene (5 mL) was heated at 110 °C for 24 h. The reaction mixture was cooled down and quenched with brine (20 mL). The product was extracted with ethyl acetate (3 × 20 mL), and the combined organic phase was washed with brine and then dried over Na2SO4. Upon removal of solvent, the residue was purified with column chromatography (silica gel, hexanes : ethyl acetate, 8: 1) to give the target compounds 1, 22–24, 26–28, 35–42, 44–52 as a pale yellow solid in 30–81% yield.

To the solution of compound 24 (0.1 mmol) in dichloromethane (2 mL) was added dropwise HCl (0.2 mL, 4N in p-dioxane) at 0 °C. The reaction mixture was warmed to room temperature and stirred for 12 hours. The solid powder thus obtained was filtered and washed with in dichloromethane to give compound 25 as a hydrochloride salt.

6-Diethylamino-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (1)

1H NMR (CDCl3, 400 MHz) δ 7.76 – 7.59 (m, 2H), 7.54 – 7.37 (m, 4H), 7.30 (d, J = 2.0 Hz, 1H), 7.21 (d, J = 9.2 Hz, 1H), 7.11 (ddd, J = 7.4, 2.1, 1.2 Hz, 1H), 7.00 (d, J = 14.9 Hz, 1H), 6.84 (d, J = 7.2 Hz, 1H), 6.57 (d, J = 8.3 Hz, 1H), 3.57 (s, 3H), 3.46 (q, J = 7.1 Hz, 4H), 1.31 (s, 9H), 1.21 (t, J = 7.1 Hz, 6H). 13C NMR (CDCl3, 100 MHz) δ 163.0, 162.2, 153.1, 151.1, 147.3, 146.8, 138.8, 137.4, 135.1, 129.2, 128.5, 125.7, 125.5, 124.3, 122.2, 120.0, 117.5, 111.3, 105.6, 52.5, 44.6, 34.8, 31.2, 12.4. MS (ESI) calculated for (C30H35N4O2)+ [M+H]+ 483.3, found 483.2.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (22)

1H NMR (CDCl3, 400 MHz) δ 7.71 (d, J = 4.2 Hz, 1H), 7.69 – 7.62 (m, 2H), 7.48 (ddd, J = 11.3, 10.7, 5.7 Hz, 4H), 7.31 (s, 1H), 7.13 (d, J = 7.3 Hz, 1H), 7.02 (d, J = 14.9 Hz, 1H), 6.85 (d, J = 7.1 Hz, 1H), 6.58 (d, J = 8.2 Hz, 1H), 3.58 (s, 3H), 3.36 – 3.26 (m, 4H), 1.72 (d, J = 4.4 Hz, 4H), 1.62 (d, J = 4.7 Hz, 2H), 1.32 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.2, 162.4, 153.3, 151.2, 150.7, 148.9, 140.7, 139.0, 137.5, 135.9, 129.4, 128.4, 125.9, 125.9, 125.7, 124.8, 124.4, 122.0, 117.8, 111.7, 110.1, 52.7, 50.3, 35.0, 31.4, 25.7, 24.3. HRMS (ESI) calculated for (C31H35N4O2)+ [M+H]+ 495.2760, found 495.2754.

6-(Pyrrolidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (23)

1H NMR (CDCl3, 400 MHz) δ 7.78 – 7.66 (m, 2H), 7.55 – 7.44 (m, 3H), 7.34 – 7.28 (m, 2H), 7.15 – 7.09 (m, 2H), 7.01 (d, J = 14.9 Hz, 1H), 6.86 (d, J = 7.2 Hz, 1H), 6.58 (d, J = 8.3 Hz, 1H), 3.58 (s, 3H), 3.41 (d, J = 6.4 Hz, 4H), 2.07 (t, J = 6.4 Hz, 4H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.3, 153.3, 151.3, 147.0, 139.0, 137.6, 135.4, 129.4, 128.5, 125.9, 125.8, 122.3, 120.3, 117.8, 111.6, 110.2, 105.8, 52.8, 48.0, 35.0, 31.4, 25.7. MS (ESI) calculated for (C30H33N4O2)+ [M+H]+ 481.3, found 481.2

tert-Butyl-4-(3-(3-(tert-butyl)phenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4-oxo-3,4-dihydroquinazolin-6-yl)piperazine-1-carboxylate (24)

1H NMR (CDCl3, 400 MHz) δ 8.00 (s, 2H), 7.64 (d, J = 2.8 Hz, 1H), 7.57 – 7.44 (m, 4H), 7.29 (d, J = 1.9 Hz, 1H), 7.11 (dt, J = 7.6, 1.6 Hz, 1H), 6.99 (dd, J = 15.0, 1.4 Hz, 1H), 6.95 (d, J = 7.2 Hz, 1H), 6.62 (d, J = 8.3 Hz, 1H), 3.62 (t, J = 5.2 Hz, 4H), 3.56 (d, J = 1.3 Hz, 3H), 3.30 (t, J = 5.3 Hz, 4H), 1.46 (s, 9H), 1.32 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 161.4, 154.6, 153.5, 150.3, 149.9, 139.0, 136.5, 129.5, 129.4, 126.2, 125.5, 125.4, 124.7, 121.3, 118.7, 112.3, 110.4, 80.1, 52.5, 48.7, 43.3, 34.9, 31.2, 28.4, 28.4. MS (ESI) calculated for (C35H42N5O4)+ [M+H]+ 596.3, found 596.3.

6-(Piperazin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone hydrochloride (25)

1H NMR (DMSO-d6, 400 MHz) δ 9.54 (s, 2H), 7.93 – 7.83 (m, 2H), 7.72 – 7.62 (m, 2H), 7.55 (dt, J = 8.0, 1.5 Hz, 1H), 7.53 – 7.44 (m, 3H), 7.23 (dt, J = 7.9, 1.4 Hz, 1H), 7.08 (d, J = 7.2 Hz, 1H), 6.84 (d, J = 15.0 Hz, 1H), 6.72 (d, J = 8.3 Hz, 1H), 3.53 (t, J = 5.1 Hz, 4H), 3.46 (d, J = 1.1 Hz, 3H), 3.21 (s, 4H), 1.24 (d, J = 1.0 Hz, 9H). 13C NMR (DMSO-d6, 100 MHz) δ 163.0, 160.9, 153.1, 150.3, 150.2, 149.3, 140.5, 138.7, 138.0, 137.2, 129.7, 127.0, 126.3, 126.2, 126.0, 124.9, 122.5, 121.5, 119.3, 112.7, 109.9, 52.5, 45.4, 42.7, 35.0, 31.4. MS (ESI) calculated for (C30H34N5O2)+ [M+H]+ 496.3, found 496.3

6-(4-Methylpiperazin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (26)

1H NMR (CDCl3, 400 MHz) δ 7.75 – 7.62 (m, 3H), 7.54 – 7.41 (m, 4H), 7.29 (q, J = 3.4, 2.5 Hz, 1H), 7.14 – 7.06 (m, 1H), 7.04 – 6.95 (m, 1H), 6.84 (d, J = 7.2 Hz, 1H), 6.58 (d, J = 8.6 Hz, 1H), 3.56 (d, J = 1.4 Hz, 3H), 3.43 – 3.31 (m, 4H), 2.63 (t, J = 4.9 Hz, 4H), 2.38 (d, J = 2.6 Hz, 3H), 1.31 (d, J = 1.3 Hz, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.2, 153.2, 150.9, 149.7, 149.1, 138.8, 137.2, 136.0, 129.3, 128.4, 125.8, 125.7, 125.5, 124.2, 124.0, 121.8, 117.7, 111.6, 110.1, 54.7, 52.5, 48.5, 45.9, 34.8, 31.2. MS (ESI) calculated for (C31H36N5O2)+ [M+H]+ 510.3, found 510.3

