ABSTRACT
Antimicrobial resistance has made a sizeable impact on public health and continues to threaten the effectiveness of antibacterial therapies. Novel bacterial topoisomerase inhibitors (NBTIs) are a promising class of antibacterial agents with a unique binding mode and distinct pharmacology that enables them to evade existing resistance mechanisms. The clinical development of NBTIs has been plagued by several issues, including cardiovascular safety. Herein, we report a sub-series of tricyclic NBTIs bearing an amide linkage that displays promising antibacterial activity, potent dual-target inhibition of DNA gyrase and topoisomerase IV (TopoIV), as well as improved cardiovascular safety and metabolic profiles. These amide NBTIs induced both single- and double-strand breaks in pBR322 DNA mediated by Staphylococcus aureus DNA gyrase, in contrast to prototypical NBTIs that cause only single-strand breaks. Unexpectedly, amides 1a and 1b targeted human topoisomerase IIα (TOP2α) causing both single- and double-strand breaks in pBR322 DNA, and induced DNA strand breaks in intact human leukemia K562 cells. In addition, anticancer drug-resistant K/VP.5 cells containing decreased levels of TOP2α were cross-resistant to amides 1a and 1b. Together, these results demonstrate broad spectrum antibacterial properties of selected tricyclic NBTIs, desirable safety profiles, an unusual ability to induce DNA double-stranded breaks, and activity against human TOP2α. Future work will be directed toward optimization and development of tricyclic NBTIs with potent and selective activity against bacteria. Finally, the current results may provide an additional avenue for development of selective anticancer agents.
KEYWORDS: multidrug resistance, novel bacterial topoisomerase inhibitors, anticancer drugs
INTRODUCTION
Bacterial infections result in extensive mortality. In 2019, there were an estimated 7.7 million deaths globally from the 33 most significant bacterial pathogens (1). Additionally, the rise in antibiotic resistance threatens the effectiveness of current treatment options, resulting in 1.3 million directly attributable deaths in 2019 (2). Within the United States, the Centers for Disease Control and Prevention has prioritized antibiotic-resistant bacteria based on their impact on public health. Examples include the serious threats from methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant (MDR) Pseudomonas aeruginosa and the urgent threats from carbapenem-resistant Enterobacterales and Acinetobacter baumannii (3). Therefore, the development of novel therapies to address these and other bacterial pathogens is warranted.
Novel bacterial topoisomerase inhibitors (NBTIs) (4) target the bacterial type II topoisomerases DNA gyrase and topoisomerase IV (TopoIV), enzymes responsible for modulating the topology of DNA during replication, transcription, and chromosomal dysjunction during binary fission. DNA gyrase uniquely introduces negative supercoils into relaxed DNA and also functions ahead of the replication fork to remove positive supercoiling, whereas TopoIV serves to decatenate DNA (5). These enzymes carry out their respective functions via transient double-strand DNA cleavage reactions followed by religation (5, 6). Previous research has demonstrated that NBTIs primarily stabilize single-strand DNA breaks (SSBs) generated by DNA gyrase and TopoIV (7 – 9). The unique binding mode (7, 8) and distinct pharmacological properties of NBTIs make them an advantageous choice for targeting MDR pathogens.
The fluoroquinolone (FQ) class of antibacterial agents also targets DNA gyrase and TopoIV. In contrast to most NBTIs, FQs stabilize double-strand DNA breaks (DSBs), preventing religation of DNA and resulting in bacterial cell death (9 – 12). However, resistance conferred by mutations in the target enzymes has eroded the effectiveness of FQs (9 – 12). NBTIs, with their distinctive structures and mode of action, largely evade cross-resistance with FQs (7, 8), making them a promising class of antibacterial agents for clinical development. Indeed, gepotidacin is currently in Phase III clinical trials for the treatment of uncomplicated urinary tract infections and urogenital gonorrhea. Gepotidacin has proven efficacious in patients against many pathogenic bacteria including Escherichia coli, MRSA, and Neisseria gonorrhoeae (13 – 15), laying a powerful foundation for the development of future NBTIs.
The advancement of additional NBTI clinical candidates has been hindered by several issues, especially cardiovascular toxicity driven by inhibition of the human ether-à-go-go-related gene (hERG) K+ ion channel (16). Gepotidacin, by contrast, is a weak hERG inhibitor (IC50 = 588 µg/mL) with a low risk of proarrhythmia at clinically relevant dosing/exposures (17). However, gepotidacin and other NBTIs typically target DNA gyrase more potently than TopoIV in S. aureus and have additional liabilities associated with emergence of resistance via mutations to gyrase (14, 18 – 20). We have sought structurally distinctive NBTIs with potent anti-MRSA activity [minimal inhibitory concentrations (MICs) of ≤0.25 µg/mL], potent inhibition of both supercoiling by S. aureus DNA gyrase, and decatenation by TopoIV, in vitro cardiac safety (hERG IC50 ≥ 100 µM), and selectivity for targeting bacterial vs human type II topoisomerases. Owing to the need for new therapies directed at life-threatening Gram-negative bacteria such as Enterobacteriaceae (including E. coli) and A. baumannii, we also routinely test these NBTIs against representative Gram-negative pathogens.
We previously reported a series of novel bicyclic 5-amino-1,3-dioxane-linked inhibitors with potent antibacterial activity against Gram-positive pathogens and improved cardiovascular safety profiles (20). This series, however, lacked potent Gram-negative activity and generally displayed rapid metabolism in microsomal assays. Optimization efforts of this series included increasing rigidity and polarity, resulting in analogs with tricyclic DNA-binding moieties (21), e.g., amines 2a and 2b (Fig. 1). Selected azatricyclic and diazatricyclic amine NBTIs displayed potent anti-MRSA activity, reduced hERG inhibition, and improved metabolic stability compared to the bicyclic inhibitors (20, 21).
Fig 1.
Design rationale for scaffold modification of tricyclic analogs.
Herein, we describe a subseries of tricyclic NBTIs bearing an amide linkage to the enzyme-binding moiety. This strategy further increases rigidity with the removal of a rotatable bond and replaces the weakly basic secondary amine with a non-basic amide. The potent antibacterial activity of these amides against both Gram-positive and Gram-negative pathogens prompted us to conduct biochemical and whole-cell experiments. These studies revealed an atypical mechanism of action vs DNA gyrase and unexpected targeting of human topoisomerase IIα (TOP2α).
RESULTS
Tricyclic amide NBTIs possess in vitro antibacterial activity
Antibacterial activity was measured as MICs against both Gram-positive and Gram-negative pathogens as previously described (20, 21). The azatricyclic amide-linked pyridooxazinone 1a was the most potent compound against a fluoroquinolone-resistant MRSA strain, with an MIC of 0.125 µg/mL (Table 1); slightly more potent than the clinical candidate gepotidacin. Amide 1a also exhibited promising Gram-negative activity against A. baumannii and E. coli, with an MIC of 2 µg/mL; the activity against A. baumannii was comparable to that of ciprofloxacin. We previously reported (21) that 2a, the amine counterpart to amide 1a, also had potent anti-MRSA activity, with an MIC of 0.25 µg/mL. In contrast, MICs for 2a were eightfold higher than 1a against the two Gram-negative bacteria (Table 1). The diazatricyclic amide-linked pyridooxazinone 1b was substantially weaker than 1a, with an eightfold loss in activity against MRSA, 16-fold loss against A. baumannii, and fourfold loss against E. coli (Table 1). These results suggest that the presence of the additional nitrogen in the diazatricyclic scaffold of 1b led to reduced activity. Amide 1b and its amine counterpart, 2b, displayed similar antibacterial activity, although 1b was fourfold more potent against E. coli (Table 1).
TABLE 1.
Minimal inhibitory concentrations (µg/mL) a of NBTIs
Tricyclic amide NBTI cardiovascular safety and metabolic profiles
Cardiovascular safety is a common concern for the development of NBTIs (16), and we have prioritized compounds with hERG IC50 values ≥100 µM. We previously demonstrated that reducing the lipophilicity of dioxane-linked amine analogs (targeting cLogP < 1.5) generally resulted in low hERG inhibition (20). Notably, the pyridooxazinone enzyme-binding moiety common to 1a,b and 2a,b routinely afforded more potent hERG inhibition, even for highly polar compounds. Amides 1a and 1b replace the weakly basic amine, a strategy our laboratories (22) and others (23, 24) have previously successfully employed to reduce hERG inhibition. hERG inhibition was measured using an automated electrophysiology assay (IonWorks Barracuda system), as previously reported in reference (25) and shown in Table 2. Amides 1a and 1b both displayed favorable hERG IC50 values, meeting our goal criteria of ≥100 µM. This finding is especially notable given the somewhat higher lipophilicity of 1a (cLogP = 1.82). Notably, both amides 1a and 1b were superior to their previously reported matched pairs, amines 2a and 2b, respectively, supporting the hERG design strategy.
