Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 15.
Published in final edited form as: Bioorg Med Chem Lett. 2018 Nov 22;29(2):257–261. doi: 10.1016/j.bmcl.2018.11.044

Triazolopyrimidine and triazolopyridine scaffolds as TDP2 inhibitors

Carlos J A Ribeiro a,§, Jayakanth Kankanala a,§, Jiashu Xie a, Jessica Williams a, Hideki Aihara b, Zhengqiang Wang a,*
PMCID: PMC6339461  NIHMSID: NIHMS1005343  PMID: 30522956

Abstract

Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (TOP2) mediated DNA damages and causes cellular resistance to clinically used TOP2 poisons. Inhibiting TDP2 can potentially sensitize cancer cells toward TOP2 poisons. Commercial compound P10A10, to which the structure was assigned as 7-phenyl triazolopyrimidine analogue 6a, was previously identified as a TDP2 inhibitor hit in our virtual and fluorescence-based biochemical screening campaign. We report herein that the hit validation through resynthesis and structure elucidation revealed the correct structure of P10A10 (Chembridge ID 7236827) to be the 5-phenyl triazolopyrimidine regioisomer 7a. Subsequent structure-activity relationship (SAR) via the synthesis of a total of 47 analogues of both the 5-phenyl triazolopyrimidine scaffold (7) and its bioisosteric triazolopyridine scaffold (17) identified four derivatives (7a, 17a, 17e, and 17z) with significant TDP2 inhibition (IC50 < 50 μM), with 17z showing excellent cell permeability and no cytotoxicity.

Keywords: Tyrosyl-DNA phosphodiesterase 2 (TDP2), Anti-cancer, Triazolo-pirimidines, Triazolo-pyridines

Graphical Abstract

graphic file with name nihms-1005343-f0001.jpg


Topoisomerase II (TOP2) resolves DNA catenanes and knots and relax both positive and negative supercoils generated during normal physiological processes, such as transcription and replication.1 TOP2 manages these topological transactions via producing transient TOP2 cleavage complexes (TOP2cc) which feature a unique covalent bond between its active site tyrosine and the 5´-phosphate terminus of the DNA backbone at the break sites.1 In the presence of TOP2 poisons, such as anticancer drugs etoposide and doxorubicin, these transient TOP2cc can be stabilized and become abortive, resulting in DNA double-strand breaks (DSBs) and ultimately leading to cell death.23 Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs these DSBs by specifically cleaving the 5’-phosphotyrosine bond that links TOP2 to DNA to create competent DSB termini ready for ligation through the non-homologous end joining (NHEJ).47 By virtue of its cellular function TDP2 allows cancer cells to become resistant toward TOP2 poisons,6 a major class of anticancer drugs widely used in clinic. Evidence of TDP2 function in cancer cell damage repair also includes the observation that TDP2-deleted chicken lymphoma DT40 cells were hypersensitive to TOP2 poison etoposide, but not to the TOP1 poison camptothecin.8 Therefore, TDP2 represents an attractive anticancer drug target since its inhibition can sensitize cancer cells against TOP2 poisons. In addition, since treatment with TOP2 poisons is associated with non-specific toxicity due to limited tumor selectivity, co-administration with TDP2 inhibitors will potentially allow therapeutic efficacy with much lower doses of TOP2 poisons.5,9

Interestingly, TDP2 is also implicated in the genome repair of certain viruses. TDP2 was identified as the host VPg-unlinkase enzyme necessary to cleave VPg-RNA bonds during picornavirus replicative cycle.1012 In addition, TDP2 could also play a role in hepatitis B virus (HBV) cccDNA formation by facilitating the release of P-protein from viral DNA.13

Phosphodiesterase activity of TDP2 was only discovered in 2009,8 and since then only a small number of scaffolds have been reported with TDP2 inhibitory activity (Fig. 1).9, 1423 In addition, most of these scaffolds, such as compound 1 (NSC111041)23 are poorly characterized, moderately active and lacking drug-like properties. Deazaflavin (e.g. 2, Fig. 1),1921 is the only inhibitor type with nanomolar biochemical potency19 and well characterized mechanism of inhibition,21 yet these inhibitors did not exhibit strong efficacy in cells20 due to their poor cellular permeability.19 As part of our own efforts in developing drug like small molecule TDP2 inhibitors, we have previously identified an attractive inhibitor core isoquinoline-1,3-diones (e.g. 3).22

Figure 1.

Figure 1.

Representative reported TDP2 inhibitors

Recently we also developed the first fluorescence-based high throughput assay14 and identified new scaffolds with TDP2 inhibitory activity (e.g. compound 4, 5, and P10A10, Fig.1) during HTS of small library of 1600 compounds curated via virtual screening. In particular, [1,2,4]triazolo[1,5-a]pyrimidine compound P10A10 was found to inhibit 14M_zTDP2, a validated surrogate protein for hTDP2. Triazolopyrimidine is a highly privileged core in medicinal chemistry, with several biological activities reported just in the past three years: anti-Alzheimer’s disease,2425 antibacterial,2627 anticancer,2829 antileishmanial,30 antimalarial,3132 antiviral,3334 CB2 cannabinoid receptor inverse agonists,35 and phosphodiesterase 2 (PDE2a) inhibition.36 Similar core is also featured in various herbicidal compounds.37 In the present report, we describe our hit confirmation efforts including resynthesis and structural elucidation of hit P10A10, and the subsequent SAR of the triazolopyrimidine scaffold as TDP2 inhibitors.