6-Morpholino-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (27)

1H NMR (CDCl3, 400 MHz) δ 7.78 – 7.68 (m, 2H), 7.65 (d, J = 2.9 Hz, 1H), 7.55 – 7.38 (m, 4H), 7.29 (d, J = 2.0 Hz, 1H), 7.11 (d, J = 7.3 Hz, 1H), 7.00 (d, J = 14.9 Hz, 1H), 6.85 (d, J = 7.1 Hz, 1H), 6.59 (d, J = 8.3 Hz, 1H), 4.00 – 3.80 (m, 4H), 3.57 (s, 3H), 3.29 (dd, J = 5.9, 3.8 Hz, 4H), 1.31 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.2, 153.2, 150.8, 149.9, 149.3, 138.9, 137.1, 129.3, 128.4, 125.9, 125.6, 125.5, 123.7, 121.8, 117.7, 111.7, 109.9, 66.7, 52.5, 48.9, 34.8, 31.2; HRMS (ESI) calculated for (C30H33N4O3)+ [M+H]+ 497.2553, found 497.2536.

6-(4-Morpholinopiperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxyl-2-pyridinyl)ethenyl]-4(3H)-quinazolinone (28)

1H NMR (CDCl3, 400 MHz) δ 7.70 (d, J = 3.0 Hz, 1H), 7.66 (dd, J = 5.8, 2.9 Hz, 2H), 7.54 – 7.42 (m, 4H), 7.29 (d, J = 2.0 Hz, 1H), 7.10 (dt, J = 7.2, 1.5 Hz, 1H), 6.99 (d, J = 14.8 Hz, 1H), 6.84 (d, J = 7.1 Hz, 1H), 6.58 (d, J = 8.3 Hz, 1H), 3.90 (d, J = 12.8 Hz, 2H), 3.74 (t, J = 4.7 Hz, 4H), 3.56 (s, 3H), 2.85 (t, J = 12.2 Hz, 2H), 2.59 (s, 4H), 2.39 (s, 1H), 1.98 (d, J = 12.4 Hz, 2H), 1.67 (q, J = 11.6 Hz, 2H), 1.31 (s, 9H). 13C NMR (CDCl3, 100 MHz) 163.1, 153.1, 150.9, 149.7, 138.8, 137.2, 135.9, 129.3, 128.4, 125.8, 125.7, 125.5, 124.5, 124.2, 117.7, 111.6, 110.1, 61.9, 52.5, 49.7, 48.6, 48.5, 34.8, 31.2, 27.8. MS (ESI) calculated for (C35H42N5O3)+ [M+H]+ 580.3, found 580.3.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-trifluoromethyl-3-pyridinyl)ethenyl]-4(3H)-quinazolinone (35)

1H NMR (CDCl3, 400 MHz) δ 8.62 (s, 1H), 7.83 (d, J = 15.7 Hz, 1H), 7.68 (dd, J = 12.1, 6.1 Hz, 3H), 7.62 – 7.45 (m, 4H), 7.29 (s, 1H), 7.16 – 7.09 (m, 1H), 6.50 (d, J = 15.7 Hz, 1H), 3.40 – 3.29 (m, 4H), 1.74 (s, 4H), 1.64 (d, J = 4.7 Hz, 2H), 1.34 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.2, 153.6, 148.8, 147.6, 136.9, 135.2, 134.4, 132.1, 129.7, 128.6, 126.5, 125.9, 125.8, 125.0, 122.2, 120.6, 110.0, 50.1, 35.1, 31.4, 25.6, 24.3. MS (ESI) calculated for (C31H32F3N4O)+ [M+H]+ 533.3, found 533.2

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(4-trifluoromethyl-3-pyridinyl)ethenyl]-4(3H)-quinazolinone (36)

1H NMR (CDCl3, 100 MHz) δ 7.82 (d, J = 15.6 Hz, 1H), 7.72 – 7.61 (m, 2H), 7.58 – 7.44 (m, 5H), 7.33 (d, J = 8.1 Hz, 2H), 7.27 (d, J = 2.0 Hz, 1H), 7.13 (ddd, J = 7.5, 2.1, 1.1 Hz, 1H), 6.40 (d, J = 15.6 Hz, 1H), 3.32 (t, J = 5.4 Hz, 4H), 1.73 (t, J = 5.8 Hz, 4H), 1.62 (p, J = 5.9 Hz, 2H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.1, 153.2, 148.2, 139.0, 136.8, 135.9, 129.4, 128.3, 127.4, 126.1, 125.7, 125.6, 125.6, 122.8, 121.8, 34.8, 31.2, 29.6, 25.4, 24.1. LC-MS (M + H) = 532.3. MS (ESI) calculated for (C32H33F3N3O)+ [M+H]+ 532.3, found 532.3.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(4-pyridinyl)ethenyl]-4(3H)-quinazolinone (37)

1H NMR (CDCl3, 400 MHz) δ 8.51 (d, J = 5.2 Hz, 2H), 7.77 – 7.61 (m, 3H), 7.59 – 7.44 (m, 3H), 7.26 (d, J = 1.8 Hz, 1H), 7.17 – 7.04 (m, 3H), 6.51 (dd, J = 15.6, 1.2 Hz, 1H), 3.32 (t, J = 5.4 Hz, 4H), 1.72 (t, J = 5.6 Hz, 4H), 1.61 (p, J = 5.7 Hz, 2H), 1.32 (d, J = 1.3 Hz, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.0, 153.3, 150.8, 149.9, 147.6, 143.2, 140.1, 136.7, 134.4, 129.4, 128.4, 126.2, 125.7, 125.6, 125.1, 124.4, 122.0, 121.3, 109.8, 49.9, 34.8, 31.2, 29.6, 25.4, 24.1. LC-MS (M + H) = 465.3. MS (ESI) calculated for (C30H33N4O)+ [M+H]+ 465.3, found 465.3.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(2-naphthalenyl)ethenyl]-4(3H)-quinazolinone (38)

1H NMR (CDCl3, 400 MHz) δ 8.00 (d, J = 15.5 Hz, 1H), 7.82 – 7.62 (m, 6H), 7.61 – 7.38 (m, 5H), 7.33 – 7.25 (m, 2H), 7.17 (dt, J = 7.5, 1.6 Hz, 1H), 6.45 (d, J = 15.5 Hz, 1H), 3.31 (t, J = 5.4 Hz, 4H), 1.74 (s, 4H), 1.62 (s, 2H), 1.34 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.1, 153.2, 136.9, 133.7, 133.3, 133.0, 129.4, 129.1, 128.4, 128.3, 127.6, 126.8, 126.5, 126.1, 125.7, 125.7, 123.1, 121.5, 34.9, 31.2, 29.6, 25.3, 23.9. MS (ESI) calculated for (C35H36N3O)+ [M+H]+ 514.3, found 514.3.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-(2,6-dimethoxystyryl)-4(3H)-quinazolinone (39)

