Abstract
β-Lactams are the most widely used antibiotics for the treatment of bacterial infections because of their proven track record of safety and efficacy. However, susceptibility to β-lactam antibiotics is continually eroded by resistance mechanisms. Emerging multidrug-resistant (MDR) Neisseria gonorrhoeae strains possessing altered penA alleles (encoding PBP2) pose a global health emergency as they threaten the utility of ceftriaxone, the last remaining outpatient antibiotic. Here we disclose a novel benzoxaborinine-based penicillin-binding protein inhibitor series (boro-PBPi) that is envisioned to address penA-mediated resistance while offering protection against evolution and expansion of β-lactamases. Optimization of boro-PBPi led to the identification of compound 21 (VNRX-14079) that exhibits potent antibacterial activity against MDR N. gonorrhoeae achieved by high affinity binding to the PBP2 target. Boro-PBPi/PBP2 complex structures confirmed covalent interaction of the boron atom with Ser310 and the importance of the β3-β4 loop for improved affinity. 21 elicits bactericidal activity, a low frequency of resistance, a good safety profile, suitable pharmacokinetic properties, and in vivo efficacy in a murine infection model against ceftriaxone-resistant N. gonorrhoeae. 21 is a promising anti-gonorrhea agent poised for further advancement.
Keywords: non-β-lactam, PBP inhibitor, boron, penicillin-binding protein, gonorrhea, antibiotic
INTRODUCTION
For the treatment of bacterial infections, β-lactam antibiotics, including penicillins, cephalosporins, monobactams, and carbapenems, represent over 60% of the total antibiotic market, reflecting their proven safety and efficacy record over >70 years of clinical use1. However, bacterial pathogens are increasingly resistant to β-lactam drugs, mainly due to the rapid evolution of β-lactamase enzymes that chemically inactivate these clinically important agents2, 3. The therapeutic targets of β-lactams are the penicillin-binding proteins (PBPs), which are unique to bacteria, essential for cell wall synthesis, and indispensable for bacterial proliferation4, 5. The clinical success of β-lactams validates PBPs as extremely valuable antibacterial targets. To overcome β-lactamase-mediated resistance, combination therapies of BL with β-lactamase inhibitors (BLI) have been developed, but resistance to these β-lactam/BLI combinations is rapidly developing6.
An alternative approach that enables continued targeting of PBPs is to develop non-β-lactam PBP inhibitors that avoid degradation by β-lactamases5. Since serine β-lactamases and PBPs share similar active site residues and catalytic mechanism, PBP inhibition utilizing current β-lactamase inhibitor scaffolds that are refractory to inactivation by β-lactamases (e.g., diazabicyclooctanes (DBOs) and benzoxaborinine-based inhibitors) has been explored5, 7. Since non-β-lactam PBP inhibitors will likely evade the action of all known and future β-lactamases, these novel PBP inhibitors may circumvent the evolution of β-lactamases and reset the resistance clock for PBP targeting agents.
Gonorrhea, caused by Neisseria gonorrhoeae, is the second most prevalent sexually transmitted bacterial infection with an estimated 82.4 million cases worldwide in 20208. If left untreated, gonorrhea can lead to serious health complications, including infertility in both men and women, pelvic inflammatory disease, and increased risk of HIV/AIDS8. Since the introduction of the sulfa drugs in the 1930s, N. gonorrhoeae has evolved under constant antimicrobial drug pressure. As such, owing to acquired resistance to nearly every antibiotic class including sulfa drugs, penicillins, tetracycline, ciprofloxacin, azithromycin, and cephalosporins, there has been a parallel evolution of therapeutic recommendations9, 10, 11, 12, 13, 14. Although gonorrhea is treatable and can be cured by current antibiotics, the emergence of N. gonorrhoeae resistant to antibiotics is making treatment of gonorrhea more challenging, forecasting future failings of outpatient therapy. Therefore, multidrug-resistant N. gonorrhoeae has been classified as an “urgent threat” by the Centers for Disease Control and Prevention (CDC) and the World Health Organization (WHO) has declared dual cephalosporin-resistant and fluoroquinolone-resistant N. gonorrhoeae as a “High Priority” pathogen for the discovery and development of new antibiotics15, 16, 17. The current last-line therapeutic option to treat uncomplicated gonorrhea is a single intramuscular (IM) dose (250–1000 mg) of the third-generation cephalosporin, ceftriaxone (CRO)18. Unfortunately, global CRO-resistant N. gonorrhoeae infections are increasing in frequency, foreshadowing an era of untreatable gonorrhea infections in the outpatient setting that would result in an unsustainable burden on healthcare systems19, 20. As there are no effective vaccines, antibiotic therapy remains the only option for treatment21.
Protein variants of penA, encoding penicillin-binding protein 2 (PBP2) (functionally equivalent to the cell division protein PBP3 in Escherichia coli), are the predominant mechanism of CRO-resistance in N. gonorrhoeae with the most urgent threat mediated by the mosaic penA-60 allele (a “mosaic PBP2” refers to a variant of PBP2 protein composed of segments derived from several Neisseria species and are often associated with decreased susceptibility to cephalosporin antibiotics)13, 20. Notably, N. gonorrhoeae strains possessing penA-60 have been spreading globally since 2015 when it was first discovered in the FC428 strain isolated in Japan19, 22, 23, 24, 25, 26, 27. In addition, N. gonorrhoeae can acquire genetic elements to produce an extended spectrum β-lactamase (ESBL) or carbapenemase which can hydrolyze CRO and other third generation cephalosporins28. Although production of the ESBL TEM-20 imposes a fitness cost in N. gonorrhoeae29, the recent report of an N. gonorrhoeae clinical isolate producing the ESBL CTX-M-1 serves as an early harbinger of the possible emergence of ESBL-mediated CRO resistance30.