The synthesis of triazolopyrimidines is well documented.3840 In our studies, [1,2,4]triazolo[1,5-a]pyrimidin-2-yl)benzamides 6 and 7 (scheme 1) were synthesized in two steps starting with phenylpropene-2-one (8) or phenylpropan-2-one (9) to obtain 7-phenyl isomer 6 or 5-phenyl isomer 7, respectively. In both cases, compounds 8 or 9 were condensed with diaminotriazole 10 in the presence of acetic acid under MW conditions to yield [1,2,4]triazolo[1,5-a]pyrimidin-2-amine intermediates 11 and 12, respectively. Acylation with different acid chlorides, followed by treatment with ammonia solution 7N in methanol, provided the desired final compounds in good yields (73 – 83%). [1,2,4]Triazolo[1,5-a]pyridin-2-amine scaffold 13a was constructed by reacting 2-amino-5-bromopyridine (14) with ethoxycarbonyl isothiocyanate in dichloromethane at room temperature to form thiourea 15, which was cyclized by reacting with hydroxylamine under basic conditions at reflux in ethanol/methanol. Intermediate 13 then underwent microwave-irradiated Suzuki coupling with different boronic acids in the presence of Pd(PPh3)4 to afford [1,2,4]triazolo[1,5-a]pyridin-2-amines 16 in moderate yields (46 – 81%). Compounds 17 and 18 were obtain by acylation of 16 in the same manner as described for compounds 6 and 7 (45–96% yield). Compound 19 was synthesized via reductive amination involving compound 16a and 3-chlorobenzaldehyde. Treating compound 16a with 3-chlorobenzenesulphonyl chloride in methanol at room temperature for 2 h afforded compound 20 in 53% yield.

Scheme 1.

Scheme 1.

Reagents and conditions: (a) AcOH, reflux, 4h, 72%; (b) (i) acid chloride (2.5 equiv), pyridine, 0°C → r.t., 1–2 h, (ii) 7N NH3 in MeOH, o.n., 45 – 96% (c) (i) AcOH, reflux, 6h; (ii) NBS, EtOH, reflux, 5h, 29% (d) ethoxycarbonylisothiocyanate (1 equiv), dioxane, under argon, 0°C to r.t., 12 h; (e) NH2OH·HCl (5 equiv), DIPEA (3 equiv), EtOH/MeOH 1:1, 2 h r.t. then 60°C, 3 h, 65% (over 2 steps); (f) boronic acid, Pd(PPh3)4, K2CO3, EtOH, H2O (1:1), M.W.,150°C, 30 min, 46 – 81% (g) 3 chlorobenzaldehyde (1.1 equiv), acetic acid (cat.), MeOH, r.t., 2 h; then sodium cyanoborohydride (2.0 equiv), r.t., 2 h, 76 %; (h) 3-chlorobenzenesulfonyl chloride (2.5 equiv), pyridine, 0°C → r.t., 2 h, 53%.

Our hit validation started with resynthesizing hit compound P10A10 (6a)41 and closely related analogues 6b-c to confirm the inhibitory capability of this scaffold (Scheme 1, Table 1). Surprisingly, all three analogues were found to be completely inactive at 100 μM, indicating that the structural assignment of compound P10A10 as 6a could be questionable. In further structural analysis, although mass spectrometry showed the same mass for P10A10 and 6a, from 1H-NMR spectra the proton chemical shifts, and specially the coupling constant, for the pyrimidine hydrogens differed significantly (J= 7.1 versus J= 4.7 Hz, respectively, Figure 2) between P10A10 and 6a. In addition, literature search revealed that reacting aminoazoles with α,β-unsaturated compounds can generate different triazolopyrimidine regioisomers depending on the reaction conditions employed.4243 These observations suggest that either our synthesized compound 6a or the commercial P10A10 could be the 5-phenyl regioisomer 7a. To further elucidate the structure, we obtained the crystal structure for an advanced intermediate 11 (step a, Scheme 1), which confirmed that the phenyl ring is indeed on the 7 position (Figure 2) as reported in literature for the same reaction.44 In the meantime, we synthesized compound 7a, the 5-phenyl isomer of compound 6a. Comparison of 7a to commercial compound P10A10 showed identical 1H-NMR spectrum and same potency inhibiting TDP2. With all these, we corrected the structure for P10A10 (Chembridge ID 7236827) to be 7a, not 6a as assigned by the vendor.

Table 1.