1H NMR (CDCl3, 400 MHz) δ 8.27 (d, J = 15.9 Hz, 1H), 7.72 (d, J = 9.0 Hz, 1H), 7.65 (d, J = 2.6 Hz, 1H), 7.55 – 7.43 (m, 3H), 7.27 (d, J = 2.0 Hz, 1H), 7.18 – 7.08 (m, 2H), 6.95 (d, J = 15.8 Hz, 1H), 6.45 (d, J = 8.4 Hz, 2H), 3.65 (s, 6H), 3.28 (t, J = 5.4 Hz, 4H), 1.73 (s, 4H), 1.61 (q, J = 5.9 Hz, 2H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.5, 159.6, 152.9, 137.7, 130.0, 129.1, 128.3, 125.8, 125.7, 125.5, 123.2, 121.3, 113.3, 103.5, 55.3, 50.5, 34.8, 31.2, 29.6, 25.4, 25.2, 24.0. LC-MS (M + H) = 524.3. MS (ESI) calculated for (C33H38N3O3)+ [M+H]+ 524.3, found 524.3.

6-(4-Methylpiperazin-1-yl)-3-(3-tert-butylphenyl)-2-(2,6-dimethoxystyryl)-4(3H)-quinazolinone (40)

1H NMR (CDCl3, 400 MHz) δ 8.28 (d, J = 15.8 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H), 7.64 (d, J = 2.9 Hz, 1H), 7.52 (dt, J = 8.1, 1.4 Hz, 1H), 7.48 (d, J = 7.6 Hz, 1H), 7.46 – 7.39 (m, 1H), 7.27 (d, J = 1.9 Hz, 1H), 7.15 (d, J = 8.4 Hz, 1H), 7.13 – 7.08 (m, 1H), 6.94 (d, J = 15.9 Hz, 1H), 6.45 (d, J = 8.4 Hz, 2H), 3.65 (s, 6H), 3.39 (t, J = 5.0 Hz, 4H), 2.70 (t, J = 4.9 Hz, 4H), 2.42 (s, 1H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 159.5, 152.9, 148.9, 137.7, 130.0, 129.1, 128.8, 128.5, 125.8, 125.7, 125.5, 124.2, 123.3, 121.3, 113.3, 110.3, 103.5, 55.3, 55.3, 54.6, 48.4, 45.6, 34.8, 31.2, 31.1. MS (ESI) calculated for (C33H39N4O3)+ [M+H]+ 539.3, found 539.3.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-(3-methoxystyryl)-4(3H)-quinazolinone (41)

1H NMR (CDCl3, 400 MHz) δ 7.78 (d, J = 15.5 Hz, 1H), 7.71 – 7.62 (m, 2H), 7.57 – 7.42 (m, 3H), 7.27 (d, J = 4.0 Hz, 1H), 7.18 (t, J = 7.9 Hz, 1H), 7.12 (ddd, J = 7.5, 2.1, 1.2 Hz, 1H), 6.89 – 6.83 (m, 1H), 6.83 – 6.76 (m, 2H), 6.32 (d, J = 15.6 Hz, 1H), 3.73 (s, 3H), 3.30 (t, J = 5.4 Hz, 4H), 1.73 (m, 4H), 1.61 (t, J = 5.3 Hz, 2H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 159.7, 153.1, 148.9, 137.8, 137.0, 137.0, 129.6, 129.3, 128.2, 126.0, 125.7, 125.7, 124.8, 121.6, 120.7, 120.0, 114.8, 112.6, 55.1, 34.8, 31.2, 29.6, 25.4, 24.1. MS (ESI) calculated for (C32H36N3O2)+ [M+H]+ 494.3, found 494.3.

tert-Butyl 4-[3-(3-tert-butylphenyl)-2-(3-methoxystyryl)-4-oxo-3,4-dihydroquinazolin-6-yl]piperazine-1-carboxylate (42)

1H NMR (CDCl3, 400 MHz) δ 7.82 (d, J = 15.5 Hz, 1H), 7.73 (d, J = 9.0 Hz, 1H), 7.64 (d, J = 2.8 Hz, 1H), 7.54 (dt, J = 8.1, 1.5 Hz, 1H), 7.52 – 7.41 (m, 2H), 7.27 (d, J = 3.9 Hz, 1H), 7.18 (t, J = 7.9 Hz, 1H), 7.12 (ddd, J = 7.5, 2.0, 1.2 Hz, 1H), 6.85 (d, J = 7.7 Hz, 1H), 6.83 – 6.79 (m, 1H), 6.78 (t, J = 2.0 Hz, 1H), 6.31 (d, J = 15.5 Hz, 1H), 3.73 (s, 3H), 3.60 (t, J = 5.1 Hz, 4H), 3.26 (t, J = 5.1 Hz, 4H), 1.47 (s, 9H), 1.32 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 162.0, 159.7, 154.6, 153.2, 149.7, 149.5, 136.9, 136.8, 129.6, 129.4, 128.2, 126.1, 125.6, 125.6, 124.7, 121.5, 120.0, 115.0, 112.7, 110.5, 80.0, 55.1, 48.9, 34.8, 31.2, 28.4. MS (ESI) calculated for (C36H43N4O4)+ [M+H]+ 595.3, found 595.3.

6-(Piperidin-1-yl)-3-(m-tolyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (44)

1H NMR (CDCl3, 400 MHz) δ 7.75 – 7.63 (m, 3H), 7.48 (dd, J = 8.3, 7.2 Hz, 2H), 7.42 (t, J = 7.7 Hz, 1H), 7.29 (d, J = 7.7 Hz, 1H), 7.16 – 7.03 (m, 3H), 6.86 (d, J = 7.2 Hz, 1H), 6.59 (d, J = 8.3 Hz, 1H), 3.61 (d, J = 1.2 Hz, 3H), 3.31 (t, J = 5.4 Hz, 4H), 2.41 (s, 3H), 1.74 (p, J = 5.6 Hz, 4H), 1.66 – 1.60 (m, 2H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.2, 151.0, 139.7, 138.9, 137.4, 129.5, 129.4, 129.2, 128.3, 125.7, 124.1, 121.8, 117.6, 111.5, 52.5, 29.6, 25.5, 24.1, 21.2. MS (ESI) calculated for (C28H29N4O2)+ [M+H]+ 453.2, found 453.2.

6-Morpholino-3-(m-tolyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (45)

1H NMR (CDCl3, 400 MHz) δ 7.81 – 7.69 (m, 2H), 7.64 (d, J = 2.9 Hz, 1H), 7.51 – 7.39 (m, 3H), 7.29 (d, J = 7.7 Hz, 1H), 7.15 – 7.04 (m, 3H), 6.87 (d, J = 7.2 Hz, 1H), 6.60 (d, J = 8.3 Hz, 1H), 3.89 (t, J = 4.8 Hz, 4H), 3.61 (s, 3H), 3.38 – 3.17 (m, 4H), 2.41 (s, 3H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.0, 150.8, 149.9, 149.2, 139.8, 138.9, 137.2, 129.7, 129.5, 129.1, 128.2, 125.6, 123.7, 121.7, 117.9, 111.8, 109.9, 66.6, 52.5, 48.8, 21.2. MS (ESI) calculated for (C27H27N4O3)+ [M+H]+ 455.2, found 455.2.