TABLE 2.
In vitro hERG IC50 and metabolic stability
| Compound | hERG IC50 (µM) a | Half-life (min) b | cLogP |
|---|---|---|---|
| 1a | >100 | 28 | 1.82 |
| 1b | >100 | 41 | 0.95 |
| 2a c | 30 | 18.4 | 1.32 |
| 2b c | 97 | 66.3 | 0.43 |
Charles River (Cleveland, OH, USA).
Charles River (Worcester, MA, USA).
Values for amine analogs were previously reported (21).
Metabolic clearance is also strongly correlated with lipophilicity (26), and dioxane-linked amine NBTIs generally required cLogP < 1.5 to achieve a half-life >30 min in mouse microsomes (20). The incorporation of the amide moiety blocks a potential site of metabolism (20), and azatricyclic amide 1a was less rapidly metabolized than amine 2a (Table 2). The diazatricyclic series showed the opposite trend; amide 1b had a shorter half-life than amine 2b. Thus, the incorporation of the amide linkage did not consistently improve the half-life of this subseries of compounds. The more polar diazatricyclic compounds 1b and 2b were more stable than their azatricyclic counterparts 1a and 2a, suggesting that reducing lipophilicity continues to be a productive strategy to enhance metabolic stability.
NBTIs inhibit supercoiling by DNA gyrase and decatenation by TopoIV
Isolated S. aureus DNA gyrase and TopoIV were used to assess inhibition of supercoiling and decatenation, respectively, by tricyclic NBTIs. IC50 values are shown in Table 3. Amide 1a was the most potent inhibitor of supercoiling and decatenation, with IC50 values of 150 nM and 653 nM, respectively. Notably, 1a was equipotent to gepotidacin against DNA gyrase and fourfold more potent against TopoIV. Amide 1a was also 10-fold more potent than previously reported amine 2a against DNA gyrase and twofold more potent against TopoIV. A similar trend was seen with the diazatricyclic compounds. Amide 1b was 18-fold more active than amine 2b against DNA gyrase and twofold improved for TopoIV. The azatricyclic amide 1a was superior to diazatricyclic 1b (Table 3), as we had previously reported for amines 2a and 2b (21). Taken together, these data support an important role for the amide moiety in disrupting enzyme function and suggest that lipophilicity may also contribute to activity.
TABLE 3.
Target inhibition by tricyclic NBTIs
| Compound | S. aureus DNA gyrase IC50 (nM) a | S. aureus TopoIV IC50 (nM) b |
|---|---|---|
| 1a | 150 | 653 |
| 1b | 229 | 755 |
| 2a c | 1,600 | 1,300 (n = 5) |
| 2b c | 4,200 | 1,400 |
| Gepotidacin (racemic) c | 170 | 2,800 |
| Ciprofloxacin c | 28,000 | 8,600 |
DNA gyrase supercoiling inhibition assay; n = 2.
TopoIV decatenation inhibition assay; n = 2 unless otherwise noted in parentheses.
Values for amine analogs, gepotidacin, and ciprofloxacin were previously reported (21).
Tricyclic NBTI effects on DNA cleavage induced by S. aureus DNA gyrase
NBTIs primarily enhance gyrase-mediated SSBs, as has been well characterized with gepotidacin (8). In contrast, ciprofloxacin, a fluoroquinolone, primarily enhances gyrase-mediated DSBs (10, 11). We previously reported that bicyclic dioxane-linked amide NBTIs atypically stabilize both SSBs and DSBs (22). Therefore, we next evaluated the tricyclic amide 1a for its activity in cleavage assays using isolated S. aureus DNA gyrase. Incubation with 1a resulted in a concentration-dependent increase in both SSBs and DSBs at nanomolar concentrations (Fig. 2A), whereas the gepotidacin control at a concentration of 20 µM showed only SSBs. Interestingly, SSBs and DSBs induced by amide 1a were nearly equivalent (Fig. 2B and C). In contrast, amine 2a displayed the prototypical behavior for NBTIs, inducing a concentration-dependent increase in SSBs with no appreciable DSBs (Fig. 3). These results suggest that the amide motif plays a critical role in accumulation of DSBs, consistent with results from pyridooxazinone and pyridothiazinone amides in our bicyclic series (22).
Fig 2.
Compound 1a induction of DNA strand breaks in the presence of gyrase. (A) The ethidium-stained gel indicates positions of DNA after incubation with enzyme in the absence or presence of compound 1a (0.2–50 nM), a dioxane-linked azatricyclic NBTI containing an amide-linked enzyme-binding moiety. The various DNA forms are indicated as negatively supercoiled pBR322 DNA {[-(SC)]}, linearized DNA (Lin); nicked open-circular DNA (Nick). Ciprofloxacin and gepotidacin were included as controls at the indicated concentrations. (B) Quantitation of compound 1a induced DNA single-strand breaks and double-strand breaks was accomplished by first measuring fluorescence in each lane with corrections for differential fluorescent emission efficiency in (-) SC compared to linearized (Lin) and nicked (Nick) DNA bands (27) followed by the calculation of percent total fluorescence in the respective bands. Percent cleavage in enzyme controls was subtracted to yield final results. (C) Percent DNA cleavage induced by compound 1a (50 nM). Results shown are the mean ± standard deviation (SD) from six separate experiments performed on separate days.
Fig 3.
Compound 2a induction of DNA strand breaks in the presence of gyrase. (A) The ethidium-stained gel indicates positions of DNA after incubation with enzyme in the absence or presence of compound 2a (0.01–10 µM), a dioxane-linked azatricyclic NBTI containing an amine-linked enzyme-binding moiety. Ciprofloxacin and gepotidacin were included as controls at the indicated concentrations. (B) Quantitation of compound 2a induced DNA single-strand breaks and double-strand breaks was obtained by the same method as that explained in Fig. 2B. (C) Percent DNA cleavage induced by compound 2a (10 µM). Results shown are the mean ± SD from five separate experiments performed on separate days.
Growth inhibitory effects of tricyclic NBTIs in human leukemia cells
Given the ubiquity and essentiality of type II topoisomerases across kingdoms, achieving selective targeting and activity against bacterial compared to human type II topoisomerases (e.g., TOP2α) is an important consideration for antibacterial development. We have routinely assessed NBTI compounds for potential TOP2α targeting by measuring growth inhibition in a human chronic myelogenous leukemia cell line (K562) and in an acquired anticancer drug (etoposide)-resistant clonal subline (K/VP.5) (20 – 22, 25). TOP2α levels are reduced in K/VP.5 cells to 15% of that found in parental K562 cells (28). Thus, selective NBTIs would be expected to exhibit little or no difference in growth inhibitory activity in K562 compared to K/VP.5 cells as an indication that these compounds lack targeting against the human enzyme, as we have found most often (20 – 22, 25).
Forty-eight-hour growth inhibition experiments were conducted as previously reported (29). The anticancer agent etoposide yielded expected results with 26-fold resistance in K/VP.5 cells, coordinate with a reduction in the level of TOP2α (Fig. 4A; Table 4). Surprisingly, 1a inhibited the growth of K562 cells with an IC50 value of 0.44 µM (Table 4). Importantly, 10-fold cross-resistance was observed in K/VP.5 cells (Fig. 4B; Table 4), yielding an IC50 value of 4.57 µM (Table 4) and suggesting that the amide 1a targets TOP2α. In stark contrast, amine 2a exhibited much less potent growth inhibition in K562 with no difference in activity in K/VP.5 cells (Fig. 4C; Table 4), an indication that this amine-containing NBTI did not target human TOP2α. Diazatricyclic amide 1b and amine 2b exhibited a similar trend (Table 4), indicating a critical role for the amide moiety in targeting TOP2α.
Fig 4.
Growth inhibitory effects of etoposide and NBTIs in K562 and K/VP.5 cells. Log-phase cells were incubated for 48 h with various concentrations of (A) etoposide, (B) 1a, (C) 2a after which cells were counted on a model Z1 Coulter counter (Beckman Coulter, Danvers, MA, USA). The extent of growth beyond the starting concentration in drug treated vs dimethyl sulfoxide (DMSO) controls was expressed ultimately as percent inhibition. The concentration-response curves for each of the tested drugs are shown as scattergrams from multiple experiments. The 50% inhibitory concentrations (IC50 values) are shown in Table 4, averaging results from multiple experiments performed on separate days.