TDP2 inhibitory activities of derivatives 6a-c, 7 a-f, 17a-o and 18.

graphic file with name nihms-1005343-f0002.jpg

Compds R1 IC50 (μM)a
6a 3-NO2 >100
6b H >100
6c 4-NO2 >100
7a 3-NO2 22.0 ± 2.5
7b H >100
7c 4-NO2 >100
7d 3-CN >100
7e 3-CF3 >100
7f 3-Br 80.9 ± 4.0
17a 3-NO2Ph 16.6 ± 0.4
17b 3-OCH3Ph >100
17c 3-CH3Ph >100
17d 3-FPh >100
17e 3-ClPh 43.8 ± 3.9
17f pyridin-3-yl >100
17g pyridin-4-yl >100
17h 4-OCH3Ph >100
17i 4-N(CH3)2 >100
17j 3,4-FPh >100
17k furan-2-yl >100
17l 4-ClPh >100
17m 2-ClPh >100
17n 3,5-ClPh >100
17o 2-Cl, 4-FPh >100
18 - 77.3 ± 7.5
a

IC50 values are the mean of three independent experiments performed in triplicate.

Figure 2.

Figure 2.

Comparison of pyrimidine hydrogens NMR coupling constant from both isomers 6a and 7a, and X-ray crystallography studies of intermediate 11 (CCDC 1869530) reveals that P10A10 is not isomer 6a, but 7a instead.

All 47 final compounds were evaluated in a fluorescence-based biochemical assay14 measuring the catalytic activity of TDP2. In addition, permeability of selected derivatives was assessed by parallel artificial membrane permeability assay (PAMPA) and cytotoxicity was determined for the two overall best derivatives.

With the confirmation of 7a as a TDP2 inhibitor hit, we started exploring the SAR to see if we could improve the hit. We were particularly interested in replacing the nitro group as nitroarenes are associated with potential toxicity in medicinal chemistry with negative impact to the pharmacokinetic and physicochemical profile.4546 To probe the importance of meta-nitro group of the benzamide ring to activity, we first synthesized analogues without nitro (7b), with nitro at para position (7c), or with nitro replaced with another electron withdrawing group, such as nitrile (7d), trifluoromethyl, (7e) and bromo (7f). As shown in Table 1, these substitutions had a dramatic impact on TDP2 inhibition as significant activity was not observed with compounds 7b-e whereas compound 7f exhibited only weak inhibitory activity (IC50= 80.9 μM). One of the challenges we had technically with these compounds was their poor solubility in DMSO. In an attempt to improve solubility and to further probe the SAR, we decided to synthesize the 4-deaza bioisosteric core, the triazolopyridine (17). Interestingly, compound 17a which bears the same meta-nitrophenyl moiety as hit 7a was found to be slightly more active (IC50= 16.6 μM vs. 22.0 μM) with a better solubility profile in both DMSO and buffer. Therefore, we decided to continue our study focusing on the triazolopyridine scaffold 17 instead. Successful nitro bioisosters47 described in literature include nitrile,4850 pyridine,5152 and halogenated analogues.5354 In addition, nitroarenes can behave as halo- or alkylarenes when noncovalently interacting with proteins.46 Unfortunately, all these bioisosteric replacements led to less active compounds (17b-o), with only 3-chloro derivative (17e) exhibiting an IC50 lower than 50 μM. Moreover, changing chloro to para and ortho positions (17l-m) or adding a second chloro (17o) further reduced activity.

Another SAR trend was that the phenyl group at different positions in the main core favored position 7 (17a, IC50= 16.6 μM vs. 18, IC50= 77.3 μM vs. 6a, IC50> 100 μM). Although the potency of compound 17e was found to be 2-fold lower than the hit compound (IC50= 43.8 μM vs 22.0 μM), its permeability in PAMPA assay was 6-fold higher (Table 3), and therefore we decided to maintain a 3-chlorobenzamide moiety when probing substitutions at position 7 of the pyridine ring (17p-ac, Table 2). Only 17s with 4-methoxyphenyl group was able to retain almost the same activity (IC50= 59.0 μM), with pyridine-3-yl substitution leading to a two-fold improve of potency (17z, IC50= 21.0 μM vs. 17e, 43.8 μM) and 7-fold increase in permeability (Table 3). The increase in potency observed for derivative 17z led us to further test substitution at meta position with potential H bond acceptor properties (3-methylsulfonyl, 17ad; 3-acetyl, 17ae; 3-carboxylate 17af) and also additional derivatives with aromatic nitrogens (pyridine-4-yl, 17ag; quinoline-3-yl, 17ah; pyrimidin-5-yl, 17ai). However, all modifications led to a decrease in potency.

Table 3.

TDP2 inhibition, permeability and cytotoxicity of the four best derivatives

Compds TDP2 IC50 (μM)a Pe (10−6 cm/s)b HepG2 CC50 (μM)c HeLa CC50 (μM)c
7a 22.0 ± 2.5 0.16 -- --
17a 16.6 ± 0.4 0.15 -- --
17e 43.8 ± 3.9 0.9 >100 >100
17z 21.0 ± 2.1 6.3 >100 >100
1 0.040 0.095 -- --
a

IC50 values are the mean of three independent experiments performed in triplicate;

b

permeability is considered high when Pe > 1.5;

c

compounds were tested at 50 μM and 100 μM (two independent experiments were performed in triplicate).