Of the recent agents developed for the treatment of multi-drug-resistant N. gonorrhoeae, only zoliflodacin and gepotidacin, both of which target bacterial DNA replication (i.e., topoisomerases/gyrase inhibitors), have successfully completed phase 3 clinical evaluation for uncomplicated gonorrhea31, 32, 33. Although both agents are expected to gain approval, there continues to be a need for new anti-gonorrhea agents — particularly those with differing modes of action. Notably, while zoliflodacin-resistant strains have only been isolated in vitro, gepotidacin-resistant strains have already been identified within the community setting34. These observations portend emerging resistance to even these novel agents35, 36. Aside from the topoisomerase/gyrase inhibitors, single-dose IM-ertapenem (1 g) was recently found to be non-inferior to IM-CRO (500 mg) in a phase 3 trial37. Clearly, there is an urgent need for the discovery and development of additional safe and effective anti-gonorrhea therapies for use in the outpatient setting.
We have identified a series of non-β-lactam inhibitors of PBP transpeptidase function, i.e., PBPi, based on the α-alkylamido benzoxaborinine scaffold also present in the novel BLIs taniborbactam38, 39 and ledaborbactam etzadroxil (Figure S1)40, 41. Owing to the similarity within the β-lactamase and PBP active sites, boronate-based inhibitors can exploit several common binding elements, including the slowly reversible interaction of the active site serine hydroxyl group with the boron atom of the benzoxaborinine core38, 39, 42, 43, 44. Here we present a novel benzoxaborinine-based PBP inhibitor series (boro-PBPi) that exhibits activity against CRO-resistant, mosaic PBP2-containing N. gonorrhoeae variants. Moreover, we provide in vitro and in vivo data to support further preclinical development of boro-PBPi 21 (VNRX-14079) as a potential new agent for the treatment of N. gonorrhoeae infections, including those resistant to the current standard of care agent, CRO.
RESULTS
Identification of benzoxaborinine-based PBPi with stand-alone antibacterial activity
To adapt the BLI-focused benzoxaborinine template for use as a PBPi, we first introduced the aminothiazole-containing oxyimino-acetamide sidechain present in the third generation cephalosporins, cefdinir, and CRO (Figure S2), onto the benzoxaborinine warhead, generating boro-PBPi 1 (Figure 1). In vitro testing of 1 against a selected panel of Gram-negative organisms revealed modest single-agent activity against certain E. coli and K. pneumoniae strains (Table S1). Notably, 1 exhibited only 4-fold improved activity (minimal inhibitory concentration, MIC of 8 μg/mL) against the hyperpermeable E. coli BAS901C strain relative to the ATCC 25922 wild type strain, suggesting that its antibacterial activity might be limited by weak affinity for target PBPs (E. coli PBP3 IC50 of 113 μM). Importantly, 1 possessed modest activity against non-mosaic PBP2-producing N. gonorrhoeae ATCC 49226 (MIC of 16 μg/mL, relative to ceftriaxone MIC of 0.008 μg/mL) with an IC50 of 100 μM against wild type PBP2 (PBP2WT) isolated from the N. gonorrhoeae FA19 strain. However, 1 was inactive (MIC of >128 μg/mL) against the H041 strain producing the mosaic PBP2 that mediates CRO-resistance.
Figure 1. Evolution of boro-PBPi from BLI-containing benzoxaborinine core to ureido-containing lead compound 21 (VNRX-14079).
The table shows PBP2 binding and MIC data for ceftriaxone, cefoperazone, and selected boro-PBPi.
Incorporation of the ureido moiety for improved activity against N. gonorrhoeae
We next chose to replace the oxyimino amide sidechain with the ureido-based aryl glycine motif present in piperacillin and cefoperazone (Figure S2). The initial ureido-containing benzoxaborinines were prepared as near equimolar mixtures of the R,R- and S,R-diastereomers (Supplementary Information); when required, the desired R,R-diastereomers (infra) were isolated following preparative reversed-phase high performance liquid chromatography (RP-HPLC). The parent phenol-bearing boro-PBPi 2 (Figure 1), a benzoxaborinine linked to the ureido side chain of cefoperazone (Figure S2), showed moderate antibacterial activity against representative N. gonorrhoeae strains producing non-mosaic PBP2 and exhibited good in vitro binding to PBP2WT (IC50 of 2.8 μM). However, poor binding affinity was observed for 2 against the mosaic PBP2 isolated from the CRO-resistant H041 strain (PBP2H041, IC50 of 770 μM). Correspondingly, across a panel of N. gonorrhoeae strains, MICs for 2 were higher in the mosaic PBP2-containing strains compared to the strains with non-mosaic PBP2 (Figure 1).
A series of phenol-bearing boro-PBPi was prepared and evaluated for PBP2WT binding and antibacterial activity against both CRO-susceptible ATCC 49226 and CRO-resistant H041 strains – an abbreviated, yet informative list of analogs is provided in Figure S3. The introduction of a fluorine atom ortho to the parent phenol provided R,R-3 as a single isomer after RP-HPLC-mediated separation of the diastereomers. In both CRO-susceptible and resistant strains, 3 exhibited comparable MICs to 2, which had been tested as a ~1:1 mixture of active and inactive diastereomers. R,R-4 proved less active against ATCC 49226 than 3, while exhibiting only modest activity against the CRO-resistant H041 strain (MIC of 16 μg/mL). Additional fluorinated analogs 5–9 revealed improved PBP2WT binding relative to non-fluorinated 2 and had comparable or lower MIC values against ATCC 49226. Although most of these analogs plateaued at MIC of 16–32 μg/mL when tested against CRO-resistant H041, trifluorinated 9 displayed a slightly improved MIC of 8 μg/mL against this mosaic PBP2-containing strain (Figure S3), suggesting that inclusion of fluoro groups may have a positive effect on PBP2 binding and/or periplasmic accumulation.
To better understand the relationship between fluoro substitution and PBP2 binding and/or antibacterial activity, the pKa of the phenol proton for these modestly active analogs was calculated. While unsubstituted phenol has a pKa of 10.0, the inductive effect of fluoro substitution lowers the pKa of 2-fluoro- and 3-fluoro-phenol to 8.7 and 9.3, respectively. The 2,3,6-trifluoro-4-hydroxyphenyl glycine of 9 was calculated to have a pKa (Ar-OH) of 6.7, which implies ionization of the phenol at physiological pH. These data suggest that 9 likely exists as a phenoxide at physiological pH, which may be contributing to the observed antibacterial activity of these fluorine-containing boro-PBPi.