TABLE 4.
Growth inhibitory activity of tricyclic NBTIs in K562 and K/VP.5 cells
| Compound | K562, IC50 (µM) b | K/VP.5, IC50 (µM) | Fold resistance c |
|---|---|---|---|
| Etoposide | 0.10 ± 0.02 d | 2.56 ± 1.50 | 25.6 |
| 1a | 0.44 ± 0.17 (6) d | 4.57 ± 1.66 (6) | 10.4 |
| 1b | 2.26 ± 0.79 (3) | 22.11 ± 11.02 (3) | 9.8 |
| 2a a | 100.66 ± 0.25 | 127.44 ± 25.02 | 1.26 |
| 2b a | >200 | >200 | NA |
Values for amine analogs were previously reported (21).
IC50 in a 48-h growth inhibition assay.
IC50 of K/VP.5 divided by that of the parental K562 cell line.
Mean ± range or SD; number of independent experiments performed on different days; n = 2 unless otherwise noted in parentheses.
Tricyclic amide NBTI-induced DNA damage in human leukemia cells
The results from growth inhibition experiments prompted us to investigate the action of amide 1a in intact mammalian cells. We conducted single-cell gel electrophoresis (Comet assay) (30) experiments to assess DNA damage in K562 and K/VP.5 cells (29). K562 cells were incubated for 30 or 60 min with DMSO vehicle control and 10–50 µM 1a followed by quantitation of DNA damage (Fig. 5A) (29). Additionally, amine 2a (50 µM) was used as a comparator, given its low growth inhibitory activity in K562 cells and lack of cross-resistance in K/VP.5 cells (Fig. 4C; Table 4). A concentration-dependent increase in DNA damage was observed with 1a with no appreciable DNA damage seen with 2a compared to DMSO control (Fig. 5A). Compounds 1a and 2a were compared across multiple experiments after 30-min incubation at 50 µM validating the activity of 1a and the relative lack of activity of 2a (Fig. 5B). The direct comparison between 1a and 2a further highlights the distinct effects of the amide in causing the accumulation of DNA strand breaks. No further increase in 1a-induced DNA damage was observed after 60 min (Fig. 5B). DNA damage induced by 1a was dramatically attenuated in K/VP.5 compared to K562 cells consistent with the diminished cellular level of TOP2α in these anticancer drug-resistant cells (Fig. 5C). In addition, 2a induced little or no DNA damage in either cell line (Fig. 5C). Together, results are consistent with the amide 1a targeting human TOP2α.
Fig 5.
NBTI induction of DNA strand breaks in human leukemia K562 cells. (A) Effects of 1a and 2a on DNA damage in K562 cells. K562 cells were incubated with 1a (10, 25, and 50 µM), 2a (50 µM), or DMSO (control) for 30 min followed by alkaline (pH 13) Comet assays. Results shown are from all data points (cells/nuclei) within a representative experiment. (B) Effects of 1a and 2a (50 µM) on DNA damage (Comet assays) in K562 cells incubated for 30 min and 1a (50 µM) for 60 min followed by alkaline (pH 13) Comet assays. Results shown are the mean ± SD from 4–5 experiments run on separate days. (C) K562 and K/VP.5 cells were incubated for 30 min at 37°C with DMSO (control) and 1a (50 µM) or 2a (50 µM) followed by evaluation of DNA damage by Comet assays. For all experimental conditions, more than 100 cells were evaluated by OpenComet software (31). *P < 0.001 compared to respective DMSO controls.
Tricyclic NBTI effects on DNA cleavage induced by human TOP2α
To provide additional evidence for human TOP2α targeting, DNA cleavage experiments using isolated human TOP2α enzyme were conducted. Enzyme alone controls revealed conversion of negatively supercoiled pBR322 substrate to relaxed DNA forms (RLX) when ethidium bromide was included in the agarose gels for electrophoresis as well as some SSBs (Nick) (Fig. 6A) (27, 32). Incubation with the anticancer agent etoposide (100 µM), a positive control, resulted in an accumulation of SSBs (Nick) and DSBs (Lin) (Fig. 6A). Compound 1a resulted in a concentration-dependent increase in both SSBs and DSBs, though SSBs predominated (Fig. 6A). Results from replicate experiments using 50 µM 1a are shown in Fig. 6B. In contrast, amine 2a did not induce DNA cleavage compared to enzyme controls (Fig. 6C). These results further confirm targeting of TOP2α by amide 1a.
Fig 6.
Compounds 1a, 2a induction of DNA strand breaks in the presence of TOP2α. (A) The ethidium-stained gel shows positions of DNA after incubation with enzyme in the absence or presence of 1a (0.1–100 µM) or etoposide (100 µM). The forms of DNA are indicated as relaxed (RLX), negatively supercoiled [(-) SC], nicked open-circular (Nick), and linearized (Lin). Formation of SSBs (Nick) and DSBs (Lin) was observed by fluorescence band intensity during conversion of (-) SC to Nick and Lin reaction products. EcoRI digested pBR322 was run on one lane as a control for identification of linearized DNA and for quantitation of %DNA cleavage. (B) Percent DNA cleavage (SSB and DSB) induced by compound 1a (50 µM) was quantified relative to intensity of the EcoRI band after subtraction of enzyme alone SSB or DSB controls. Results shown are the mean ± SD from six separate experiments. (C) The ethidium-stained gel shows positions of DNA forms after incubation with enzyme in the absence or presence of 2a (0.1–100 µM) or etoposide (100 µM).
DISCUSSION
The development of NBTIs for the treatment of MDR bacterial infections is challenging. Optimization efforts require balancing multiple parameters including antibacterial activity, inhibition of DNA gyrase and TopoIV, cardiovascular safety, and metabolic stability. The previously described amine 2a demonstrated potent antibacterial activity against Gram-positive pathogens but suffered from poor Gram-negative activity, relatively potent hERG inhibition, and poor metabolic stability (21). Here, we hypothesized that replacing the amine of 2a with an amide in 1a would reduce hERG inhibition (21, 24) and improve metabolic stability by blocking the potentially oxidatively labile amino methylene with a carbonyl group (20). We postulated that increased rigidity through the removal of one rotatable bond could improve downstream pharmacokinetic properties such as oral bioavailability (33) and enhance accumulation in Gram-negative bacteria (34). Indeed, amide 1a demonstrated several notable advantages over amine 2a, including a greater than threefold increase in hERG IC50 and modest improvement in microsomal metabolic stability (Table 2). We were particularly encouraged by the substantial improvement in Gram-negative antibacterial activity for 1a (Table 1). The MIC of 2 µg/mL against E. coli was eightfold better than for 2a and on par with NBTI 5463, an earlier NBTI optimized for Gram-negative activity (35). Similar improvements were observed for A. baumannii. However, not all of these findings were generalizable. Whereas amide 1b also demonstrated reduced hERG inhibition vs amine 2b, improvements in metabolic stability and Gram-negative activity were not observed. Whereas amides 1a,1b both targeted S. aureus DNA gyrase and TopoIV more potently than amines 2a,2b, the increased inhibition in these biochemical assays did not translate to marked improvements in S. aureus MICs. This observation may reflect reduced penetration into S. aureus cells by the amides, but we have not yet carried out such measurements.
Amide 1a also exhibited intriguing pharmacological properties demonstrating the formation of DNA DSBs with S. aureus DNA gyrase, in contrast to the exclusive SSBs traditionally observed with NBTIs (7 – 9). We have previously reported DSBs with other amide NBTIs bearing oxazinone and thiazinone amide moieties, whereas benzamide NBTIs yielded only SSBs (22). The basis of this differentiation is not currently understood, although structural biology efforts are underway in an attempt to elucidate mechanism(s). Such research has provided powerful insight into the mechanism of other classes of inhibitors, including the very recent cryoelectron microscopy studies with the albicidins and E. coli DNA gyrase (36). Whether other amide-type NBTIs also induce type II topoisomerase-mediated DSBs is an open question; DNA cleavage results for such compounds have not been reported (24, 35).
Additionally, the tricyclic amide 1a but not the amine 2a inhibited the growth of human leukemia K562 cells in the low micromolar range (Table 4). The observed cross-resistance (Fig. 4B) in an acquired anticancer drug-resistant clonal cell line (K/VP.5), containing reduced TOP2α, suggests that this tricyclic amide NBTI has the capability to target TOP2α. A limitation of the present work is that both amides 1a,1b and amines 2a,2b were assayed as racemic mixtures. It is not certain whether the TOP2α-targeting activity resides in one or both enantiomers. Amide NBTIs, in general, appear to lack potent targeting of TOP2α (22, 24); we have only rarely observed such cross-resistance in our previous research, for example, with compound 4e in reference (21).