6-(4-Morpholinopiperidin-1-yl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-3-(m-tolyl)-4(3H)-quinazolinone (46)

1H NMR (CDCl3, 400 MHz) δ 7.74 – 7.61 (m, 3H), 7.51 – 7.38 (m, 3H), 7.29 (d, J = 7.7 Hz, 1H), 7.15 – 7.03 (m, 3H), 6.85 (d, J = 7.1 Hz, 1H), 6.59 (d, J = 8.2 Hz, 1H), 3.90 (d, J = 12.5 Hz, 2H), 3.75 (bs, 4H), 3.60 (s, 3H), 2.91 – 2.77 (m, 2H), 2.61 (bs, 4H), 2.40 (s, 4H), 2.05 – 1.92 (m, 2H), 1.68 (d, J = 12.2 Hz, 2H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 150.9, 149.7, 148.7, 140.8, 139.8, 138.9, 137.4, 136.0, 129.5, 129.5, 129.2, 128.4, 125.7, 124.5, 124.1, 121.8, 117.6, 111.6, 110.2, 62.0, 49.7, 48.5, 21.2. MS (ESI) calculated for (C32H36N5O3)+ [M+H]+ 538.3, found 538.2

6-(Piperidin-1-yl)-3-(3-methoxyphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (47)

1H NMR (CDCl3, 400 MHz) δ 7.74 – 7.62 (m, 3H), 7.53 – 7.39 (m, 3H), 7.09 (d, J = 14.9 Hz, 1H), 7.02 (ddd, J = 8.5, 2.6, 1.0 Hz, 1H), 6.90 (ddd, J = 7.7, 1.9, 0.9 Hz, 1H), 6.88 – 6.83 (m, 2H), 6.59 (d, J = 8.2 Hz, 1H), 3.80 (s, 3H), 3.63 (s, 3H), 3.31 (t, J = 5.4 Hz, 4H), 1.73 (t, J = 5.7 Hz, 4H), 1.62 (q, J = 6.0 Hz, 2H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.1, 160.6, 151.0, 148.5, 138.9, 138.6, 136.0, 130.3, 128.3, 124.6, 124.0, 121.7, 121.0, 117.6, 115.0, 114.1, 111.5, 55.4, 52.6, 25.5, 24.1; HRMS (ESI) calculated for (C28H29N4O3)+ [M+H]+ 469.2240, found 469.2227.

6-Morpholino-3-(3-methoxyphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (48)

1H NMR (CDCl3, 400 MHz) δ 7.80 – 7.69 (m, 2H), 7.64 (d, J = 2.9 Hz, 1H), 7.53 – 7.39 (m, 3H), 7.09 (d, J = 14.8 Hz, 1H), 7.03 (dd, J = 8.5, 2.7 Hz, 1H), 6.94 – 6.83 (m, 3H), 6.60 (d, J = 8.2 Hz, 1H), 3.89 (t, J = 4.8 Hz, 4H), 3.81 (d, J = 1.7 Hz, 3H), 3.63 (d, J = 1.7 Hz, 3H), 3.29 (dd, J = 5.8, 3.7 Hz, 4H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 161.9, 160.7, 150.8, 149.9, 149.1, 138.9, 138.3, 130.4, 128.2, 123.7, 121.6, 120.9, 117.8, 115.1, 114.0, 111.8, 109.9, 66.6, 55.5, 52.6, 48.8. MS (ESI) calculated for (C27H27N4O4)+ [M+H]+ 471.2, found 471.2

6-(4-Morpholinopiperidin-1-yl)-3-(3-methoxyphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (49)

1H NMR (CDCl3, 400 MHz) δ 7.74 – 7.62 (m, 3H), 7.51 – 7.40 (m, 3H), 7.09 (d, J = 14.9 Hz, 1H), 7.03 (ddd, J = 8.5, 2.6, 1.0 Hz, 1H), 6.90 (ddd, J = 7.8, 1.9, 1.0 Hz, 1H), 6.85 (d, J = 6.8 Hz, 2H), 6.59 (d, J = 8.3 Hz, 1H), 3.91 (d, J = 12.5 Hz, 2H), 3.80 (d, J = 1.1 Hz, 3H), 3.75 (s, 4H), 3.62 (d, J = 1.1 Hz, 3H), 2.85 (td, J = 12.5, 2.2 Hz, 2H), 2.62 (d, J = 6.1 Hz, 4H), 2.07 – 1.93 (m, 2H), 1.77 – 1.62 (m, 2H). 13C NMR (CDCl3, 100 MHz)) δ 163.1, 162.3, 153.3, 151.6, 150.9, 150.6, 139.4, 138.9, 136.9, 136.7, 133.3, 129.4, 127.7, 126.7, 126.0, 125.9, 125.7, 125.5, 124.6, 121.3, 118.1, 112.0, 52.6, 34.8, 34.6, 31.3, 31.2. MS (ESI) calculated for (C32H36N5O4)+ [M+H]+ 554.3, found 554.3.

6-(Piperidin-1-yl)-3-(3-trifluoromethylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (50)

1H NMR (CDCl3, 400 MHz) δ 7.83 – 7.59 (m, 6H), 7.58 – 7.42 (m, 3H), 6.97 (dd, J = 14.8, 1.7 Hz, 1H), 6.86 (d, J = 7.1 Hz, 1H), 6.60 (t, J = 6.1 Hz, 1H), 3.56 (s, 3H), 3.42 – 3.23 (m, 4H), 1.81 – 1.68 (m, 4H), 1.63 (q, J = 5.8 Hz, 2H). 13C NMR (CDCl3, 100 MHz) δ 163.2, 162.1, 150.6, 138.9, 138.2, 136.5, 132.6, 130.3, 128.5, 126.0, 125.6, 124.6, 123.2, 121.5, 117.9, 111.9, 52.4, 25.4, 24.1. MS (ESI) calculated for (C28H26F3N4O2)+ [M+H]+ 507.2, found 507.2.

6-Morpholino-3-(3-trifluoromethylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (51)

1H NMR (CDCl3, 400 MHz) δ 7.80 – 7.67 (m, 4H), 7.65 (s, 1H), 7.62 (d, J = 2.9 Hz, 1H), 7.55 – 7.42 (m, 3H), 6.97 (d, J = 14.7 Hz, 1H), 6.86 (d, J = 7.1 Hz, 1H), 6.61 (d, J = 8.2 Hz, 1H), 4.00 – 3.82 (m, 4H), 3.56 (s, 3H), 3.38 – 3.22 (m, 4H). 13C NMR (CDCl3, 100 MHz) δ 163.2, 161.9, 150.4, 150.1, 148.3, 138.9, 138.1, 137.2, 132.5, 132.2, 130.4, 128.5, 126.0, 125.9, 125.8, 125.7, 124.7, 123.8, 122.9, 121.5, 118.0, 112.1, 109.7, 66.6, 52.4, 48.7. MS (ESI) calculated for (C27H24F3N4O3)+ [M+H]+ 509.2, found 509.2.