Benzoate- and phosphonate-containing benzoxaborinines improves boro-PBPi activity
Based on the hypothesis that a phenoxide anion might be beneficial at the para position of the aryl glycine moiety, a series of benzoate-containing boro-PBPi was investigated, with the expectation that a carboxylate residue might be similarly preferred. To this end, a suitable benzoate-based phenyl glycine intermediate (42) was prepared (Supplementary Information). The results from this set of benzoate-containing benzoxaborinines supported the hypothesis that an anion is preferred at the 4-position of the aryl glycine moiety. Both 10 and 11 displayed good activity against ATCC 49226 (MIC of 1 and 4 μg/mL, respectively), and importantly, both boro-PBPi exhibited activity against H041 (MIC of 8 μg/mL) at a level comparable to trifluorinated phenol 9 (Figure S3), with 11 showing measurable binding to mosaic PBP2H041 (Figure S4). Separation of the active isomer of 10, i.e., R,R-12, allowed identification of the first benzoxaborinine-based PBPi with MIC of 4 μg/mL against H041 (Figure 1). Both fluorinated single isomers, R,R-13 and R,R-14, also showed MICs of 4 μg/mL against H041 (Figure S4), further supporting this subseries as a promising lead benzoxaborinine PBPi for CRO-resistant gonococcal infection.
Seeking to identify alternatives to the putative phenoxide and carboxylate hydrogen bonding residues, a phosphonate group was incorporated onto the phenyl glycine moiety, preparing the non-fluorinated parent boro-PBPi R,R-15 (Figure 1). Strikingly, the addition of a phosphonate residue to these boro-PBPi resulted in a marked improvement in mosaic PBP2H041 binding and antibacterial activity against both CRO-susceptible and -resistant strains when compared to the phenol- or benzoate-containing boro-PBPi (Figure 1 and Figure S5). The 2,3-dioxapiperidine-containing analogs 15–17 showed good binding to WT and mosaic PBP2 and excellent MICs of 0.06–0.12 and 0.5 μg/mL against ATCC 49226 and H041 strains, respectively.
Having discovered the importance of the terminal phosphonate group for PBP2 engagement and bactericidal activity, a series of boro-PBPi bearing the N-methylsulfonyl-2-imidazolinone motif present in mezlocillin was prepared (Figures S2 and S6). These new boro-PBPi, represented by R,R-18 (Figure 1), had similar PBP2 binding affinity and antibacterial activity as the 2,3-dioxapiperidine-containing analogs. The incorporation of multiple fluorine atoms (boro-PBPi 19–23) led to R,R-21 (VNRX-14079) that showed a 2–8-fold improvement in MICs relative to 18 in mosaic PBP2-producing strains. In addition, 21 exhibited 8-fold lower MICs than CRO against the CRO-resistant H041 and WHO Q strains that produce mosaic PBP2H041 and PBP2FC428 (encoded by penA-60), respectively.
Structural evidence for high affinity binding of boro-PBPi to mosaic PBP2
Mutations in the transpeptidase (TPase) domain of PBP2 are firmly established as the primary determinant of cephalosporin resistance in N. gonorrhoeae45 and such mutations appear to act by restricting the conformational dynamics of the protein46, 47. We obtained the structures of benzoate-containing 12 and phosphonate-containing 15 complexes in the mosaic PBP2 transpeptidase domain isolated from the extended-spectrum-cephalosporin-decreased susceptibility strain 35/02, i.e., tPBP235/02 (Figure 2)47. In both structures, essentially complete electron density is observed for the inhibitors, including a covalent interaction between the boro-PBPi boron atom and Ser310 of the tPBP235/02 active site. In addition, the benzoxaborinine-associated carboxylate group is hydrogen bonded to Thr498 and Thr500, while Ser362 forms a hydrogen bond with the boronate ester oxygen atom. Furthermore, a hydrogen bond between Asn364 and the carbonyl group of the α-amido benzoxaborinine is observed. Notably, the para-carboxylate of 12 forms a hydrogen bond with Tyr543 in the β5-α11 loop (Figure 2A). Replacement of the benzoate moiety with a phosphonate residue in 15 leads to the formation of additional hydrogen bonds with Arg502 and His514, in addition to that with Tyr543 (Figure 2B).
Figure 2. Crystal structures of tPBP235/2 in complex with boro-PBPi 12 and 15.
A and B) The crystal structures of mosaic Ng tPBP235/2 bound to R,R-12 and R,R-15, respectively. Right hand panels show the interactions made by the para-carboxylate in 12 and phosphonate of 15. C) Superimposition of the crystal structures of Ng tPBP235/2 in complex with 15 and apo Ng tPBP235/2 (PDB: 6VBL) showing the movement of the β3-β4 loop toward the active site in the complex (arrow).
One of the most significant conformational changes observed upon boro-PBPi binding is the movement of the β3-β4 loop from a so-called “outbent” conformation as observed for apo tPBP235/02 to an inward conformation closer to the active site (Figure 2C). The inward movement of the β3-β4 loop in the crystal structures in complex with the boro-PBPi is highly significant, as its failure to move in PBP2 variants derived from extended-spectrum-cephalosporin-resistant strains of N. gonorrhoeae is hypothesized to be an important contributor to CRO-resistance47. The observed binding modes of 12 and 15, together with the movement of the β3-β4 loop, are similar to the binding modes of cefoperazone and the ureidopenicillins piperacillin and azlocillin observed in the tPBP2H041 protein48. These β-lactams exhibit faster second-order acylation rates against tPBP2H041 compared to the oxyimino-containing CRO48. These similar findings with the boro-PBPi are therefore consistent with the idea that higher-activity inhibitors work by overcoming the conformational barrier created by resistance mutations present in tPBP2 from resistant strains.