Assessment of amide 1a-induced DNA damage in anticancer drug-resistant K/VP.5 cells compared to K562 cells (Fig. 5C) is another indication that 1a is capable of targeting TOP2α. No appreciable DNA damage was manifest with 2a in intact cells, in accord with a virtual lack of growth inhibition in intact cells (Fig. 5A through C). In addition, biochemical analysis of the effects of 1a and 2a using isolated TOP2α enzyme provided evidence that amide 1a stabilized SSBs and DSBs generated by TOP2α (Fig. 6A and B), whereas amine 2a had no apparent DNA damaging effects (<1% due to nicking) on the human enzyme (Fig. 6C).
In summary, azatricyclic amide 1a delivered several key advances relative to our earlier NBTIs, most notably its low hERG inhibition, favorable dual-target inhibition of DNA gyrase and TopoIV, and potent activity against Gram-negative pathogens. The unexpected targeting of TOP2α precludes its further development as an antibacterial candidate. This discovery may, however, pave the way for the identification of potential anticancer therapeutics based on this chemically attractive scaffold with good drug-like properties. There have been previous reports of TOP2-targeting anticancer agents derived from classical antibacterial scaffolds, for example, the quinolone voreloxin (37). Finally, future work will be directed toward optimization and development of tricyclic NBTIs with both potent and selective activity against bacteria.
MATERIALS AND METHODS
Synthesis
General amide formation method: 3-oxo-3,4-dihydro-2H-pyrido3,2-b][1,4]oxazine-6-carboxylic acid (1–1.5 equiv), hydroxybenzotriazole (HOBT, 1.1 equiv), the requisite primary amine (1 equiv), 4-dimethylaminopyridine (DMAP, 0.1 equiv), and N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide (EDC, 1.1 equiv) were dissolved in tetrahydrofuran (THF, 0.1 M), and triethylamine (4 equiv) was added. The resulting mixture was heated at reflux overnight, then cooled to room temperature, diluted with dichloromethane, washed with saturated NaHCO3 solution, dried over Na2SO4, and concentrated in vacuo. The residue was purified by chromatography on silica gel with dichloromethane/methanol (0%–25% gradient) to give the desired compound. Purity was determined by high-performance liquid chromatography (HPLC, “Method 2”) as previously reported (21).
N-(2-((9-fluoro-4-oxo-1,2-dihydro-4H-pyrrolo[3,2,1-ij]quinolin-1-yl)methyl)- trans -1,3-dioxan-5-yl)-3-oxo-3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine-6-carboxamide (1a)
The title compound as a white solid (0.033 g, 0.069 mmol, 57%) was prepared following the general amide formation method with the previously described (21) primary amine (1-((5-amino-trans-1,3-dioxan-2-yl)methyl)-9-fluoro-1,2-dihydro-4H-pyrrolo[3,2,1-ij]quinolin-4-one): 1H NMR (400 MHz, DMSO-d 6) δ: 11.31 (s, 1H), 7.93–7.88 (m, 2H), 7.63–7.58 (m, 2H), 7.47 (d, J = 8.2 Hz, 1H), 7.02 (dd, J = 9.8, 8.8 Hz, 1H), 6.51 (d, J = 9.4 Hz, 1H), 4.77 (t, J = 4.7 Hz, 1H), 4.74 (s, 2H), 4.46 (dd, J = 12.6, 9.5 Hz, 1H), 4.21–4.00 (m, 5H), 3.69–3.58 (m, 2H), 2.24 (dt, J = 13.8, 4.2 Hz, 1H), 2.02 (ddd, J = 14.0, 10.3, 4.6 Hz, 1H). 13C NMR (100 MHz, DMSO-d 6) δ: 165.31, 163.47, 159.70, 158.73 (d, J = 248.4 Hz), 144.21 (d, J = 12.1 Hz), 141.83, 141.04, 140.41, 137.13, 127.11 (d, J = 9.3 Hz), 123.47, 122.10 (d, J = 2.6 Hz), 118.20, 118.00 (d, J = 20.6 Hz), 113.87 (d, J = 2.0 Hz), 111.41 (d, J = 23.5 Hz), 99.52, 68.41, 68.38, 66.76, 53.69, 42.24, 37.80, 33.79 (d, J = 1.2 Hz). HRMS (ESI) m/z calc’d for C24H22FN4O6 [M + H]+: 481.1523; found: 481.1529. HPLC: ELSD rt: 14.871 min, purity: 100%.
N-(2-((3-fluoro-7-oxo-4,5-dihydro-7H-pyrrolo[3,2,1-de][1,5]naphthyridin-4-yl)methyl)-1,3- trans -dioxan-5-yl)-3-oxo-3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine-6-carboxamide (1b)
The title compound as a white solid (0.036 g, 0.075 mmol, 50%) was prepared following the general amide formation method with the previously described (21) primary amine (4-((5-amino-1,3-trans-dioxan-2-yl)methyl)-3-fluoro-4,5-dihydro-7H-pyrrolo[3,2,1-de][1,5]naphthyridin-7-one). 1H NMR (400 MHz, DMSO-d 6) δ: 11.30 (s, 1 h), 8.44 (d, J = 1.4 Hz, 1 h), 7.97 (d, J = 9.7 Hz, 1 h), 7.91 (d, J = 8.0 Hz, 1 h), 7.60 (d, J = 8.1 Hz, 1 h), 7.47 (d, J = 8.2 Hz, 1 h), 6.76 (d, J = 9.7 Hz, 1 h), 4.78 (t, J = 4.7 Hz, 1 h), 4.74 (s, 2 h), 4.49 (dd, J = 12.0, 9.0 Hz, 1 h), 4.22–4.07 (m, 5 h), 3.69–3.59 (m, 2 h), 2.24 (dt, J = 14.0, 4.1 Hz, 1 h), 2.12 (ddd, J = 13.8, 10.0, 4.8 Hz, 1 h). 13C NMR (100 MHz, DMSO-d 6) δ: 165.34, 163.49, 159.30, 155.21 (d, J = 258.3 Hz), 141.86, 141.05, 140.50 (d, J = 8.9 Hz), 140.43, 137.94, 135.22 (d, J = 26.2 Hz), 131.42 (d, J = 2.6 Hz), 126.94 (d, J = 17.7 Hz), 125.89 (d, J = 2.5 Hz), 123.50, 118.23, 99.25, 68.40, 68.37, 66.77, 53.95, 42.22, 36.88, 34.59. HRMS (ESI) m/z calc’d for C23H21FN5O6 [M + H]+: 482.1476; found: 482.1482. HPLC: ELSD rt: 13.177 min, purity: 99.4%.
The preparation of amines 2a and 2b has been previously described (21).
Minimal inhibitory concentration assay
Minimal inhibitory concentrations were assayed in triplicate (at a minimum) by microbroth dilution according to CLSI guidelines (38).
Microsomal stability
The stability in mouse microsomes was determined at Charles River (Worcester, MA, USA) using the previously described methodology (20).
hERG IC50
IC50 values were measured at Charles River (Cleveland, OH, USA) using the IonWorks Barracuda system (Molecular Devices Corporation, Union City, CA, USA) as previously reported (25).
Staphylococcus aureus gyrase supercoiling assay
Assays were performed using an Inspiralis Ltd (Norwich, UK) kit with modifications. DNA gyrase was added to initiate reactions and was present at a level that was just able to supercoil all relaxed pBR322 under the experimental conditions utilized. DNA gyrase was incubated with 250 ng of relaxed pBR322 DNA, DMSO vehicle control (1.2%–2.7% final), and varying concentrations of drugs/compounds diluted in DMSO (1.2%–2.7% final) in 30 µL total reaction volume at 37°C for 30 min under the following conditions: 40 mM HEPES-KOH (pH 7.6), 10 mM magnesium acetate, 10 mM DTT, 2 mM ATP, 500 mM potassium glutamate and 0.05 mg/mL BSA. Each reaction was stopped with 30 µL STEB buffer [40% sucrose, wt/vol; 100 mM Tris-HCl (pH 8); 10 mM EDTA (pH 8); 0.5 mg/mL bromophenol blue] followed by the addition of 30 µL chloroform/iso-amyl alcohol (24:1) and loaded on a 1% agarose gel run at 70 V for 2 h. Gels were stained with ethidium bromide (1 µg/mL) followed by UV visualization.