Table 2.

TDP2 inhibitory activities of derivatives 17p-ai, 19–20.

graphic file with name nihms-1005343-f0003.jpg

Compds R1 IC50 (μM)a
17p 4-ClPh 80.8 ± 7.5
17q 4-OHPh >100
17r 4-CF3Ph >100
17s 4-OCH3Ph 59.0 ± 3.7
17t 4-FPh 85.2 ± 4.6
17u 3-ClPh >100
17v 3-OCH3Ph >100
17w 3-CF3Ph >100
17x 2-OCH3Ph >100
17y 2-ClPh >100
17z pyridin-3-yl 21.0 ± 2.1
17aa furan-2-yl 94.5 ± 6.8
17ab thiophen-2-yl >100
17ac furan-3-yl >100
17ad 3-SO2CH3Ph >100
17ae 3-COCH3Ph >100
17af 3-COOHPh >100
17ag pyridin-4-yl >100
17ah quinolin-3-yl >100
17ai pyrimidin-5-yl >100
Compds X IC50 (μM)
19 CH2 >100
20 SO2 >100
a

IC50 values are the mean of three independent experiments performed in triplicate.

Lastly, we synthesized compounds in which the carboxamide moiety of 17 is replaced by an amine (19) and or a sulfonamide (20). Both substitutions led to inactive derivatives, highlighting the importance of the amide group to activity.

Since the goal of developing TDP2 inhibitors is to sensitize cancer cells toward TOP2 poisons to ultimately overcome drug resistance and potentially also allow TOP2 therapeutic efficiency at lower and less toxic concentrations, it is imperative that these inhibitors are cell permeable and non-cytotoxic. Toward these ends, selected active compounds (7a, 17a, 17e, and 17z) were first tested in the PAMPA permeability assay along with the most potent deazaflavin inhibitor 1 (Table 3). From this assay, excellent permeability was observed only with our compound 17z. Another analogue 17e also exhibited borderline Pe value which is about 10 times higher than that of 1. These findings are consistent with the report that deazaflavin compounds lacked cellular permeability. We then assessed the two cell-permeable compounds 17e and 17z for cytotoxicity in two different cell lines (HepG2 and HeLa), from which no cytotoxicity was observed at concentrations up to 100 μM (Table 3). These results indicate that despite the relatively moderate TDP2 inhibitory activity, scaffold 17 has the potential to be developed into cell permeable and noncytotoxic chemical probes for studying cellular functions.

From a previous screening campaign against TDP2, compound P10A10, reported as 7-phenyl triazolopyrimidine 6a, was identified as a hit. The present study on hit validation via resynthesis and structure elucidation revealed that the correct structure for P10A10 (Chembridge ID 7236827) should be 5-phenyl isomer 7a. To probe TDP2 inhibition potentialities of this scaffold we synthesized a total of 47 compounds: 11 derivatives inhibited TDP2 (IC50 < 100 μM), with four derivatives (7a, 17a, 17e, and 17z) reaching potencies below 50 μM. Although we were unable to increase significantly the potency of the original hit, compounds 17e and 17z, particularly the latter, demonstrated good cellular permeability and no cytotoxicity and can be further developed into useful chemical probes for studying cellular functions of TDP2 in host DNA repair as well as virus genome repair.

Supplementary Material

Suppl

Acknowledgments

This research was supported by the Academic Health Center Faculty Research Development Grant Program (FRD #14.23), University of Minnesota, and partially by the Center for Drug Design, University of Minnesota, and NIH grant GM118047 to HA. We acknowledge Professor Bert Semler at University of California, Irvine and Professor Haitao Guo at Indiana University School of Medicine, for providing HeLa and HepG2 Cells, respectively. We also acknowledge Victor G. Young, Jr. and the X-Ray Crystallographic Laboratory for the X-ray crystallography studies. The Bruker-AXS D8 Venture diffractometer was purchased through a grant from NSF/MRI (#1224900) and the University of Minnesota.

Footnotes

Appendix A. Supplementary data

Experimental procedures, NMR characterization, as well as crystal data and structure refinement for compound 11, can be found in the supporting information. Supplementary data associated with this article can be found online at