Pharmacokinetic evaluation and in vivo efficacy of boro-PBPi
In advance of the murine efficacy studies49, the plasma protein binding (PPB) and pharmacokinetic (PK) profiles of 18 and 21 were determined in mice. In mouse plasma, PPB of 18 and 21 was 7% and 19%, respectively, whereas PPB in human plasma was 24% and 39%, respectively. As shown in Figure S7, boro-PBPi 18 (formulated at 100 mg/mL in PBS, pH 6.5–7.0) and 21 (formulated at 75 mg/mL in PBS, pH 6–7) exhibited similar plasma-time concentration profiles for each compound, when administered to mice by either intravenous (IV) bolus or subcutaneous (SC) injection. Boro-PBPi 18 and 21 were well tolerated up to the highest dose tested (1,000 and 300 mg/kg SC doses, respectively). Exposure, in terms of maximum concentrations (Cmax) and total overall exposures (AUC), was less than dose-proportional for 21 following SC administration from 10 to 300 mg/kg, while for 18, exposure following SC exposure was less than dose proportional for Cmax at ≥300 mg/kg and was generally dose-proportional for AUCinf across all dose levels administered (Tables S2 and S3). The terminal half-life (t½) of 18 was approximately 4 h following SC dosing with good exposure at all doses. The terminal half-life of 21 was ~4 h at ≥100 mg/kg, whereas the half-life at 10 and 30 mg/kg was calculated to be short at <0.9 h because the terminal half-life of the lower doses was not determined as concentrations at later timepoints were below the limit of detection.
Assuming all drugs that target PBPs display time-dependent antibacterial activity in vivo as demonstrated for β-lactam-based PBP inhibitors50, 51, we selected the boro-PBPi dosages of 0.25, 7.5, 50, and 200 mg/kg every 12 h (q12h or BID) for the in vivo efficacy study in the murine vaginal infection model with the CRO-susceptible FA1090 strain [agar MIC of 0.016 μg/mL for 18 and 0.008 μg/mL for CRO determined by CLSI guidelines52 (Table 1)]. In vivo efficacy of 18 was observed at ≥7.5 mg/kg SC q12h, as PK simulations predicted that these doses would provide unbound boro-PBPi concentrations in plasma above the MIC for at least 40% of the 24 h treatment duration (i.e., ≥40% fT>MIC); bacterial growth stasis was observed at 0.25 mg/kg SC q12h (NB: ≤13% PK-simulated fT>MIC) (Figure S8). The efficacious doses of 18 were similar to the efficacy achieved by CRO at 1 mg/kg (IV, single dose).
Table 1.
Agar MIC of boro-PBPi and comparator agents against N. gonorrhoeae reference strains
| Strain | PBP2 allele | 12 | 15 | 18 | 21 | CRO | ZOLI | GEPO | AZM | CIP | PEN |
|---|---|---|---|---|---|---|---|---|---|---|---|
| ATCC 49226 | penA-22 | 0.5 | 0.06 | 0.06 | 0.03 | 0.016 | 0.12 | 0.25 | 0.25 | 0.004 | 0.5 |
| FA19 | penA-15 | ≤0.016 | 0.008 | 0.008 | 0.008 | 0.004 | 0.03 | 0.12 | 0.06 | ≤0.002 | ≤0.016 |
| FA1090 | penA-1 | 0.06 | 0.016 | 0.016 | 0.016 | 0.008 | 0.03 | 0.12 | 0.06 | 0.004 | 0.06 |
| WHO F | penA-15 | ≤0.016 | ≤0.004 | 0.008 | 0.008 | ≤0.002 | 0.06 | 0.12 | 0.06 | 0.004 | ≤0.016 |
| WHO G | penA-2 | 0.25 | 0.03 | 0.06 | 0.03 | 0.016 | 0.12 | 1 | 0.12 | 0.12 | 0.5 |
| WHO M | penA-2 | 0.5 | 0.06 | 0.06 | 0.06 | 0.016 | 0.06 | 1 | 0.25 | 2 | >64 |
| MS11 | penA-22 | 0.5 | 0.03 | 0.06 | 0.03 | 0.016 | 0.12 | 0.5 | 0.25 | 0.004 | 0.5 |
| WHO L | penA-7 | 8 | 1 | 1 | 0.5 | 0.25 | 0.12 | 4 | 0.25 | 16 | 2 |
| WHO K | Mosaic penA-10 | 4 | 0.25 | 0.5 | 0.12 | 0.12 | 0.12 | 0.25 | 0.25 | >16 | 2 |
| H041 | Mosaic penA-37 | 16 | 1 | 1 | 0.25 | 1 | 0.12 | 0.5 | 0.25 | >16 | 2 |
| WHO Z | Mosaic penA-64 | 2 | 0.25 | 0.25 | 0.06 | 0.5 | 0.12 | 0.25 | 1 | >16 | 2 |
| WHO Q (G7944) | Mosaic penA-60 | 4 | 0.5 | 0.25 | 0.06 | 0.5 | 0.06 | 1 | >4 | 16 | 1 |
| CDC-0197 | Mosaic penA-34 | 2 | 0.25 | 0.25 | 0.06 | 0.06 | 0.06 | 0.25 | >4 | 16 | 1 |
| F89 | Mosaic penA-42 | >16 | >8 | >8 | >8 | 2 | 0.12 | 0.5 | 0.25 | 16 | 2 |
Agar MIC (μg/mL)
Shown in red are resistant/non-susceptible MIC values based on the CLSI guideline77 where MIC breakpoints are defined only for ceftriaxone, azithromycin, ciprofloxacin, and penicillin G.
Abbreviations: CRO, ceftriaxone; ZOLI, zoliflodacin; GEPO, gepotidacin; AZM, azithromycin; CIP, ciprofloxacin; PEN G, penicillin G.
In vivo efficacy of 21 was evaluated in the murine vaginal infection model using the CRO-resistant H041 strain (agar MIC of 0.25 μg/mL for 21 and 1 μg/mL for CRO) as previously described51 (Figure S9). Four dosing groups were selected to provide %fT>MIC for 21, ranging from 20% to 100% when corrected for 19% mouse plasma protein binding. In this model, 21 showed good efficacy against the CRO-resistant H041 strain by demonstrating a statistically significant reduction in the percentage of culture-positive mice relative to vehicle control (p <0.0001) (Figure 3 and Table S5). The 150 mg/kg SC twice-a-day (q12h) and three-times-a-day (q8h) regimens resulted in 90% and 100% bacterial clearance by day 2, respectively, whereas 200 mg/kg SC single dose (q24h) achieved 100% clearance by day 3. Results with 10 mg/kg SC q12h were also significant with 80% clearance by day 4 relative to vehicle control (p <0.01). Bacterial bioburden (CFU/mL) in vaginal swab suspension was consistent with bacterial clearance (% mice cured of infection) in all treatment groups (Figure 3). Based on the PK data and simulations, the in vivo breakpoint of 21 for clearance of infections by H041 was a single dose of 200 mg/kg, which corresponds to a predicted fT/MIC of 44% on day 1 and is consistent with that observed for 18 against FA1090 (≥40% fT>MIC). Notably, neither the evolution of spontaneous gonococcal mutants nor a significant change in vaginal polymorphonuclear leukocytes (PMNs) was observed between groups during this study (Figure S10). No adverse effects were observed in mice treated with any dose of 21.