6-(4-Morpholinopiperidin-1-yl)-3-(3-(trifluoromethyl)phenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (52)

1H NMR (CDCl3, 400 MHz) δ 7.82 – 7.56 (m, 6H), 7.56 – 7.38 (m, 3H), 6.97 (dt, J = 14.9, 3.0 Hz, 1H), 6.86 (dd, J = 7.1, 1.9 Hz, 1H), 6.60 (dd, J = 8.3, 1.9 Hz, 1H), 3.91 (d, J = 12.4 Hz, 2H), 3.74 (s, 4H), 3.56 (d, J = 1.9 Hz, 3H), 2.87 (t, J = 12.5 Hz, 2H), 2.60 (s, 4H), 2.48 – 2.35 (m, 1H), 1.99 (d, J = 12.4 Hz, 2H), 1.76 – 1.57 (m, 2H). 13C NMR (CDCl3, 100 MHz) δ 163.2, 162.0, 150.5, 149.9, 147.9, 140.5, 138.9, 138.2, 136.7, 132.5, 130.3, 128.6, 125.9, 125.7, 124.6, 123.2, 121.5, 117.9, 112.0, 110.0, 61.9, 52.4, 49.7, 48.4, 27.7. LC-MS (M + H) = 592.3. MS (ESI) calculated for (C32H33F3N5O3)+ [M+H]+ 592.3, found 592.2

General synthetic methods for compounds 29–34:

A mixture of compound 58 (0.25 mmol, 1 eq), an aryl or vinyl boronic acid (0.30 mmol, 1.2 eq), tetrakis(triphenylphosphine)palladium(0) (0.029 g, 0.0125 mmol, 0.05 eq), and Na2CO3 (0.053 g, 0.5 mmol, 2 eq) in 1,4-dioxane/H2O (8/2 mL) was heated to 110 °C for 24 h. The reaction mixture was cooled down and quenched with brine (20 mL). The product was extracted with ethyl acetate (3 × 20 mL), and the combined organic phase was washed with brine and then dried over Na2SO4. Upon removal of solvent, the residue was purified with column chromatography (silica gel, hexanes : ethyl acetate, 9:1 – 3:1) to give the target compounds as an off-white or pale yellow solid in 45–80% yield.

6-(Pyridin-4-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone dihydrochloride (29)

1H NMR (CDCl3, 400 MHz) δ 8.71 (d, J = 5.4 Hz, 2H), 8.62 (d, J = 2.2 Hz, 1H), 8.08 (dd, J = 8.6, 2.2 Hz, 1H), 7.91 (d, J = 8.5 Hz, 1H), 7.85 (d, J = 14.8 Hz, 1H), 7.77 – 7.70 (m, 2H), 7.65 (ddd, J = 12.0, 8.2, 1.4 Hz, 1H), 7.58 – 7.43 (m, 4H), 7.32 (t, J = 1.9 Hz, 1H), 7.14 (ddd, J = 7.4, 2.2, 1.2 Hz, 1H), 7.04 (d, J = 14.8 Hz, 1H), 6.91 (d, J = 7.1 Hz, 1H), 6.63 (d, J = 8.3 Hz, 1H), 3.58 (s, 3H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.2, 162.0, 153.5, 152.9, 150.4, 148.7, 148.6, 138.9, 138.5, 136.6, 135.2, 132.8, 132.1, 132.0, 132.0, 131.9, 131.9, 129.5, 128.6, 128.5, 128.4, 126.2, 125.9, 125.5, 125.4, 123.6, 121.8, 121.6, 118.3, 112.3, 52.6, 34.9, 31.2. MS (ESI) calculated for (C31H29N4O2)+ [M+H]+ 489.2, found 489.2

6-(3-Methoxyphenyl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (30)

1H NMR (CDCl3, 400 MHz) δ 8.56 – 8.50 (m, 1H), 8.03 (dd, J = 8.5, 2.2 Hz, 1H), 7.90 – 7.78 (m, 2H), 7.57 – 7.44 (m, 3H), 7.38 (t, J = 7.9 Hz, 1H), 7.35 – 7.26 (m, 2H), 7.23 (dd, J = 2.5, 1.6 Hz, 1H), 7.14 (ddd, J = 7.4, 2.1, 1.3 Hz, 1H), 7.04 (d, J = 14.8 Hz, 1H), 6.93 (ddd, J = 8.2, 2.6, 1.0 Hz, 1H), 6.90 (d, J = 7.1 Hz, 1H), 6.62 (dd, J = 8.3, 0.7 Hz, 1H), 3.87 (s, 3H), 3.58 (s, 3H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.3, 160.1, 153.3, 151.8, 150.6, 141.1, 139.4, 138.9, 137.7, 136.9, 133.5, 129.9, 129.4, 127.8, 126.0, 125.6, 125.5, 125.0, 123.8, 121.3, 119.6, 118.1, 113.4, 112.6, 112.0, 55.3, 52.6, 34.8, 31.2. MS (ESI) calculated for (C33H32N3O3)+ [M+H]+ 518.2, found 518.2.

6-(4-fluorophenyl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (31)

1H NMR (CDCl3, 400 MHz) δ 8.47 (d, J = 2.2 Hz, 1H), 7.99 (dd, J = 8.5, 2.2 Hz, 1H), 7.90 – 7.78 (m, 2H), 7.70 – 7.61 (m, 2H), 7.57 – 7.45 (m, 3H), 7.32 (d, J = 2.0 Hz, 1H), 7.20 – 7.11 (m, 3H), 7.04 (d, J = 14.8 Hz, 1H), 6.89 (d, J = 7.1 Hz, 1H), 6.62 (d, J = 8.3 Hz, 1H), 3.58 (s, 3H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 153.4, 151.8, 150.6, 138.9, 138.5, 136.8, 133.2, 129.4, 128.8, 128.7, 127.9, 126.1, 125.6, 125.5, 124.8, 121.3, 118.1, 116.0, 115.8, 112.1, 52.6, 34.8, 31.2. MS (ESI) calculated for (C32H29N3O2)+ [M+H]+ 506.2, found 506.2

6-(4-dimethylaminophenyl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (32)

1H NMR (CDCl3, 400 MHz) δ 8.49 (d, J = 2.3 Hz, 1H), 8.02 (ddd, J = 8.5, 2.3, 0.9 Hz, 1H), 7.85 – 7.74 (m, 2H), 7.68 – 7.59 (m, 2H), 7.56 – 7.44 (m, 3H), 7.32 (d, J = 2.0 Hz, 1H), 7.18 – 7.11 (m, 1H), 7.04 (d, J = 14.8 Hz, 1H), 6.88 (d, J = 7.1 Hz, 1H), 6.83 (d, J = 8.3 Hz, 2H), 6.60 (d, J = 8.2 Hz, 1H), 3.58 (d, J = 1.0 Hz, 3H), 3.01 (s, 6H), 1.32 (d, J = 1.0 Hz, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.4, 153.2, 151.0, 150.8, 146.0, 139.6, 138.9, 137.1, 137.0, 132.6, 129.3, 127.7, 127.7, 125.9, 125.7, 125.5, 124.1, 123.3, 121.3, 117.9, 112.8, 111.8, 52.6, 40.5, 34.8, 31.2, 29.6. MS (ESI) calculated for (C34H35N4O2)+ [M+H]+ 531.3, found 531.3.