References

  • 1.Pommier Y; Sun Y; Huang SN; Nitiss JL, Roles of eukaryotic topoisomerases in transcription, replication and genomic stability. Nat. Rev. Mol. Cell Biol 2016, 17 (11), 703–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pommier Y, Drugging topoisomerases: lessons and challenges. ACS Chem. Biol 2013, 8 (1), 82–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Fortune JM; Osheroff N, Topoisomerase II as a target for anticancer drugs: when enzymes stop being nice. Prog. Nucleic Acid Res. Mol. Biol 2000, 64, 221–53. [DOI] [PubMed] [Google Scholar]
  • 4.Menon V; Povirk LF, End-processing nucleases and phosphodiesterases: An elite supporting cast for the non-homologous end joining pathway of DNA double-strand break repair. DNA Repair (Amst) 2016, 43, 57–68. [DOI] [PubMed] [Google Scholar]
  • 5.Pommier Y; Huang S.-y. N.; Gao R; Das BB; Murai J; Marchand C, Tyrosyl-DNA-phosphodiesterases (TDP1 and TDP2). DNA Repair 2014, 19, 114–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Do PM; Varanasi L; Fan S; Li C; Kubacka I; Newman V; Chauhan K; Daniels SR; Boccetta M; Garrett MR; Li R; Martinez LA, Mutant p53 cooperates with ETS2 to promote etoposide resistance. Genes Dev 2012, 26 (8), 830–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zeng Z; Cortés-Ledesma F; El Khamisy SF; Caldecott KW, TDP2/TTRAP is the major 5′-tyrosyl DNA phosphodiesterase activity in vertebrate cells and is critical for cellular resistance to topoisomerase II-induced DNA damage. J. Biol. Chem 2011, 286 (1), 403–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cortes Ledesma F; El Khamisy SF; Zuma MC; Osborn K; Caldecott KW, A human 5’-tyrosyl DNA phosphodiesterase that repairs topoisomerase-mediated DNA damage. Nature 2009, 461 (7264), 674–8. [DOI] [PubMed] [Google Scholar]
  • 9.Laev SS; Salakhutdinov NF; Lavrik OI, Tyrosyl-DNA phosphodiesterase inhibitors: Progress and potential. Bioorg. Med. Chem 2016, 24 (21), 5017–5027. [DOI] [PubMed] [Google Scholar]
  • 10.Virgen-Slane R; Rozovics JM; Fitzgerald KD; Ngo T; Chou W; van Noort GJV; Filippov DV; Gershon PD; Semler BL, An RNA virus hijacks an incognito function of a DNA repair enzyme. Proc. Natl. Acad. Sci. U. S. A 2012, 109 (36), 14634–14639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Maciejewski S; Nguyen JHC; Gomez-Herreros F; Cortes-Ledesma F; Caldecott KW; Semler BL, Divergent requirement for a DNA repair enzyme during enterovirus infections. Mbio 2016, 7 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Maciejewski S; Ullmer W; Semler BL, VPg unlinkase/TDP2 in cardiovirus infected cells: Re-localization and proteolytic cleavage. Virology 2018, 516, 139–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Koniger C; Wingert I; Marsmann M; Rosler C; Beck J; Nassal M, Involvement of the host DNA-repair enzyme TDP2 in formation of the covalently closed circular DNA persistence reservoir of hepatitis B viruses. Proc. Natl. Acad. Sci. U. S. A 2014, 111 (40), E4244–E4253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ribeiro CJA; Kankanala J; Shi K; Kurahashi K; Kiselev E; Ravji A; Pommier Y; Aihara H; Wang Z, New fluorescence-based high-throughput screening assay for small molecule inhibitors of tyrosyl-DNA phosphodiesterase 2 (TDP2). Eur. J. Pharm. Sci 2018, 118, 67–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Thomson G; Watson A; Caldecott K; Denneny O; Depledge P; Hamilton N; Hopkins G; Jordan A; Morrow C; Raoof A; Waddell I; Ogilvie D, Generation of assays and antibodies to facilitate the study of human 5′-tyrosyl DNA phosphodiesterase. Anal. Biochem 2013, 436 (2), 145–150. [DOI] [PubMed] [Google Scholar]