Figure 3. In vivo efficacy of boro-PBPi 21 in the murine vaginal infection model with ceftriaxone-resistant Neisseria gonorrhoeae H041.
A) The dosing scheme to examine in vivo efficacy of 21 against the ceftriaxone-resistant H041-STMR strain in the murine vaginal infection model is shown with predicted % fT/MIC. 21 was dosed only on day 1. Vaginal swab samples from infected mice were cultured to confirm H041 infection on day 0 prior to administration of test compound or controls on day 0 (green box). Vaginal cultures were collected on days 1–8 post treatment to determine the bacterial burden (CFU/mL). B) The percentage of mice infected over time with H041, and the average bacterial burden recovered over the course of the experiment. Over 85% of vehicle control mice remained colonized through day 5, with 60% colonized by day 8. SC-administered CRO, used as a positive control, yielded unanticipated underperformance relative to traditional intraperitoneal (IP) injection51. The horizontal dashed line denotes the limit of detection (20 CFU/mL). C) The average bacterial burden recovered over the course of the experiment. Error bars indicate standard error of the mean. All bacterial burden data are presented in Table S5.
Microbiological activity of boro-PBPi 21
21 showed comparable MICs to CRO against strains producing non-mosaic PBP2 and lower MICs against mosaic PBP2-producing strains, including the CRO-non-susceptible strain WHO Q, which belongs to the globally expanding FC428 lineage (Tables 1 and S7). The WHO M strain that produces TEM-1 β-lactamase was susceptible to 21, which is consistent with the stability of benzoxaborinines towards β-lactamase inactivation. Two strains, WHO L (containing non-mosaic PBP2) and F89 (containing mosaic PBP2), were the least susceptible strains to 21. WHO L carries a PBP2 A501V variant (Table S8) that reduces CRO susceptibility53, 54 and likely also affects activity of 21. The F89 strain has mosaic PBP2 (penA-42) which is identical to PBP2 (penA-34) of the CDC-0197 strain but with an additional A501P alteration (Table S8). The difference in MIC of boro-PBPi against N. gonorrhoeae F89 and CDC-0197 strains indicates that the A501P mutation impacts the activity of multiple boro-PBPi including 21. Notably, F89 and closely related strains were isolated in France and Spain in 2010 and 201155, 56, but since then, no clinical isolate carrying a PBP2 A501P variant has been reported likely owing to the fitness costs of producing this mosaic PBP257.
Zoliflodacin and gepotidacin, two novel topoisomerase inhibitors, are the most advanced agents in clinical development for the treatment of gonorrhea. Zoliflodacin, which had comparable activity to 21 in the mosaic PBP2-producing strains (except F89 as stated above), was the most active comparator agent (Table 1). Gepotidacin was slightly less active than zoliflodacin against the strain panel. As N. gonorrhoeae have evolved resistance to all current or former antibiotic regimens, resistance to these new agents is anticipated as well. In fact, resistance to these inhibitors mediated by topoisomerase mutations has already been reported34, 35, 36. Since the molecular target of boro-PBPi like 21 is PBP2, no cross-resistance to the topoisomerase inhibitors zoliflodacin or gepotidacin is expected. To confirm this, we isolated mutants with reduced susceptibility to zoliflodacin from WHO K (all mutants possessing GyrB D429N) and showed no cross-resistance to 21 in these mutants (Table S9).
Boro-PBPi were tested against the CDC panel of N. gonorrhoeae clinical isolates (n = 44) that include isolates with diverse antimicrobial susceptibilities to a variety of known drugs used to treat gonorrhea58. 21 showed MIC50 and MIC90 of 0.06 and 0.12 μg/mL, respectively, which were equivalent to CRO and slightly more active than zoliflodacin (Tables 2 and S10). As expected, based on the primary strain panel data, 18 showed a 4-fold higher MIC50 and MIC90 than 21, while the MIC50/MIC90 of benzoate-containing 12 was 8-fold higher than 18. In time-kill assays using three strains (ATCC 49226, WHO Q, and H041), both 21 and CRO reduced CFU/mL by ≥2 log10 in 6 h and below limit of detection at 24 h relative to untreated controls (i.e., 108 CFU/mL by 6 h) (Figure S11), confirming similar levels of bactericidal activity of 21 to CRO. The frequencies of resistance (FoR) to 21 and CRO were determined using ATCC 49226, WHO Q, and H041 on agar containing 4× and 16× MIC of each agent. No colonies arose on agar containing either 21 or CRO by 24 h, establishing FoR as <10−9 against all three strains (Table S11).
Table 2.
MIC50/MIC90 and MIC distribution of boro-PBPi and competitor compounds against 44 CDC isolates
| Compound | MIC50 | MIC90 | Number of strains at agar MIC (μg/mL) | |||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| ≤0.016 | 0.03 | 0.06 | 0.12 | 0.25 | 0.5 | 1 | 2 | 4 | ≥8 | |||
| Ceftriaxone | 0.06 | 0.12 | 4 | 2 | 25 | 12 | 1 | |||||
| 12 | 2 | 4 | 2 | 1 | 1 | 2 | 22 | 15 | 1 | |||
| 15 | 0.25 | 0.5 | 3 | 1 | 3 | 21 | 16 | |||||
| 18 | 0.25 | 0.5 | 3 | 1 | 3 | 16 | 21 | |||||
| 21 | 0.06 | 0.12 | 3 | 1 | 21 | 18 | 1 | |||||
| Zoliflodacin | 0.12 | 0.12 | 8 | 32 | 4 | |||||||
| Gepotidacin | 0.5 | 1 | 7 | 21 | 16 | |||||||
| Azithromycin | 0.5 | 1 | 1 | 18 | 17 | 4 | 1 | 3 | ||||
| Ciprofloxacin | 16 | 16 | 5 | 1 | 7 | 2 | 29 | |||||
| Penicillin G | 2 | 2 | 3 | 12 | 25 | 2 | ||||||
MIC values against each strain are shown in Table S10.