Staphylococcus aureus TopoIV decatenation assay
Assays were performed using an Inspiralis Ltd (Norwich, UK) kit according to protocol with slight modifications. Decatenation of 100 ng of kDNA was assessed after 20 min incubation at 37°C in a final 30 µL reaction volume with DMSO vehicle control (1.7% final) and varying concentrations of drugs/compounds diluted in DMSO in a buffer containing 50 mM Tris-HCl (pH 7.5), 5 mM magnesium chloride, 5 mM dithiothreitol (DTT), 1.5 mM ATP, 350 mM potassium glutamate, 0.05 mg/mL bovine serum albumin (BSA). TopoIV was added to initiate reactions and was present at a level that was just able to decatenate 80%–100% of the kDNA under the experimental conditions utilized. Reactions were stopped with 30 µL STEB buffer [40% sucrose, wt/vol; 100 mM Tris-HCl (pH 8); 10 mM EDTA (pH 8); 0.5 mg/mL bromophenol blue] followed by the addition of 30 µL chloroform/iso-amyl alcohol (24:1). After mixing and centrifugation, aqueous fractions (20 µL) were loaded on a 1% agarose gel and run at 70 V for 2 h. Gels were stained with ethidium bromide (1 µg/mL) for UV visualization and quantitation of the percent decatenation in the presence or absence of various concentrations of test compounds for the assessment of 50% inhibitory concentrations.
Staphylococcus aureus DNA gyrase cleavage assay
Assays were performed using modifications of a kit from Inspiralis Ltd (Norwich, UK). Reactions in 30 µL total volume were incubated with varying concentrations of drug/compound diluted in DMSO (3% final DMSO concentrations) at 37°C for 30 min in assay buffer [40 mM HEPES-KOH (pH 7.6), 10 mM magnesium acetate, 10 mM DTT, 100 mM potassium glutamate and 0.05 mg/mL albumin] containing pBR322 (250 ng/condition) and DNA gyrase (15 nM). Complexes were trapped with the addition of 2% (wt/vol) SDS (3 µL) and proteinase K (10 mg/mL) (1.5 µL) and incubated at 45°C for an additional 30 min to digest enzyme and reveal single-strand (Nick) and double-strand (Lin) breaks. STEB buffer (30 µL) [40% (wt/vol) sucrose, 100 mM Tris-HCl (pH 8), and 10 mM EDTA, 0.5 mg/mL bromophenol blue] and chloroform:isoamyl alcohol (24:1) (30 µL) were added followed by vigorous mixing and centrifugation for 2 min. The aqueous phase (20 µL) was loaded onto a 1% agarose gel containing 0.7 µg/mL ethidium bromide. Gel electrophoresis was conducted (60 V, 2 h) in 1× Tris acetate EDTA (TAE) buffer containing 0.7 µg/mL ethidium bromide. DNA bands were visualized using ImageLab software (Bio-Rad Laboratories, Hercules, CA, USA). Total fluorescence in each lane was determined as previously reported in reference (22) using corrections for differential emission in supercoiled DNA compared to linearized and nicked DNA bands according to reference (27) followed by calculation of percent total fluorescence in these bands as a measure of DNA cleavage. Percent cleavage in enzyme alone controls was subtracted to yield final results plotted using Sigmaplot 14.5 (Systat Software, Inc., San Jose, CA, USA).
Human topoisomerase II DNA cleavage assay
The formation of nicked (Nick; SSBs), linear (Lin; DSBs), and relaxed (RLX) DNA from negatively supercoiled pBR322 (substrate) was detected by separating reaction products using ethidium bromide gel electrophoresis as previously described (27, 32). The cleavage assay reaction mixture (20 µL total) contained 300 ng of isolated TOP2α protein, 160 ng of pBR322 DNA (NEB, Ipswich, MA, USA), and 1 mM ATP in an assay buffer [10 mM (pH 7.5), 50 mM KCL, 50 mM NaCl, 0.1 mM EDTA, 5 mM MgCl2, 2.5% glycerol] along with DMSO solvent and 0.1–100 µM compound 1a or 2a in DMSO. The anticancer agent etoposide was also evaluated at the indicated concentrations. Assay buffer (17 µL) and compound/DMSO (1 µL) were mixed at room temperature for 30 min followed by addition of TOP2α (2 µL) to initiate the reaction which proceeded at 37°C for 30 min. Reactions were quenched with 10% SDS (2 µL) and 5 min later with 10 mM EDTA-200 mM NaCl mixture (2 µL) followed by 2 µL proteinase K (10 mg/mL). Tubes were incubated at 55°C for 1 h followed by addition of pre-heated 10× DNA loading solution (3.5 µL) (glycerol, H2O, and bromophenol blue) and run on a 1.3% agarose gel containing ethidium bromide (0.7 µg/mL) in 1× TAE buffer also containing ethidium bromide (0.7 µg/mL) at 20V for 18 h. EcoRI digested pBR322 (160 ng) was run on one lane as a control for identification of linearized DNA and or quantitation of %DNA cleavage.
Mammalian cell growth inhibition
Growth inhibition was conducted in a human myelogenous leukemia cell line (K562) and an etoposide-resistant clonal subline (K/VP.5) using methods previously reported (29). Varying concentrations of drugs were incubated (48 h) in a 24-well plate containing log-phase parental K562 and clonal drug-resistant K/VP.5 cells adjusted to 1–1.5 × 106 cells. Cells were counted using a Z1 Coulter counter. Growth inhibition (IC50 value) was determined based on comparison with DMSO control growth using non-linear regression plots.
DNA damage (Comet) assay
Alkaline single-cell gel electrophoresis was utilized to assess DNA damage induced by compounds 1a and 2a as previously reported (29). K562 cells were washed and resuspended (1 × 106 cell/mL) in warm buffer (25 mM HEPES, 10 mM glucose, 1 mM MgCl2, 5 mM KCl, 130 mM NaCl, 5 mM NaH2PO4, pH 7.4) followed by incubation with 10, 25 or 50 µM of 1a and 50 µM of 2a or DMSO (solvent control) for 30 or 60 min at 37°C. The treated cells were washed with ice-cold buffer (1× PBS) and resuspended to 0.28 × 106 cell/mL, and then further diluted in low melt agarose. Following alkaline electrophoresis (of ~2,000 cells) and staining with fluorescent DNA intercalating dye (SYBR Gold), DNA damage was visualized and quantified by fluorescence microscopy. Migrating fragments (comet tails) from the nucleoids (comet heads) were visualized and used to quantify the Olive tail moment (30) using the ImageJ processing program with the open-source software tool OpenComet (31). Olive tail moments from more than 100 cells per sample condition were determined (29).
ACKNOWLEDGMENTS
This project was supported by funding from the Dr. Ralph and Marian Falk Medical Research Trust (Transformational Award to M.J.M.-F.) and by the National Institutes of Health (R21 AI148986 and R01 AI173072 to M.J.M.-F.). C.A.M. was partially supported by the Jack L. Beal Fellowship, and Y.L. was partially supported by the Chih-Ming and Jane Chen Graduate Fellowship Fund in Medicinal Chemistry and Pharmacognosy.
Contributor Information
Jack C. Yalowich, Email: yalowich.1@osu.edu.
Mark J. Mitton-Fry, Email: mitton-fry.1@osu.edu.
Anne-Catrin Uhlemann, Columbia University Irving Medical Center, New York, New York, USA .