  • 16.Kossmann BR; Abdelmalak M; Lopez S; Tender G; Yan C; Pommier Y; Marchand C; Ivanov I, Discovery of selective inhibitors of tyrosyl-DNA phosphodiesterase 2 by targeting the enzyme DNA-binding cleft. Bioorg. Med. Chem. Lett 2016, 26 (14), 3232–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Beck DE; Lv W; Abdelmalak M; Plescia CB; Agama K; Marchand C; Pommier Y; Cushman M, Synthesis and biological evaluation of new fluorinated and chlorinated indenoisoquinoline topoisomerase I poisons. Bioorg. Med. Chem 2016, 24 (7), 1469–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wang P; Elsayed MSA; Plescia CB; Ravji A; Redon CE; Kiselev E; Marchand C; Zeleznik O; Agama K; Pommier Y; Cushman M, Synthesis and biological evaluation of the first triple inhibitors of human topoisomerase 1, tyrosyl–DNA phosphodiesterase 1 (tdp1), and tyrosyl–DNA phosphodiesterase 2 (tdp2). J. Med. Chem 2017, 60 (8), 3275–3288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Raoof A; Depledge P; Hamilton NM; Hamilton NS; Hitchin JR; Hopkins GV; Jordan AM; Maguire LA; McGonagle AE; Mould DP; Rushbrooke M; Small HF; Smith KM; Thomson GJ; Turlais F; Waddell ID; Waszkowycz B; Watson AJ; Ogilvie DJ, Toxoflavins and deazaflavins as the first reported selective small molecule inhibitors of tyrosyl-DNA phosphodiesterase II. J. Med. Chem 2013, 56 (16), 6352–6370. [DOI] [PubMed] [Google Scholar]
  • 20.Marchand C; Abdelmalak M; Kankanala J; Huang SY; Kiselev E; Fesen K; Kurahashi K; Sasanuma H; Takeda S; Aihara H; Wang Z; Pommier Y, Deazaflavin inhibitors of tyrosyl-DNA phosphodiesterase 2 (TDP2) specific for the human enzyme and active against cellular TDP2. ACS Chem. Biol 2016, 11 (7), 1925–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hornyak P; Askwith T; Walker S; Komulainen E; Paradowski M; Pennicott LE; Bartlett EJ; Brissett NC; Raoof A; Watson M; Jordan AM; Ogilvie DJ; Ward SE; Atack JR; Pearl LH; Caldecott KW; Oliver AW, Mode of action of DNA-competitive small molecule inhibitors of tyrosyl DNA phosphodiesterase 2. Biochem. J 2016, 473, 1869–1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kankanala J; Marchand C; Abdelmalak M; Aihara H; Pommier Y; Wang Z, Isoquinoline-1,3-diones as selective inhibitors of tyrosyl DNA phosphodiesterase II (TDP2). J. Med. Chem 2016, 59 (6), 2734–2746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kont YS; Dutta A; Mallisetty A; Mathew J; Minas T; Kraus C; Dhopeshwarkar P; Kallakury B; Mitra S; Uren A; Adhikari S, Depletion of tyrosyl DNA phosphodiesterase 2 activity enhances etoposide-mediated double-strand break formation and cell killing. DNA Repair 2016, 43, 38–47. [DOI] [PubMed] [Google Scholar]
  • 24.Kumar J; Meena P; Singh A; Jameel E; Maqbool M; Mobashir M; Shandilya A; Tiwari M; Hoda N; Jayaram B, Synthesis and screening of triazolopyrimidine scaffold as multi-functional agents for Alzheimer’s disease therapies. Eur. J. Med. Chem 2016, 119, 260–277. [DOI] [PubMed] [Google Scholar]
  • 25.Kovalevich J; Cornec AS; Yao Y; James M; Crowe A; Lee VMY; Trojanowski JQ; Smith AB; Ballatore C; Brunden KR, Characterization of Brain-Penetrant Pyrimidine-Containing Molecules with Differential Microtubule-Stabilizing Activities Developed as Potential Therapeutic Agents for Alzheimer’s Disease and Related Tauopathies. J. Pharmacol. Exp. Ther 2016, 357 (2), 432–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jung IP; Ha NR; Lee SC; Ryoo SW; Yoon MY, Development of potent chemical antituberculosis agents targeting Mycobacterium tuberculosis acetohydroxyacid synthase. Int. J. Antimicrob. Agents 2016, 48 (3), 247–258. [DOI] [PubMed] [Google Scholar]
  • 27.Wang H; Lee M; Peng Z; Blázquez B; Lastochkin E; Kumarasiri M; Bouley R; Chang M; Mobashery S, Synthesis and Evaluation of 1,2,4-Triazolo[1,5-a]pyrimidines as Antibacterial Agents Against Enterococcus faecium. J. Med. Chem 2015, 58 (10), 4194–4203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wang S; Zhao L-J; Zheng Y-C; Shen D-D; Miao E-F; Qiao X-P; Zhao L-J; Liu Y; Huang R; Yu B; Liu H-M, Design, synthesis and biological evaluation of [1,2,4]triazolo[1,5-a]pyrimidines as potent lysine specific demethylase 1 (LSD1/KDM1A) inhibitors. Eur. J. Med. Chem 2017, 125, 940–951. [DOI] [PubMed] [Google Scholar]
  • 29.Arenas-Gonzalez A; Mendez-Delgado LA; Merino-Montiel P; Padron JM; Montiel-Smith S; Vega-Baez JL; Meza-Reyes S, Synthesis of monomeric and dimeric steroids containing 1,2,4 triazolo 1,5-a pyrimidines. Steroids 2016, 116, 13–19. [DOI] [PubMed] [Google Scholar]