Boro-PBPi 21 exhibits promising pharmaceutical properties
21 showed no hemolytic activity when tested at 1 mg/mL in human red blood cells, no cytotoxicity at 256 μg/mL against 3 mammalian cell lines (HeLa, MRC-5, and 3T3), no mitochondrial toxicity (IC50 >100 μM) in a Glu/Gal assay using SKOV-3 human ovarian cancer cells, and no chromosomal aberrations (IC50 >300 μM) in the micronucleus assay using CHO-K1 cells with and without liver S9 fraction pre-incubation (Tables 3 and S12). When tested at 30 μM, 21 showed no noteworthy inhibition (≥10% decrease of baseline activity) of seven CYP enzymes (CYP1A2, CYP2D6, CYP3A4, CYP2E1, CYP2B6, CYP2C9, and CYP2C8) and low level inhibition (decrease of 34.6% of baseline activity) of CYP 2C19 and three mammalian serine proteases (decreases of 17.8%, 9.3% and 7.6% of baseline activity for chymotrypsin, trypsin and thrombin A, respectively). In addition, 21 at 30 μM had no noteworthy binding to the hERG channel, decreasing binding of the tracer by 8.34%. The results suggest favorable antibacterial selectivity, low potential drug-drug interactions, and minimal cardiac channel-mediated toxicity.
Table 3.
Safety and selectivity of boro-PBPi 21
| In vitro ADMET assay | Boro-PBPi 21 results |
|---|---|
| Solubility (formulated) | 75 mg/mL in PBS adjusted to pH 6.5 – 7 |
| Plasma protein binding (% bound) | 39% (human), 19% (mouse) |
| Hemolysis | >1,000 μg/mL |
| Cytotoxicity | CC50 >256 μg/mL (HeLa, MRC-5, 3T3) |
| Mitochondrial toxicity | No toxicity observed at 100 μM |
| CYP inhibition | IC50 >30 μM for 8 CYP enzymes (~35% inhibition of CYP2C19 observed) |
| Micronucleus assay (mammalian chromosomal aberration assay) | No micronucleus observed up to 300 μM |
| hERG cardiac channel assay | No inhibition at 30 μM |
| Protease inhibition | Negligible inhibition of 3 human proteases at 30 μM (18% inhibition for chymotrypsin observed) |
A small-molecule therapeutic agent to treat N. gonorrhoeae infections is recommended to be oral or intramuscular administration within the outpatient setting59. Several boro-PBPi were evaluated in the Caco-2 monolayer assay to measure the potential for human gastrointestinal tract permeability60. None of the boro-PBPi tested, including several ester prodrugs, demonstrated sufficient passage across Caco-2 cell monolayers, indicating that oral administration of these boro-PBPi is impractical (data not shown). We therefore evaluated the potential for IM administration of 21 in male Sprague Dawley rats (Figure S12). Based on the pharmacokinetic results, the IM bioavailability (FIM) of 21 was estimated to be at least 66%, supporting the potential for IM administration of 21 for the treatment of gonorrhea.
DISCUSSION
Boron-containing compounds have become increasingly attractive in drug discovery because of their unique binding properties to biological targets, their favorable pharmacokinetic properties, and low overall toxicity61. Here, we describe the identification of 21 (VNRX-14079), a novel non-β-lactam benzoxaborinine PBPi, that binds to both non-mosaic and mosaic PBP2 and exhibits potent standalone in vitro antibacterial activity and in vivo efficacy against CRO-resistant N. gonorrhoeae, as well as a favorable safety, antibacterial selectivity, and pharmacokinetic profile for intramuscular administration.
During the discovery phase of this program, we first identified 2, a prototype boro-PBPi containing the ureido side chain of cefoperazone. Notably, 2 showed weaker antibacterial activity than cefoperazone against multiple N. gonorrhoeae strains. This reduced activity may be a consequence of the reversible nature of the covalent bond between 2 and the catalytic Ser310 of PBP2, whereas cefoperazone forms an irreversible acyl-enzyme complex with PBP2 that requires a separate hydrolysis step to regenerate the enzyme. We subsequently identified benzoate-containing 12 and aryl phosphonate-containing 15 as well as 18 with increased interactions to PBP2 and antibacterial activity. The preference of the phosphonate-containing boro-PBPi is partly related to the ability of the terminal phosphonate residue to interact with PBP2 residues Arg502, His514 and Tyr543, further stabilizing the boro-PBPi/PBP2 complex relative to 2. X-ray crystallographic analysis of boro-PBPi/tPBP2 complexes indicates that, following binding of the benzoxaborinines 12 and 15, the β3-β4 loop occupies an inward conformation closer to the active site of mosaic tPBP235/02. Inward movement of the β3-β4 loop is postulated to promote efficient acylation of β-lactams to PBP248, 62. The importance of β3-β4 loop movement for potent boro-PBPi activity is also supported by the observation that Ala501 mutations — known to cause ordering of the β3-β4 loop54 — reduced boro-PBPi activity in the WHO L (A501V mutation) and F89 (A501P mutation) strains. Importantly, antibacterial activities of these ureido boro-PBPi were less impacted by altered PBP2 variants (e.g., mosaic PBP2) than oxyimino-acetamide boro-PBPi (e.g., compound 1), which is consistent with the observation that ureidopenicillins and ureidocephalosporins are more potent inhibitors of mosaic PBP2 than the oxyimino-acetamide cephalosporin CRO48.