REFERENCES
- 1. Ikuta KS, Swetschinski LR, Robles Aguilar G, Sharara F, Mestrovic T, Gray AP, Davis Weaver N, Wool EE, Han C, Gershberg Hayoon A, Aali A, Abate SM, Abbasi-Kangevari M, Abbasi-Kangevari Z, Abd-Elsalam S, Abebe G, Abedi A, Abhari AP, Abidi H, Aboagye RG, Absalan A, Abubaker Ali H, Acuna JM, Adane TD, Addo IY, Adegboye OA, Adnan M, Adnani QES, Afzal MS, Afzal S, Aghdam ZB, Ahinkorah BO, Ahmad A, Ahmad AR, Ahmad R, Ahmad S, Ahmad S, Ahmadi S, Ahmed A, Ahmed H, Ahmed JQ, Ahmed Rashid T, Ajami M, Aji B, Akbarzadeh-Khiavi M, Akunna CJ, Al Hamad H, Alahdab F, Al-Aly Z, Aldeyab MA, Aleman AV, Alhalaiqa FAN, Alhassan RK, Ali BA, Ali L, Ali SS, Alimohamadi Y, Alipour V, Alizadeh A, Aljunid SM, Allel K, Almustanyir S, Ameyaw EK, Amit AML, Anandavelane N, Ancuceanu R, Andrei CL, Andrei T, Anggraini D, Ansar A, Anyasodor AE, Arabloo J, Aravkin AY, Areda D, Aripov T, Artamonov AA, Arulappan J, Aruleba RT, Asaduzzaman M, Ashraf T, Athari SS, Atlaw D, Attia S, Ausloos M, Awoke T, Ayala Quintanilla BP, Ayana TM, Azadnajafabad S, Azari Jafari A, B DB, Badar M, Badiye AD, Baghcheghi N, Bagherieh S, Baig AA, Banerjee I, Barac A, Bardhan M, Barone-Adesi F, Barqawi HJ, Barrow A, Baskaran P, Basu S, Batiha A-MM, Bedi N, Belete MA, Belgaumi UI, Bender RG, Bhandari B, Bhandari D, Bhardwaj P, Bhaskar S, Bhattacharyya K, Bhattarai S, Bitaraf S, Buonsenso D, Butt ZA, Caetano dos Santos FL, Cai J, Calina D, Camargos P, Cámera LA, Cárdenas R, Cevik M, Chadwick J, Charan J, Chaurasia A, Ching PR, Choudhari SG, Chowdhury EK, Chowdhury FR, Chu D-T, Chukwu IS, Dadras O, Dagnaw FT, Dai X, Das S, Dastiridou A, Debela SA, Demisse FW, Demissie S, Dereje D, Derese M, Desai HD, Dessalegn FN, Dessalegni SAA, Desye B, Dhaduk K, Dhimal M, Dhingra S, Diao N, Diaz D, Djalalinia S, Dodangeh M, Dongarwar D, Dora BT, Dorostkar F, Dsouza HL, Dubljanin E, Dunachie SJ, Durojaiye OC, Edinur HA, Ejigu HB, Ekholuenetale M, Ekundayo TC, El-Abid H, Elhadi M, Elmonem MA, Emami A, Engelbert Bain L, Enyew DB, Erkhembayar R, Eshrati B, Etaee F, Fagbamigbe AF, Falahi S, Fallahzadeh A, Faraon EJA, Fatehizadeh A, Fekadu G, Fernandes JC, Ferrari A, Fetensa G, Filip I, Fischer F, Foroutan M, Gaal PA, Gadanya MA, Gaidhane AM, Ganesan B, Gebrehiwot M, Ghanbari R, Ghasemi Nour M, Ghashghaee A, Gholamrezanezhad A, Gholizadeh A, Golechha M, Goleij P, Golinelli D, Goodridge A, Gunawardane DA, Guo Y, Gupta RD, Gupta S, Gupta VB, Gupta VK, Guta A, Habibzadeh P, Haddadi Avval A, Halwani R, Hanif A, Hannan MdA, Harapan H, Hassan S, Hassankhani H, Hayat K, Heibati B, Heidari G, Heidari M, Heidari-Soureshjani R, Herteliu C, Heyi DZ, Hezam K, Hoogar P, Horita N, Hossain MM, Hosseinzadeh M, Hostiuc M, Hostiuc S, Hoveidamanesh S, Huang J, Hussain S, Hussein NR, Ibitoye SE, Ilesanmi OS, Ilic IM, Ilic MD, Imam MT, Immurana M, Inbaraj LR, Iradukunda A, Ismail NE, Iwu CCD, Iwu CJ, J LM, Jakovljevic M, Jamshidi E, Javaheri T, Javanmardi F, Javidnia J, Jayapal SK, Jayarajah U, Jebai R, Jha RP, Joo T, Joseph N, Joukar F, Jozwiak JJ, Kacimi SEO, Kadashetti V, Kalankesh LR, Kalhor R, Kamal VK, Kandel H, Kapoor N, Karkhah S, Kassa BG, Kassebaum NJ, Katoto PD, Keykhaei M, Khajuria H, Khan A, Khan IA, Khan M, Khan MN, Khan MA, Khatatbeh MM, Khater MM, Khayat Kashani HR, Khubchandani J, Kim H, Kim MS, Kimokoti RW, Kissoon N, Kochhar S, Kompani F, Kosen S, Koul PA, Koulmane Laxminarayana SL, Krapp Lopez F, Krishan K, Krishnamoorthy V, Kulkarni V, Kumar N, Kurmi OP, Kuttikkattu A, Kyu HH, Lal DK, Lám J, Landires I, Lasrado S, Lee S, Lenzi J, Lewycka S, Li S, Lim SS, Liu W, Lodha R, Loftus MJ, Lohiya A, Lorenzovici L, Lotfi M, Mahmoodpoor A, Mahmoud MA, Mahmoudi R, Majeed A, Majidpoor J, Makki A, Mamo GA, Manla Y, Martorell M, Matei CN, McManigal B, Mehrabi Nasab E, Mehrotra R, Melese A, Mendoza-Cano O, Menezes RG, Mentis A-FA, Micha G, Michalek IM, Micheletti Gomide Nogueira de Sá AC, Milevska Kostova N, Mir SA, Mirghafourvand M, Mirmoeeni S, Mirrakhimov EM, Mirza-Aghazadeh-Attari M, Misganaw AS, Misganaw A, Misra S, Mohammadi E, Mohammadi M, Mohammadian-Hafshejani A, Mohammed S, Mohan S, Mohseni M, Mokdad AH, Momtazmanesh S, Monasta L, Moore CE, Moradi M, Moradi Sarabi M, Morrison SD, Motaghinejad M, Mousavi Isfahani H, Mousavi Khaneghah A, Mousavi-Aghdas SA, Mubarik S, Mulita F, Mulu GBB, Munro SB, Muthupandian S, Nair TS, Naqvi AA, Narang H, Natto ZS, Naveed M, Nayak BP, Naz S, Negoi I, Nejadghaderi SA, Neupane Kandel S, Ngwa CH, Niazi RK, Nogueira de Sá AT, Noroozi N, Nouraei H, Nowroozi A, Nuñez-Samudio V, Nutor JJ, Nzoputam CI, Nzoputam OJ, Oancea B, Obaidur RM, Ojha VA, Okekunle AP, Okonji OC, Olagunju AT, Olusanya BO, Omar Bali A, Omer E, Otstavnov N, Oumer B, P A M, Padubidri JR, Pakshir K, Palicz T, Pana A, Pardhan S, Paredes JL, Parekh U, Park E-C, Park S, Pathak A, Paudel R, Paudel U, Pawar S, Pazoki Toroudi H, Peng M, Pensato U, Pepito VCF, Pereira M, Peres MFP, Perico N, Petcu I-R, Piracha ZZ, Podder I, Pokhrel N, Poluru R, Postma MJ, Pourtaheri N, Prashant A, Qattea I, Rabiee M, Rabiee N, Radfar A, Raeghi S, Rafiei S, Raghav PR, Rahbarnia L, Rahimi-Movaghar V, Rahman M, Rahman MA, Rahmani AM, Rahmanian V, Ram P, Ranjha MMAN, Rao SJ, Rashidi M-M, Rasul A, Ratan ZA, Rawaf S, Rawassizadeh R, Razeghinia MS, Redwan EMM, Regasa MT, Remuzzi G, Reta MA, Rezaei N, Rezapour A, Riad A, Ripon RK, Rudd KE, Saddik B, Sadeghian S, Saeed U, Safaei M, Safary A, Safi SZ, Sahebazzamani M, Sahebkar A, Sahoo H, Salahi S, Salahi S, Salari H, Salehi S, Samadi Kafil H, Samy AM, Sanadgol N, Sankararaman S, Sanmarchi F, Sathian B, Sawhney M, Saya GK, Senthilkumaran S, Seylani A, Shah PA, Shaikh MA, Shaker E, Shakhmardanov MZ, Sharew MM, Sharifi-Razavi A, Sharma P, Sheikhi RA, Sheikhy A, Shetty PH, Shigematsu M, Shin JI, Shirzad-Aski H, Shivakumar KM, Shobeiri P, Shorofi SA, Shrestha S, Sibhat MM, Sidemo NB, Sikder MK, Silva LMLR, Singh JA, Singh P, Singh S, Siraj MS, Siwal SS, Skryabin VY, Skryabina AA, Socea B, Solomon DD, Song Y, Sreeramareddy CT, Suleman M, Suliankatchi Abdulkader R, Sultana S, Szócska M, Tabatabaeizadeh S-A, Tabish M, Taheri M, Taki E, Tan K-K, Tandukar S, Tat NY, Tat VY, Tefera BN, Tefera YM, Temesgen G, Temsah M-H, Tharwat S, Thiyagarajan A, Tleyjeh II, Troeger CE, Umapathi KK, Upadhyay E, Valadan Tahbaz S, Valdez PR, Van den Eynde J, van Doorn HR, Vaziri S, Verras G-I, Viswanathan H, Vo B, Waris A, Wassie GT, Wickramasinghe ND, Yaghoubi S, Yahya GATY, Yahyazadeh Jabbari SH, Yigit A, Yiğit V, Yon DK, Yonemoto N, Zahir M, Zaman BA, Zaman SB, Zangiabadian M, Zare I, Zastrozhin MS, Zhang Z-J, Zheng P, Zhong C, Zoladl M, Zumla A, Hay SI, Dolecek C, Sartorius B, Murray CJL, Naghavi M. 2022. Global mortality associated with 33 bacterial pathogens in 2019: a systematic analysis for the global burden of disease study 2019. The Lancet 400:2221–2248. doi: 