6-[4-(Pyrrolidin-1-yl)phenyl]-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (33)

1H NMR (CDCl3, 400 MHz) δ 8.48 (d, J = 2.2 Hz, 1H), 8.02 (dd, J = 8.6, 2.2 Hz, 1H), 7.85 – 7.74 (m, 2H), 7.63 (d, J = 8.6 Hz, 2H), 7.56 – 7.44 (m, 3H), 7.32 (d, J = 1.9 Hz, 1H), 7.14 (dt, J = 7.4, 1.7 Hz, 1H), 7.03 (d, J = 14.8 Hz, 1H), 6.88 (d, J = 7.1 Hz, 1H), 6.66 (d, J = 8.3 Hz, 2H), 6.60 (d, J = 8.3 Hz, 1H), 3.58 (s, 3H), 3.34 (t, J = 8.0, 4H), 2.13 – 1.91 (m, 4H), 1.33 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.4, 153.2, 150.8, 150.8, 147.6, 139.9, 138.9, 137.1, 132.5, 129.3, 127.8, 127.6, 125.9, 125.7, 125.5, 124.1, 123.1, 121.3, 117.9, 111.8, 52.6, 47.6, 34.8, 31.2, 25.4. MS (ESI) calculated for (C36H37N4O2)+ [M+H]+ 557.3, found 557.3.

6-(3,6-dihydro-2H-pyran-4-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)vinyl]-4(3H)-quinazolinone (34)

1H NMR (CDCl3, 400 MHz) δ 8.27 (d, J = 2.1 Hz, 1H), 7.88 (dd, J = 8.6, 2.2 Hz, 1H), 7.84 – 7.72 (m, 2H), 7.56 – 7.44 (m, 3H), 7.30 (d, J = 2.0 Hz, 1H), 7.12 (ddd, J = 7.4, 2.2, 1.3 Hz, 1H), 7.02 (d, J = 14.8 Hz, 1H), 6.88 (d, J = 7.2 Hz, 1H), 6.61 (d, J = 8.3 Hz, 1H), 6.32 (tt, J = 3.2, 1.5 Hz, 1H), 4.37 (q, J = 2.8 Hz, 2H), 3.96 (t, J = 5.4 Hz, 2H), 3.57 (s, 3H), 2.69 – 2.51 (m, 2H), 1.32 (s, 9H). 13C NMR (CDCl3, 100 MHz) δ 163.1, 162.2, 153.3, 151.6, 150.6, 138.9, 138.4, 136.8, 133.0, 131.0, 129.4, 127.4, 126.0, 125.6, 125.4, 123.8, 122.4, 120.9, 118.1, 112.0, 65.8, 64.3, 52.6, 34.8, 31.2, 26.9. MS (ESI) calculated for (C31H32N3O3)+ [M+H]+ 494.2, found 494.3.

General synthetic methods for compounds 53–54:

A mixture of a compound (22, 27) (0.25 mmol, 1 eq) and palladium on activated charcoal (10% w/w) in methanol was stirred under hydrogen gas. Upon completion, the reaction mixture was filtered using Celite bed, and solvent was removed under reduced pressure. Obtained residue was purified with column chromatography (silica gel, hexanes : ethyl acetate, 9:1 – 3:1) to give the target compounds as an off-white or orange solid in 80–88% yield.

6-(Piperidin-1-yl)-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)ethyl]-4(3H)-quinazolinone (53)

1H NMR (CDCl3, 400 MHz) δ 7.68 – 7.52 (m, 2H), 7.52 – 7.28 (m, 4H), 7.28 – 7.17 (m, 1H), 7.06 – 6.95 (m, 1H), 6.61 (d, J = 7.2 Hz, 1H), 6.46 (d, J = 8.2 Hz, 1H), 3.70 (s, 3H), 3.26 (t, J = 5.5 Hz, 4H), 3.08 (t, J = 7.7 Hz, 2H), 2.88 – 2.68 (m, 2H), 1.71 (q, J = 5.7 Hz, 4H), 1.59 (t, J = 6.2 Hz, 2H), 1.31 (s, 9H).13C NMR (100 MHz, cdcl3) δ 162.5, 157.8, 153.1, 138.6, 137.2, 129.3, 125.9, 125.3, 125.2, 121.3, 115.3, 107.9, 52.9, 34.8, 34.6, 34.6, 31.2, 25.4, 24.0. MS (ESI) calculated for (C31H37N4O2)+ [M+H]+ 497.3, found 497.3.

6-Morpholino-3-(3-tert-butylphenyl)-2-[2-(6-methoxypyridin-2-yl)ethyl]-4(3H)-quinazolinone (54)

1H NMR (400 MHz, Chloroform-d) δ 7.65 (d, J = 8.9 Hz, 1H), 7.61 (d, J = 2.8 Hz, 1H), 7.52 – 7.34 (m, 4H), 7.24 (d, J = 2.8 Hz, 1H), 7.01 (dt, J = 7.6, 1.6 Hz, 1H), 6.62 (d, J = 7.2 Hz, 1H), 6.47 (d, J = 8.2 Hz, 1H), 3.87 (t, J = 4.8 Hz, 4H), 3.69 (s, 3H), 3.25 (t, J = 4.8 Hz, 4H), 3.09 (t, J = 7.7 Hz, 2H), 2.85 (t, J = 7.5 Hz, 2H), 1.31 (s, 9H). 13C NMR (100 MHz, Chloroform-d) δ 163.4, 153.2, 149.8, 138.6, 129.3, 126.1, 125.2, 125.2, 123.8, 115.3, 109.9, 108.0, 66.7, 52.9, 49.0, 34.8, 34.6, 31.2, 29.6. MS (ESI) calculated for (C30H35N4O3)+ [M+H]+ 499.3, found 499.3.

Cellular antiviral assays.

The cellular antiviral activities of these compounds were determined in Vero, U87 glioma or mosquito C6/36 cells, following our previous methods 34. Briefly, 2 × 104 cells/well were seeded in 96-well plates and cultured in DMEM media with 2% FBS to form a monolayer of cells. For regular compound treatment (2h-pretreatment), 0.1 MOI (multiplicity of infection) of ZIKV or DENV, together with various concentrations of a compound, was added to each well. After incubation for 2h, the supernatant was removed and cells were washed with PBS to remove unattached viral particles. Fresh medium (150 μL/well) containing various concentrations of a compound in duplicate were added. For 0h-post-treatment, a compound was added into cell culture media right after removal of the unattached viruses. For 2h-post-treatment, a compound was added into cell culture media 2 hours after removal of the unattached viruses. Upon incubation at 37 °C for 48h, aliquots of the supernatant from each well were used to determine viral TCID50 (tissue culture infectious dose) using end-point dilution assay. Half-log serial dilution of the viral supernatant (50 μL) was added to a monolayer of Vero cells in quadruplicate in 96-well plates and cultured for 5–7 days. CPE or cell lysis was determined with microscope followed by MTT assay. TCID50 was calculated based on the highest dilution in which ≥50% (i.e., ≥2 out of the 4 quadruplicate wells) of Vero cells were infected with ZIKV. Compared to controls, the ability for a compound to reduce TCID50 can be determined. The results were from at least 2 independent experiments. Inhibition data were imported into Prism (version 5.0) and EC50 values with standard deviation were obtained by using a standard dose-response curve fitting.

Quantitative RT-PCR to determine ZIKV RNA copies.