Although the acquisition of an Ala501 mutation in PBP2 (i.e., A501P in F89) might lead to boro-PBPi resistance, recent studies have shown that the A501P mutation is associated with reduced biological fitness in vitro and in vivo57. Not surprisingly, recurrence of the A501P mutation of PBP2 has not been reported for over a decade. By contrast, the PBP2 variant of highest concern is penA-60, which has been observed globally in many CRO-resistant clinical isolates. This variant is not mutated at position 501 and overall contains similar mutations as PBP2 from H041. Unlike the PBP2 A501P variant, only minor fitness defects are observed for penA-60-possessing strains, which have likely contributed to the rapid spread of these CRO-resistant strains63. The MIC of 21 against WHO Q, which carries the penA-60 allele, was 8-fold lower than CRO (MIC of 0.06 μg/mL for 21, relative to 0.5 μg/mL for CRO). In addition, like CRO, 21 showed a low frequency of resistance (<4.17 × 10−9) and rapid cell killing at 4× MIC in WHO Q. Thus, 21 is active against CRO-resistant N. gonorrhoeae strains carrying the penA-60 allele, while retaining favorable drug-like properties akin to CRO.
Overall, boro-PBPi can exhibit potent standalone antibacterial activity, good safety and selectivity profiles, and excellent in vivo efficacy. 21 is a promising new agent to treat N. gonorrhoeae infections including CRO-resistant infections. Return on investment and other financial barriers imposes a real-world limitation to development of novel antibacterial agents such as boro-PBPi 21. An unfortunate consequence of the current antibiotic market that prevents further advancement of highly needed new antibacterial drugs64. Despite the current market situation, 21 is an outstanding candidate that could replace CRO for outpatient treatment of gonorrhea.
METHODS
Compounds and chemical synthesis
Boro-PBPi compounds were synthesized at Venatorx Pharmaceuticals, Inc. and BioDuro-Sundia as described in Supplementary Information. Ceftriaxone and azithromycin were purchased from USP. Ciprofloxacin was purchased from Sigma. Cefoperazone was purchased from Alfa Aesar. Zoliflodacin and gepotidacin were purchased from MedChemExpress. Molecular Operating Environment (MOE, Chemical Computing Group, Montreal, Canada) was used for computational chemistry and inhibitor design.
Bacterial strains and growth conditions
The strains used are listed in Table S13. FA1090, H041 (WHO X), MS11, and F89 were obtained from Dr. Ann Jerse (Uniformed Services University of the Health Sciences). The N. gonorrhoeae WHO reference strains (WHO F, WHO G, WHO L, WHO M, WHO K, WHO L, WHO Y, and WHO Z)65 were obtained from the CDC. WHO Q (G7944, NCTC 14208)66 was obtained from the National Collection of Type Cultures. FA19 was obtained from Dr. Robert A. Nicholas (University of North Carolina). A set of 50 CDC strains (AR bank #165–214) were obtained from the CDC, of which 6 strains were confirmed not to be N. gonorrhoeae and excluded from the study. All N. gonorrhoeae strains were grown at 37 °C in a 5% CO2 humidified environment.
Minimum inhibitory concentration (MIC) assays
To determine the ability of test compounds to inhibit the growth of bacterial strains, MIC assays were performed using both liquid- and agar-based microdilution methods as described previously with modifications52, 67, 68. Briefly, cryo-preserved bacterial cultures of clinical strains were streaked for isolation on Chocolate Agar [72 g/L (2×) GC Agar Base (Remel #R453502) and 2% (2×) hemoglobin (Remel #R451402) was autoclaved separately at 121 °C for 20 min to sterilize. Once cooled to ~50 °C, the 2× GC Agar Base and 2× hemoglobin solutions were combined at a 1:1 volume ratio, and 1% IsoVitaleX™ Enrichment (BD # 211876) was added to the solution] and grown for ~24 h. Freshly grown colonies were used for inoculum preparation. In the liquid broth-based MIC assay, 2-fold serial dilutions of test compounds were conducted in a 96-well plate with a final volume of 75 μL per well at 2-fold the final desired concentration in Fastidious broth (FB, prepared according to Cartwright et al. with an adjustment in final pH to 7.2 ± 0.2)67. For preparation of assay inoculum, a direct suspension was prepared by aseptically swabbing 10–15 colonies from agar plates into culture tubes containing 2 mL of fresh sterile saline. After the dilution plates were set up, direct suspensions were then diluted in a cuvette containing sterile saline and the optical density was measured at 600 nm (OD600). Direct suspensions were diluted in FB to make assay inocula (~1 × 106 CFU/mL) and 75 μL of assay inocula was added to the 96-well plates prepared with compound dilutions, yielding a starting bacterial concentration at 5 × 105 CFU/mL. The plates were incubated for ~24 h. The broth MIC was read visually as the lowest concentration well that completely inhibited bacterial growth.
In the agar-based method, GC agar was used to determine MIC. After the initial strain incubation for ~24 h, bacterial colonies were re-streaked for isolation on fresh Chocolate Agar and grown for 18–24 h for use in the assay. To prepare GC agar, GC agar base (Remel #R453502) dissolved in water was autoclaved at 121°C to sterilize, and once cooled to ~50°C, it was supplemented with IsoVitaleX™ Enrichment (BD # 211876) and 1M filter-sterilized ferric nitrate (Fisher #S25320) to achieve 1% and 12 μM final concentrations, respectively. The determined amount of test compound stock solution was pipetted directly to a 100 × 15 mm petri dish for each desired test concentration and 20 mL of the prepared GC agar (before solidification) was added to the petri dish containing test compound. Agar plates were swirled to dissolve compound and vented for solidification in the biological safety cabinet (BSC) until dry. For preparation of assay inoculum, a direct suspension was made by aseptically swabbing several colonies from the agar plates and suspending them into 2 mL of sterile cation-adjusted Mueller Hinton broth (CAMHB). Direct suspensions were then diluted with sterile CAMHB and adjusted to an OD600 of 0.1 (0.5 McFarland standard equivalent) to prepare the inocula. The Steer’s replicator was used to plate up to 32 spots of 2 μL inocula on a single agar plate from a separate 32-well plate containing inocula (500 μL each). The agar plates were vented in the BSC until dry, inverted, and incubated for 24 h. The agar MIC was read as the lowest concentration that completely inhibits bacterial growth.