10.1016/S0140-6736(22)02185-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Murray CJL, Ikuta KS, Sharara F, Swetschinski L, Robles Aguilar G, Gray A, Han C, Bisignano C, Rao P, Wool E, Johnson SC, Browne AJ, Chipeta MG, Fell F, Hackett S, Haines-Woodhouse G, Kashef Hamadani BH, Kumaran EAP, McManigal B, Achalapong S, Agarwal R, Akech S, Albertson S, Amuasi J, Andrews J, Aravkin A, Ashley E, Babin F-X, Bailey F, Baker S, Basnyat B, Bekker A, Bender R, Berkley JA, Bethou A, Bielicki J, Boonkasidecha S, Bukosia J, Carvalheiro C, Castañeda-Orjuela C, Chansamouth V, Chaurasia S, Chiurchiù S, Chowdhury F, Clotaire Donatien R, Cook AJ, Cooper B, Cressey TR, Criollo-Mora E, Cunningham M, Darboe S, Day NPJ, De Luca M, Dokova K, Dramowski A, Dunachie SJ, Duong Bich T, Eckmanns T, Eibach D, Emami A, Feasey N, Fisher-Pearson N, Forrest K, Garcia C, Garrett D, Gastmeier P, Giref AZ, Greer RC, Gupta V, Haller S, Haselbeck A, Hay SI, Holm M, Hopkins S, Hsia Y, Iregbu KC, Jacobs J, Jarovsky D, Javanmardi F, Jenney AWJ, Khorana M, Khusuwan S, Kissoon N, Kobeissi E, Kostyanev T, Krapp F, Krumkamp R, Kumar A, Kyu HH, Lim C, Lim K, Limmathurotsakul D, Loftus MJ, Lunn M, Ma J, Manoharan A, Marks F, May J, Mayxay M, Mturi N, Munera-Huertas T, Musicha P, Musila LA, Mussi-Pinhata MM, Naidu RN, Nakamura T, Nanavati R, Nangia S, Newton P, Ngoun C, Novotney A, Nwakanma D, Obiero CW, Ochoa TJ, Olivas-Martinez A, Olliaro P, Ooko E, Ortiz-Brizuela E, Ounchanum P, Pak GD, Paredes JL, Peleg AY, Perrone C, Phe T, Phommasone K, Plakkal N, Ponce-de-Leon A, Raad M, Ramdin T, Rattanavong S, Riddell A, Roberts T, Robotham JV, Roca A, Rosenthal VD, Rudd KE, Russell N, Sader HS, Saengchan W, Schnall J, Scott JAG, Seekaew S, Sharland M, Shivamallappa M, Sifuentes-Osornio J, Simpson AJ, Steenkeste N, Stewardson AJ, Stoeva T, Tasak N, Thaiprakong A, Thwaites G, Tigoi C, Turner C, Turner P, van Doorn HR, Velaphi S, Vongpradith A, Vongsouvath M, Vu H, Walsh T, Walson JL, Waner S, Wangrangsimakul T, Wannapinij P, Wozniak T, Young Sharma TEMW, Yu KC, Zheng P, Sartorius B, Lopez AD, Stergachis A, Moore C, Dolecek C, Naghavi M. 2022. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. The Lancet 399:629–655. doi: 10.1016/S0140-6736(21)02724-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Centers for Disease Control and Prevention . 2019. Antibiotic resistance threats in the United States. U.S. Department of Health and Human Services, CDC, Atlanta, GA. [Google Scholar]
- 4. Kolarič A, Anderluh M, Minovski N. 2020. Two decades of successful SAR-grounded stories of the novel bacterial topoisomerase inhibitors (NBTIs). J Med Chem 63:5664–5674. doi: 10.1021/acs.jmedchem.9b01738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Bates AD, Maxwell A. 2005. DNA topology. Oxford Press, USA. [Google Scholar]
- 6. Champoux JJ. 2001. DNA topoisomerases: structure, function, and mechanism. Annu Rev Biochem 70:369–413. doi: 10.1146/annurev.biochem.70.1.369 [DOI] [PubMed] [Google Scholar]
- 7. Bax BD, Chan PF, Eggleston DS, Fosberry A, Gentry DR, Gorrec F, Giordano I, Hann MM, Hennessy A, Hibbs M, Huang J, Jones E, Jones J, Brown KK, Lewis CJ, May EW, Saunders MR, Singh O, Spitzfaden CE, Shen C, Shillings A, Theobald AJ, Wohlkonig A, Pearson ND, Gwynn MN. 2010. Type IIA topoisomerase inhibition by a new class of antibacterial agents. Nature 466:935–940. doi: 10.1038/nature09197 [DOI] [PubMed] [Google Scholar]
- 8. Gibson EG, Bax B, Chan PF, Osheroff N. 2019. Mechanistic and structural basis for the actions of the antibacterial gepotidacin against Staphylococcus aureus gyrase. ACS Infect Dis 5:570–581. doi: 10.1021/acsinfecdis.8b00315 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Gibson EG, Oviatt AA, Cacho M, Neuman KC, Chan PF, Osheroff N. 2019. Bimodal actions of a naphthyridone/aminopiperidine-based antibacterial that targets gyrase and topoisomerase IV. Biochem 58:4447–4455. doi: 10.1021/acs.biochem.9b00805 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Aldred KJ, Kerns RJ, Osheroff N. 2014. Mechanism of quinolone action and resistance. Biochem 53:1565–1574. doi: 10.1021/bi5000564 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gibson EG, Ashley RE, Kerns RJ, Osheroff N. 2018. Bacterial type II topoisomerases and target-mediated drug resistance, p 507–529. In Fong IW, Shlaes D, Drlica K (ed), Antimicrobial resistance in the 21st century. Springer International Publishing, Cham. doi: 10.1007/978-3-319-78538-7 [DOI] [Google Scholar]
- 12. Bush NG, Diez-Santos I, Abbott LR, Maxwell A. 2020. Quinolones: mechanism, lethality and their contributions to antibiotic resistance. Molecules 25:5662. doi: 10.3390/molecules25235662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Overcash JS, Tiffany CA, Scangarella-Oman NE, Perry CR, Tao Y, Hossain M, Barth A, Dumont EF. 2020. Phase 2a pharmacokinetic, safety, and exploratory efficacy evaluation of oral gepotidacin (GSK2140944) in female participants with uncomplicated urinary tract infection (acute uncomplicated cystitis). Antimicrob Agents Chemother 64:e00199-20. doi: 10.1128/AAC.00199-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. O’Riordan W, Tiffany C, Scangarella-Oman N, Perry C, Hossain M, Ashton T, Dumont E. 2017. Efficacy, safety, and tolerability of gepotidacin (GSK2140944) in the treatment of patients with suspected or confirmed gram-positive acute bacterial skin and skin structure infections. Antimicrob Agents Chemother 61:e02095-16. doi: 10.1128/AAC.02095-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Taylor SN, Morris DH, Avery AK, Workowski KA, Batteiger BE, Tiffany CA, Perry CR, Raychaudhuri A, Scangarella-Oman NE, Hossain M, Dumont EF. 2018. Gepotidacin for the treatment of uncomplicated urogenital gonorrhea: a phase 2, randomized, dose-ranging, single-oral dose evaluation. Clin Infect Dis 67:504–512. doi: 10.1093/cid/ciy145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kolarič A, Minovski N. 2018. Novel bacterial topoisomerase inhibitors: challenges and perspectives in reducing hERG toxicity. Future Med Chem 10:2241–2244. doi: 10.4155/fmc-2018-0272 [DOI] [PubMed] [Google Scholar]