Viral RNA was extracted from the supernatant (50 μL) using TRIzol (ThermoFisher). qPCR is based on amplification of ZIKV envelope gene region 3, using ZIKV-specific primers and probes, following our previous method 34. PCR was performed using a TaqMan Fast Virus 1-step Master Mix kit on a StepOnePlus RT-PCR system (Applied Biosystems). Concentrations of ZIKV RNA (copies/mL) were calculated by using a standard curve.

Western blot.

8 × 105 Vero cells in petri dishes were infected with ZIKA (MOI = 0.1) for 2h. Upon removal of the virus, the cells were cultured with fresh media contain 2% FBS and compound 27 for 48h. The cells were harvested and lysed using ice-cold radioimmunoprecipitation assay buffer (Invitrogen). Equal amounts of total proteins were then separated on SDS-PAGE and transferred to PVDF membranes. The blots were probed and visualized with primary antibodies against Zika virus NS3 (GTX133309, GeneTex), Zika virus capsid (GTX133317, GeneTex), Zika virus NS5 (GTX133327, GeneTex), and human β-Actin (4967S, Cell Signaling).

Supplementary Material

Supporting Information PDF

Acknowledgement

This work was supported by a grant (W81XWH-18-1-0368) from USAMRAA of the US Department of Defense, a grant (RP220232) from Cancer Prevention and Research Institute of Texas, and a grant (R01CA266057) from the United States National Institute of Health/National Cancer Institute to Y.S.

Abbreviation:

SAR

structure–activity relationships

DENV

Dengue virus

ZIKV

Zika virus

RNA

Ribonucleic acid

EC50

half maximal effective concentration

WHO

World Health Organization

CPE

cytopathic effects

MOI

multiplicity of infection

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide

TCID50

half maximal tissue culture infectious dose

LCMS

liquid chromatography–mass spectrometry

ESI

electrospray ionization

HPCL

high performance liquid chromatography

DMF

N,N-dimethylformamide

Footnotes

Declaration of competing interest

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.

Supporting Information

Figure S1, HPLC tracers of selected most active compounds, and Molecular Formula Strings for all compounds are available free of charge via the Internet at http://pubs.acs.org.