  • 30.da Silva ER; Boechat N; Pinheiro LCS; Bastos MM; Costa CCP; Bartholomeu JC; da Costa TH, Novel Selective Inhibitor of Leishmania (Leishmania) amazonensis Arginase. Chem. Biol. Drug Des 2015, 86 (5), 969–978. [DOI] [PubMed] [Google Scholar]
  • 31.Phillips MA; White KL; Kokkonda S; Deng X; White J; El Mazouni F; Marsh K; Tomchick DR; Manjalanagara K; Rudra KR; Wirjanata G; Noviyanti R; Price RN; Marfurt J; Shackleford DM; Chiu FCK; Campbell M; Jimenez-Diaz MB; Bazaga SF; Angulo-Barturen I; Martinez MS; Lafuente-Monasterio M; Kaminsky W; Silue K; Zeeman A-M; Kocken C; Leroy D; Blasco B; Rossignol E; Rueckle T; Matthews D; Burrows JN; Waterson D; Palmer MJ; Rathod PK; Charman SA, A Triazolopyrimidine-Based Dihydroorotate Dehydrogenase Inhibitor with Improved Drug-like Properties for Treatment and Prevention of Malaria. ACS Infectious Diseases 2016, 2 (12), 945–957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kokkonda S; Deng XY; White KL; Coteron JM; Marco M; de las Heras L; White J; El Mazouni F; Tomchick DR; Manjalanagara K; Rudra KR; Chen G; Morizzi J; Ryan E; Kaminsky W; Leroy D; Martinez-Martinez MS; Jimenez-Diaz MB; Bazaga SF; Angulo-Barturen I; Waterson D; Burrows JN; Matthews D; Charman SA; Phillips MA; Rathod PK, Tetrahydro-2-naphthyl and 2-Indanyl Triazolopyrimidines Targeting Plasmodium falciparum Dihydroorotate Dehydrogenase Display Potent and Selective Antimalarial Activity. J. Med. Chem 2016, 59 (11), 5416–5431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Massari S; Nannetti G; Desantis J; Muratore G; Sabatini S; Manfroni G; Mercorelli B; Cecchetti V; Palu G; Cruciani G; Loregian A; Goracci L; Tabarrini O, A Broad Anti-influenza Hybrid Small Molecule That Potently Disrupts the Interaction of Polymerase Acidic Protein-Basic Protein 1 (PA-PB1) Subunits. J. Med. Chem 2015, 58 (9), 3830–42. [DOI] [PubMed] [Google Scholar]
  • 34.Huang B; Li C; Chen W; Liu T; Yu M; Fu L; Sun Y; Liu H; De Clercq E; Pannecouque C; Balzarini J; Zhan P; Liu X, Fused heterocycles bearing bridgehead nitrogen as potent HIV-1 NNRTIs. Part 3: Optimization of [1,2,4]triazolo[1,5-a]pyrimidine core via structure-based and physicochemical property-driven approaches. Eur. J. Med. Chem 2015, 92, 754–765. [DOI] [PubMed] [Google Scholar]
  • 35.Aghazadeh Tabrizi M; Baraldi PG; Ruggiero E; Saponaro G; Baraldi S; Poli G; Tuccinardi T; Ravani A; Vincenzi F; Borea PA; Varani K, Synthesis and structure activity relationship investigation of triazolo[1,5-a]pyrimidines as CB2 cannabinoid receptor inverse agonists. Eur. J. Med. Chem 2016, 113, 11–27. [DOI] [PubMed] [Google Scholar]
  • 36.Gomez L; Massari ME; Vickers T; Freestone G; Vernier W; Ly K; Xu R; McCarrick M; Marrone T; Metz M; Yan YG; Yoder ZW; Lemus R; Broadbent NJ; Barido R; Warren N; Schmelzer K; Neul D; Lee D; Andersen CB; Sebring K; Aertgeerts K; Zhou X; Tabatabaei A; Peters M; Breitenbucher JG, Design and Synthesis of Novel and Selective Phosphodiesterase 2 (PDE2a) Inhibitors for the Treatment of Memory Disorders. J. Med. Chem 2017, 60 (5), 2037–2051. [DOI] [PubMed] [Google Scholar]
  • 37.Billington R; Gehen SC; Hanley TR, Chapter 87 - Toxicology of Triazolopyrimidine Herbicides In Hayes’ Handbook of Pesticide Toxicology (Third Edition), Krieger R, Ed. Academic Press: New York, 2010; pp 1865–1885. [Google Scholar]
  • 38.Elashry ESH; Rashed N, 1,2,4-Triazolo- and Tetrazolo[x,y-z]pyrimidines In Adv. Heterocycl. Chem, Katritzky AR, Ed. Academic Press: 1998; Vol. 72, pp 127–224. [Google Scholar]
  • 39.Fischer G, Recent Progress in 1,2,4-Triazolo[1,5-a]pyrimidine Chemistry In Adv. Heterocycl. Chem, Katritzky AR, Ed. Academic Press: 2007; Vol. 95, pp 143–219. [Google Scholar]
  • 40.Fizer M; Slivka M, Synthesis of 1,2,4 triazolo 1,5-a pyrimidine (microreview). Chem. Heterocycl. Compds 2016, 52 (3), 155–157. [Google Scholar]
  • 41.https://www.molinstincts.com/chemical-structure/Cambridge-id-7236827-cstr-CT1067003447.html (accessed 9/19/2018).