- 17. Hossain M, Zhou M, Tiffany C, Dumont E, Darpo B. 2017. A phase I, randomized, double-blinded, placebo- and moxifloxacin-controlled, four-period crossover study to evaluate the effect of gepotidacin on cardiac conduction as assessed by 12-lead electrocardiogram in healthy volunteers. Antimicrob Agents Chemother 61:e02385-16. doi: 10.1128/AAC.02385-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Scangarella-Oman NE, Ingraham KA, Tiffany CA, Tomsho L, Van Horn SF, Mayhew DN, Perry CR, Ashton TC, Dumont EF, Huang J, Brown JR, Miller LA. 2020. In vitro activity and microbiological efficacy of gepotidacin from a phase 2, randomized, multicenter, dose-ranging study in patients with acute bacterial skin and skin structure infections. Antimicrob Agents Chemother 64:e01302-19. doi: 10.1128/AAC.01302-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lahiri SD, Kutschke A, McCormack K, Alm RA. 2015. Insights into the mechanism of inhibition of novel bacterial topoisomerase inhibitors from characterization of resistant mutants of Staphylococcus aureus. Antimicrob Agents Chemother 59:5278–5287. doi: 10.1128/AAC.00571-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Lu Y, Vibhute S, Li L, Okumu A, Ratigan SC, Nolan S, Papa JL, Mann CA, English A, Chen A, Seffernick JT, Koci B, Duncan LR, Roth B, Cummings JE, Slayden RA, Lindert S, McElroy CA, Wozniak DJ, Yalowich J, Mitton-Fry MJ, Optimization of TopoIV Potency . 2021. Optimization of TopoIV potency, ADMET properties, and hERG inhibition of 5-amino-1,3-dioxane-linked novel bacterial topoisomerase inhibitors: identification of a lead with In vivo efficacy against MRSA. J Med Chem 64:15214–15249. doi: 10.1021/acs.jmedchem.1c01250 [DOI] [PubMed] [Google Scholar]
- 21. Lu Y, Mann CA, Nolan S, Collins JA, Parker E, Papa J, Vibhute S, Jahanbakhsh S, Thwaites M, Hufnagel D, Hazbón MH, Moreno J, Stedman TT, Wittum T, Wozniak DJ, Osheroff N, Yalowich JC, Mitton-Fry MJ. 2022. 1,3-Dioxane-linked novel bacterial topoisomerase inhibitors: expanding structural diversity and the antibacterial spectrum. ACS Med Chem Lett 13:955–963. doi: 10.1021/acsmedchemlett.2c00111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lu Y, Papa JL, Nolan S, English A, Seffernick JT, Shkolnikov N, Powell J, Lindert S, Wozniak DJ, Yalowich J, Mitton-Fry MJ. 2020. Dioxane-linked amide derivatives as novel bacterial topoisomerase inhibitors against gram-positive Staphylococcus aureus. ACS Med Chem Lett 11:2446–2454. doi: 10.1021/acsmedchemlett.0c00428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Lyons A, Kirkham J, Blades K, Orr D, Dauncey E, Smith O, Dick E, Walker R, Matthews T, Bunt A, Finlayson J, Morrison I, Savage VJ, Moyo E, Butler HS, Newman R, Ooi N, Smith A, Charrier C, Ratcliffe AJ, Stokes NR, Best S, Salisbury A-M, Craighead M, Cooper IR. 2022. Discovery and structure-activity relationship of a novel oxazolidinone class of bacterial type II topoisomerase inhibitors. Bioorg Med Chem Lett 65:128648. doi: 10.1016/j.bmcl.2022.128648 [DOI] [PubMed] [Google Scholar]
- 24. Kokot M, Weiss M, Zdovc I, Senerovic L, Radakovic N, Anderluh M, Minovski N, Hrast M. 2023. Amide containing NBTI antibacterials with reduced hERG inhibition, retained antimicrobial activity against gram-positive bacteria and in vivo efficacy. Eur J Med Chem 250:115160. doi: 10.1016/j.ejmech.2023.115160 [DOI] [PubMed] [Google Scholar]
- 25. Li L, Okumu AA, Nolan S, English A, Vibhute S, Lu Y, Hervert-Thomas K, Seffernick JT, Azap L, Cole SL, Shinabarger D, Koeth LM, Lindert S, Yalowich JC, Wozniak DJ, Mitton-Fry MJ. 2019. 1,3-Dioxane-linked bacterial topoisomerase inhibitors with enhanced antibacterial activity and reduced hERG inhibition. ACS Infect Dis 5:1115–1128. doi: 10.1021/acsinfecdis.8b00375 [DOI] [PubMed] [Google Scholar]
- 26. Gleeson MP. 2008. Generation of a set of simple, interpretable ADMET rules of thumb. J Med Chem 51:817–834. doi: 10.1021/jm701122q [DOI] [PubMed] [Google Scholar]
- 27. Projan SJ, Carleton S, Novick RP. 1983. Determination of plasmid copy number by fluorescence densitometry. Plasmid 9:182–190. doi: 10.1016/0147-619x(83)90019-7 [DOI] [PubMed] [Google Scholar]
- 28. Ritke MK, Roberts D, Allan WP, Raymond J, Bergoltz VV, Yalowich JC. 1994. Altered stability of etoposide-induced topoisomerase II-DNA complexes in resistant human leukaemia K562 cells. Br J Cancer 69:687–697. doi: 10.1038/bjc.1994.131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Hernandez VA, Carvajal-Moreno J, Papa JL, Shkolnikov N, Li J, Ozer HG, Yalowich JC, Elton TS. 2021. CRISPR/Cas9 genome editing of the human topoisomerase IIα intron-19 5′ splice site circumvents etoposide resistance in human leukemia K562 cells. Mol Pharmacol 99:226–241. doi: 10.1124/molpharm.120.000173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Olive PL. 2002. The comet assay, p 179–194. In Didenko VV (ed), In situ detection of DNA damage: methods and protocols. Humana Press, NJ. [Google Scholar]
- 31. Gyori BM, Venkatachalam G, Thiagarajan PS, Hsu D, Clement M-V. 2014. Opencomet: an automated tool for comet assay image analysis. Redox Biol 2:457–465. doi: 10.1016/j.redox.2013.12.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Hasinoff BB, Wu X, Krokhin OV, Ens W, Standing KG, Nitiss JL, Sivaram T, Giorgianni A, Yang S, Jiang Y, Yalowich JC. 2005. Biochemical and proteomics approaches to characterize topoisomerase Iiα cysteines and DNA as targets responsible for cisplatin-induced inhibition of topoisomerase IIα. Mol Pharmacol 67:937–947. doi: 10.1124/mol.104.004416 [DOI] [PubMed] [Google Scholar]
- 33. Veber DF, Johnson SR, Cheng H-Y, Smith BR, Ward KW, Kopple KD. 2002. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem 45:2615–2623. doi: 10.1021/jm020017n [DOI] [PubMed] [Google Scholar]
- 34. Richter MF, Drown BS, Riley AP, Garcia A, Shirai T, Svec RL, Hergenrother PJ. 2017. Predictive compound accumulation rules yield a broad-spectrum antibiotic. Nature 545:299–304. doi: 10.1038/nature22308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Singh SB, Kaelin DE, Wu J, Miesel L, Tan CM, Gill C, Black T, Nargund R, Meinke PT, Olsen DB, Lagrutta A, Wei C, Peng X, Wang X, Fukuda H, Kishii R, Takei M, Takeuchi T, Shibue T, Ohata K, Takano H, Ban S, Nishimura A, Fukuda Y. 2015. Hydroxy tricyclic 1,5-napthyridinone oxabicyclooctane-linked novel bacterial topoisomerase inhibitors as broad-spectrum antibacterial agents-SAR of RHS moiety (Part 3). Bioorg Med Chem Lett 25:2473–2478. doi: 10.1016/j.bmcl.2015.04.063 [DOI] [PubMed] [Google Scholar]
- 36. Michalczyk E, Hommernick K, Behroz I, Kulike M, Pakosz-Stępień Z, Mazurek L, Seidel M, Kunert M, Santos K, von Moeller H, Loll B, Weston JB, Mainz A, Heddle JG, Süssmuth RD, Ghilarov D. 2023. Molecular mechanism of topoisomerase poisoning by the peptide antibiotic albicidin. Nat Catal 6:52–67. doi: 10.1038/s41929-022-00904-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Hawtin RE, Stockett DE, Byl JAW, McDowell RS, Nguyen T, Arkin MR, Conroy A, Yang W, Osheroff N, Fox JA. 2010. Voreloxin is an anticancer quinolone derivative that intercalates DNA and poisons topoisomerase II. PLoS One 5:e10186. doi: 10.1371/journal.pone.0010186 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Clinical and Laboratory Standards Institute . 2018. Methods for dilution antimicrobial susceptibility tests for bacteria that grow Aerobically; approved standard - 11The Ed. In CLSI standard M07. Clinical and Laboratory Standards Institute, Wayne, PA. [Google Scholar]