References

  • 1.Moreira J; Peixoto TM; Siqueira AM; Lamas CC Sexually acquired Zika virus: a systematic review. Clin Microbiol Infect 2017, 23, 296–305. [DOI] [PubMed] [Google Scholar]
  • 2.Nitsche C; Holloway S; Schirmeister T; Klein CD Biochemistry and medicinal chemistry of the dengue virus protease. Chem Rev 2014, 114, 11348–11381. [DOI] [PubMed] [Google Scholar]
  • 3.Dick GW; Kitchen SF; Haddow AJ Zika virus. I. Isolations and serological specificity. Trans R Soc Trop Med Hyg 1952, 46, 509–520. [DOI] [PubMed] [Google Scholar]
  • 4.Yun SI; Lee YM Zika virus: An emerging flavivirus. J Microbiol 2017, 55, 204–219. [DOI] [PubMed] [Google Scholar]
  • 5.Medin CL; Rothman AL Zika Virus: The Agent and Its Biology, With Relevance to Pathology. Arch Pathol Lab Med 2017, 141, 33–42. [DOI] [PubMed] [Google Scholar]
  • 6.ECDC. Rapid risk assessment: Zika virus infection outbreak, French Polynesia. European Centre for Disease Prevention and Control, Stockholm, Sweden. February 14, 2014. 2014. [Google Scholar]
  • 7.Oehler E; Watrin L; Larre P; Leparc-Goffart I; Lastere S; Valour F; Baudouin L; Mallet H; Musso D; Ghawche F Zika virus infection complicated by Guillain-Barre syndrome--case report, French Polynesia, December 2013. Euro Surveill 2014, 19. [DOI] [PubMed] [Google Scholar]
  • 8.Cao-Lormeau VM; Blake A; Mons S; Lastere S; Roche C; Vanhomwegen J; Dub T; Baudouin L; Teissier A; Larre P; Vial AL; Decam C; Choumet V; Halstead SK; Willison HJ; Musset L; Manuguerra JC; Despres P; Fournier E; Mallet HP; Musso D; Fontanet A; Neil J; Ghawche F Guillain-Barre Syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet 2016, 387, 1531–1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Schuler-Faccini L; Ribeiro EM; Feitosa IM; Horovitz DD; Cavalcanti DP; Pessoa A; Doriqui MJ; Neri JI; Neto JM; Wanderley HY; Cernach M; El-Husny AS; Pone MV; Serao CL; Sanseverino MT Possible Association Between Zika Virus Infection and Microcephaly - Brazil, 2015. MMWR Morb Mortal Wkly Rep 2016, 65, 59–62. [DOI] [PubMed] [Google Scholar]
  • 10.Victora CG; Schuler-Faccini L; Matijasevich A; Ribeiro E; Pessoa A; Barros FC Microcephaly in Brazil: how to interpret reported numbers? Lancet 2016, 387, 621–624. [DOI] [PubMed] [Google Scholar]
  • 11. https://www.cdc.gov/media/releases/2017/s0302-zika-birth-defects.html. In.
  • 12.Nash D; Mostashari F; Fine A; Miller J; O’Leary D; Murray K; Huang A; Rosenberg A; Greenberg A; Sherman M; Wong S; Layton M The outbreak of West Nile virus infection in the New York City area in 1999. N Engl J Med 2001, 344, 1807–1814. [DOI] [PubMed] [Google Scholar]
  • 13.From the Centers for Disease Control and Prevention. Update: West Nile-like viral encephalitis--New York, 1999. JAMA 1999, 282, 1714. [PubMed] [Google Scholar]
  • 14.Tricarico PM; Caracciolo I; Crovella S; D’Agaro P Zika virus induces inflammasome activation in the glial cell line U87-MG. Biochem Biophys Res Commun 2017, 492, 597–602. [DOI] [PubMed] [Google Scholar]
  • 15.Yildiz M; Ghosh S; Bell JA; Sherman W; Hardy JA Allosteric inhibition of the NS2B-NS3 protease from dengue virus. ACS Chem Biol 2013, 8, 2744–2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Murray KO; Gorchakov R; Carlson AR; Berry R; Lai L; Natrajan M; Garcia MN; Correa A; Patel SM; Aagaard K; Mulligan MJ Prolonged Detection of Zika Virus in Vaginal Secretions and Whole Blood. Emerg Infect Dis 2017, 23, 99–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Armstrong PM; Rico-Hesse R Efficiency of dengue serotype 2 virus strains to infect and disseminate in Aedes aegypti. Am J Trop Med Hyg 2003, 68, 539–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Arsuaga M; Bujalance SG; Diaz-Menendez M; Vazquez A; Arribas JR Probable sexual transmission of Zika virus from a vasectomised man. Lancet Infect Dis 2016, 16, 1107. [DOI] [PubMed] [Google Scholar]
  • 19.Quintana VM; Selisko B; Brunetti JE; Eydoux C; Guillemot JC; Canard B; Damonte EB; Julander JG; Castilla V Antiviral activity of the natural alkaloid anisomycin against dengue and Zika viruses. Antiviral Res 2020, 176, 104749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Estoppey D; Lee CM; Janoschke M; Lee BH; Wan KF; Dong H; Mathys P; Filipuzzi I; Schuhmann T; Riedl R; Aust T; Galuba O; McAllister G; Russ C; Spiess M; Bouwmeester T; Bonamy GMC; Hoepfner D The Natural Product Cavinafungin Selectively Interferes with Zika and Dengue Virus Replication by Inhibition of the Host Signal Peptidase. Cell Rep 2017, 19, 451–460. [DOI] [PubMed] [Google Scholar]
  • 21.Brecher M; Li Z; Liu B; Zhang J; Koetzner CA; Alifarag A; Jones SA; Lin Q; Kramer LD; Li H A conformational switch high-throughput screening assay and allosteric inhibition of the flavivirus NS2B-NS3 protease. PLoS Pathog 2017, 13, e1006411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Rothan HA; Zhong Y; Sanborn MA; Teoh TC; Ruan J; Yusof R; Hang J; Henderson MJ; Fang S Small molecule grp94 inhibitors block dengue and Zika virus replication. Antiviral Res 2019, 171, 104590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Oeyen M; Meyen E; Noppen S; Claes S; Doijen J; Vermeire K; Sussmuth RD; Schols D Labyrinthopeptin A1 inhibits dengue and Zika virus infection by interfering with the viral phospholipid membrane. Virology 2021, 562, 74–86. [DOI] [PubMed] [Google Scholar]
  • 24.Lin CS; Huang SH; Yan BY; Lai HC; Lin CW Effective Antiviral Activity of the Tyrosine Kinase Inhibitor Sunitinib Malate against Zika Virus. Infect Chemother 2021, 53, 730–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yang S; Xu M; Lee EM; Gorshkov K; Shiryaev SA; He S; Sun W; Cheng YS; Hu X; Tharappel AM; Lu B; Pinto A; Farhy C; Huang CT; Zhang Z; Zhu W; Wu Y; Zhou Y; Song G; Zhu H; Shamim K; Martinez-Romero C; Garcia-Sastre A; Preston RA; Jayaweera DT; Huang R; Huang W; Xia M; Simeonov A; Ming G; Qiu X; Terskikh AV; Tang H; Song H; Zheng W Emetine inhibits Zika and Ebola virus infections through two molecular mechanisms: inhibiting viral replication and decreasing viral entry. Cell Discov 2018, 4, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chen H; Lao Z; Xu J; Li Z; Long H; Li D; Lin L; Liu X; Yu L; Liu W; Li G; Wu J Antiviral activity of lycorine against Zika virus in vivo and in vitro. Virology 2020, 546, 88–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Deng YQ; Zhang NN; Li CF; Tian M; Hao JN; Xie XP; Shi PY; Qin CF Adenosine Analog NITD008 Is a Potent Inhibitor of Zika Virus. Open Forum Infect Dis 2016, 3, ofw175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moquin SA; Simon O; Karuna R; Lakshminarayana SB; Yokokawa F; Wang F; Saravanan C; Zhang J; Day CW; Chan K; Wang QY; Lu S; Dong H; Wan KF; Lim SP; Liu W; Seh CC; Chen YL; Xu H; Barkan DT; Kounde CS; Sim WLS; Wang G; Yeo HQ; Zou B; Chan WL; Ding M; Song JG; Li M; Osborne C; Blasco F; Sarko C; Beer D; Bonamy GMC; Sasseville VG; Shi PY; Diagana TT; Yeung BKS; Gu F NITD-688, a pan-serotype inhibitor of the dengue virus NS4B protein, shows favorable pharmacokinetics and efficacy in preclinical animal models. Sci Transl Med 2021, 13. [DOI] [PubMed] [Google Scholar]
  • 29.Nunes DAF; Santos F; da Fonseca STD; de Lima WG; Nizer W; Ferreira JMS; de Magalhaes JC NS2B-NS3 protease inhibitors as promising compounds in the development of antivirals against Zika virus: A systematic review. J Med Virol 2022, 94, 442–453. [DOI] [PubMed] [Google Scholar]
  • 30.Saiz JC; Martin-Acebes MA The Race To Find Antivirals for Zika Virus. Antimicrob Agents Chemother 2017, 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chung D; Schroeder CE; Sotsky J; Yao T; Roy S; Smith RA; Tower NA; Noah JW; McKellip S; Sosa M; Rasmussen L; White EL; Aube J; Golden JE ML336: Development of Quinazolinone-Based Inhibitors Against Venezuelan Equine Encephalitis Virus (VEEV). In Probe Reports from the NIH Molecular Libraries Program, Bethesda (MD), 2010. [PubMed] [Google Scholar]
  • 32.Lahon A; Arya RP; Kneubehl AR; Vogt MB; Dailey Garnes NJ; Rico-Hesse R Characterization of a Zika Virus Isolate from Colombia. PLoS Negl Trop Dis 2016, 10, e0005019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chan JF; Yip CC; Tsang JO; Tee KM; Cai JP; Chik KK; Zhu Z; Chan CC; Choi GK; Sridhar S; Zhang AJ; Lu G; Chiu K; Lo AC; Tsao SW; Kok KH; Jin DY; Chan KH; Yuen KY Differential cell line susceptibility to the emerging Zika virus: implications for disease pathogenesis, non-vector-borne human transmission and animal reservoirs. Emerg Microbes Infect 2016, 5, e93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yao Y; Huo T; Lin YL; Nie S; Wu F; Hua Y; Wu J; Kneubehl AR; Vogt MB; Rico-Hesse R; Song Y Discovery, X-ray Crystallography and Antiviral Activity of Allosteric Inhibitors of Flavivirus NS2B-NS3 Protease. J Am Chem Soc 2019, 141, 6832–6836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nie S; Yao Y; Wu F; Wu X; Zhao J; Hua Y; Wu J; Huo T; Lin YL; Kneubehl AR; Vogt MB; Ferreon J; Rico-Hesse R; Song Y Synthesis, Structure-Activity Relationships, and Antiviral Activity of Allosteric Inhibitors of Flavivirus NS2B-NS3 Protease. J Med Chem 2021, 64, 2777–2800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Nie S; Zhao J; Wu X; Yao Y; Wu F; Lin YL; Li X; Kneubehl AR; Vogt MB; Rico-Hesse R; Song Y Synthesis, structure-activity relationship and antiviral activity of indole-containing inhibitors of Flavivirus NS2B-NS3 protease. Eur J Med Chem 2021, 225, 113767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wu F; Zhou C; Yao Y; Wei L; Feng Z; Deng L; Song Y 3-(Piperidin-4-ylmethoxy)pyridine Containing Compounds Are Potent Inhibitors of Lysine Specific Demethylase 1. J Med Chem 2016, 59, 253–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wu F; Hua Y; Kaochar S; Nie S; Lin YL; Yao Y; Wu J; Wu X; Fu X; Schiff R; Davis CM; Robertson M; Ehli EA; Coarfa C; Mitsiades N; Song Y Discovery, Structure-Activity Relationship, and Biological Activity of Histone-Competitive Inhibitors of Histone Acetyltransferases P300/CBP. J Med Chem 2020, 63, 4716–4731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wu F; Nie S; Yao Y; Huo T; Li X; Wu X; Zhao J; Lin YL; Zhang Y; Mo Q; Song Y Small-molecule inhibitor of AF9/ENL-DOT1L/AF4/AFF4 interactions suppresses malignant gene expression and tumor growth. Theranostics 2021, 11, 8172–8184. [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.

Supplementary Materials

Supporting Information PDF

RESOURCES