  • 42.Allen CFH; Beilfuss HR; Burness DM; Reynolds GA; Tinker JF; VanAllan JA, The Structure of Certain Polyazaindenes. IV. Compounds from β-Keto Acetals and β-Methoxyvinyl Ketones1. J. Org. Chem 1959, 24 (6), 796–801. [Google Scholar]
  • 43.Massari S; Desantis J; Nannetti G; Sabatini S; Tortorella S; Goracci L; Cecchetti V; Loregian A; Tabarrini O, Efficient and regioselective one-step synthesis of 7-aryl-5-methyl- and 5-aryl-7-methyl-2-amino-[1,2,4]triazolo[1,5-a]pyrimidine derivatives. Org. Biomol. Chem 2017, 15 (37), 7944–7955. [DOI] [PubMed] [Google Scholar]
  • 44.Menet CJ; Fletcher SR; Van Lommen G; Geney R; Blanc J; Smits K; Jouannigot N; Deprez P; van der Aar EM; Clement-Lacroix P; Lepescheux L; Galien R; Vayssiere B; Nelles L; Christophe T; Brys R; Uhring M; Ciesielski F; Van Rompaey L, Triazolopyridines as selective JAK1 inhibitors: from hit identification to GLPG0634. J. Med. Chem 2014, 57 (22), 9323–42. [DOI] [PubMed] [Google Scholar]
  • 45.Boelsterli UA; Ho HK; Zhou S; Leow KY, Bioactivation and hepatotoxicity of nitroaromatic drugs. Curr. Drug Metab 2006, 7 (7), 715–27. [DOI] [PubMed] [Google Scholar]
  • 46.Dörwald FZ, Lead Optimization for Medicinal Chemists: Pharmacokinetic Properties of Functional Groups and Organic Compounds Wiley-VCH: Weinheim, Germany: 2012. [Google Scholar]
  • 47.Meanwell NA, Synopsis of Some Recent Tactical Application of Bioisosteres in Drug Design. J. Med. Chem 2011, 54 (8), 2529–2591. [DOI] [PubMed] [Google Scholar]
  • 48.Abeywardane A; Caviness G; Choi Y; Cogan D; Gao A; Goldberg D; Heim-Riether A; Jeanfavre D; Klein E; Kowalski JA; Mao W; Miller C; Moss N; Ramsden P; Raymond E; Skow D; Smith-Keenan L; Snow RJ; Wu F; Wu J-P; Yu Y, N-Arylsulfonyl-α-amino carboxamides are potent and selective inhibitors of the chemokine receptor CCR10 that show efficacy in the murine DNFB model of contact hypersensitivity. Bioorg. Med. Chem. Lett 2016, 26 (21), 5277–5283. [DOI] [PubMed] [Google Scholar]
  • 49.Tang H; Walsh SP; Yan Y; de Jesus RK; Shahripour A; Teumelsan N; Zhu Y; Ha S; Owens KA; Thomas-Fowlkes BS; Felix JP; Liu J; Kohler M; Priest BT; Bailey T; Brochu R; Alonso-Galicia M; Kaczorowski GJ; Roy S; Yang L; Mills SG; Garcia ML; Pasternak A, Discovery of Selective Small Molecule ROMK Inhibitors as Potential New Mechanism Diuretics. ACS Med. Chem. Lett 2012, 3 (5), 367–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cottyn B; Acher F; Ramassamy B; Alvey L; Lepoivre M; Frapart Y; Stuehr D; Mansuy D; Boucher J-L; Vichard D, Inhibitory effects of a series of 7-substituted-indazoles toward nitric oxide synthases: Particular potency of 1H-indazole-7-carbonitrile. Bioorg. Med. Chem 2008, 16 (11), 5962–5973. [DOI] [PubMed] [Google Scholar]
  • 51.Gneuwuch CT; Friedman HL, Pyridine isosteres of the .beta.-adrenergic antagonists, 2-(p-nitrophenyl)-1-isopropylamino-2-ethanol and 3-(p-nitrophenoxy)-1-isopropylamino-2-propanol. J. Med. Chem 1972, 15 (12), 1321–1324. [DOI] [PubMed] [Google Scholar]
  • 52.Julémont F; de Leval X; Michaux C; Damas J; Charlier C; Durant F; Pirotte B; Dogné J-M, Spectral and Crystallographic Study of Pyridinic Analogues of Nimesulide: Determination of the Active Form of Methanesulfonamides as COX-2 Selective Inhibitors. J. Med. Chem 2002, 45 (23), 5182–5185. [DOI] [PubMed] [Google Scholar]
  • 53.Yu L-F; Li Y-Y; Su M-B; Zhang M; Zhang W; Zhang L-N; Pang T; Zhang R-T; Liu B; Li J-Y; Li J; Nan F-J, Development of Novel Alkene Oxindole Derivatives As Orally Efficacious AMP-Activated Protein Kinase Activators. ACS Med. Chem. Lett 2013, 4 (5), 475–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nagarathnam D; Miao SW; Lagu B; Chiu G; Fang J; Murali Dhar TG; Zhang J; Tyagarajan S; Marzabadi MR; Zhang F; Wong WC; Sun W; Tian D; Wetzel JM; Forray C; Chang RSL; Broten TP; Ransom RW; Schorn TW; Chen TB; O’Malley S; Kling P; Schneck K; Bendesky R; Harrell CM; Vyas KP; Gluchowski C, Design and Synthesis of Novel α1a Adrenoceptor-Selective Antagonists. 1. Structure−Activity Relationship in Dihydropyrimidinones. J. Med. Chem 1999, 42 (23), 4764–4777. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Suppl

RESOURCES