Time-kill assay
The time-kill assay for N. gonorrhoeae was performed as described69 with the following modifications. The bacterial inoculum for the assays was prepared by suspending colonies of N. gonorrhoeae grown on Chocolate Agar for ~24 h in sterile saline. The cell suspension was adjusted to an OD600 of 0.1 and was diluted 1:500 in FB (final cell density ~1 × 105 colony forming units (CFU)/mL, 90 μL each) in a 96-well plate, which was then incubated with shaking at 200 RPM for 4 h to allow the bacteria to reach log phase of growth. After the initial growth phase, 10 μL of appropriate 10× drug dilutions (4× and 16× MIC) were added to each desired well for a total volume of 100 μL. As an untreated control, 10 μL of FB was added. At time −4 h (time of inoculation), 0 h (time of compound addition), 2 h, 4 h, 6 h, and 24 h samples were taken and CFU/mL in each sample was measured. Growth curves were analyzed by plotting the log10CFU/mL versus time using GraphPad Prism version 10.3. 2.
Frequency of resistance (FoR)
N. gonorrhoeae strains (ATCC 49226, H041, and WHO Q) were grown for ~24 h on Chocolate Agar and colonies were suspended in phosphate buffered saline (PBS) to make a cell suspension at ~5 × 108 CFU/mL. A 0.1 mL aliquot of the cell suspension was spread on 10 GC agar plates (100 mm diameter) supplemented with 1% IsoVitaleX™, 12 μM ferric nitrate and test compounds (4× and 16× MIC) for ATCC 49226 and H041. Chocolate Agar was used for WHO Q because the strain grew poorly on GC agar. The viable cell count in each suspension was determined by plating serial 10-fold dilutions onto the corresponding agar. The plates were incubated for 24 h and the number of colonies that arose were counted visually and the spontaneous mutation frequency was calculated.
Isolation of zoliflodacin-resistant mutants and mutation identification
Zoliflodacin-resistant mutants were isolated by selection of WHO K (MIC = 0.12 μg/mL) on GC agar supplemented with 4× MIC of zoliflodacin. Colonies arising on agar were streaked on Chocolate Agar and a single colony was tested with broth MIC to confirm elevated MIC of zoliflodacin. Short-read genome-sequencing was performed on parent and isolated mutants (GENEWIZ, South Plainfield, NJ, USA). Genome analysis from FASTQ files provided by GENEWIZ was performed using Geneious Prime version 2022.1.1 (Biomatters Inc., San Diego, CA, USA). The reads were trimmed with BBDuK Adapter/Quality Trimming Version 38.84 (Brian Bushnell), yielding ~10 million reads. Read mapping was performed with the Geneious Mapper using the WHO K reference genome (Genbank ID: GCF_900087865.2) to give >100 mean coverage of the entire chromosome followed by Geneious SNP analysis, resulting in the identification of the GyrB(D429N) mutation present in each mutant.
Expression and purification of N. gonorrhoeae PBP2
In the N. gonorrhoeae PBP2 inhibition assay, constructs comprising the transpeptidase (TP) domain of PBP2 from the FA19 and H041 strains (tPBP2FA19 and tPBP2H041) were used. The cloning, expression, and purification of these PBP2 constructs was described previously46, 70. Importantly, these TP domains of PBP2 are acylated by Bocillin™-FL at the same rate as for full-length PBP270.
Measurement of in vitro inhibition of N. gonorrhoeae PBP2
The inhibitory potency towards wild-type PBP2FA19 was assessed by determining the inhibitor concentration that was required to reduce PBP2FA19 binding to fluorescently labelled penicillin V, Bocillin™-FL (Thermo Fisher Scientific) by 50% (IC50), using a fluorescence polarization (FP) competitive equilibrium binding assay71. To establish assay conditions for competition binding, enzyme titration/saturation binding experiments were initially performed. Bocillin™-FL was prepared at 0.2 μM in assay buffer (50 mM Hepes-NaOH pH 8.0), 300 mM NaCl and 10% (v/v) glycerol), and saturation binding performed by mixing 40 μL of PBP2 ranging in concentrations from 0–24 μM with 40 μL of the 0.2 μM Bocillin™-FL solution, in individual wells of black 384-well microplates. Immediately upon mixing, fluorescence was monitored using a Cytation™ 3 plate reader (BioTek) with an FP cube containing polarizing filters with 485 nm excitation and 520 nm emission. The instrument gain was set to achieve minimum fluorescence values of 50–60 and each measurement was the average of 3 flashes. Fluorescence was measured continuously for up to 120 min, and the response was stabilized within 10 min, with a dose-dependence on PBP2FA19 concentration. For competition binding assays, two-fold serial dilutions of compounds were prepared in assay buffer and mixed with Bocillin™-FL (final concentration of 0.1 μM) in black 384-well microplates. PBP2FA19 was added to achieve a final concentration of 0.25 μM and fluorescence was immediately measured at 1-min intervals for up to 10 min. Fluorescence values at the reaction endpoint (typically within 8 min) were normalized to the maximal response, then plotted against inhibitor concentration and the data fit to a four-parameter inhibitor-response curve to derive the half-maximal inhibitory concentration, IC50.
The interaction between PBP2H041 and Bocillin™-FL did not produce an adequate response in the FP assay. Therefore, inhibitory potency towards tPBP2H041 was assessed by determining the concentration of compound required to inhibit the binding of Bocillin™-FL to PBP2H041 by 50% (IC50) with an SDS-PAGE-based competition binding assay. Two-fold serial dilutions of each compound were prepared in assay buffer and mixed with 1 μM tPBP2H041, then incubated at ambient temperature for 60 min. Bocillin™-FL (1 μM) was added, and reaction mixtures were further incubated for 60 min before resolution by SDS-PAGE using 10% NuPAGE Bis-Tris Mini Protein Gels (Invitrogen). The amount of Bocillin™-FL incorporated into tPBP2H041 at each inhibitor concentration was detected by fluorescence scanning of PAGE gels with an Azure 600 Imager (Azure Biosystems, Inc.) and quantitated using ImageJ software72. Data were normalized to the maximum fluorescence, then plotted against inhibitor concentration and fit to a four-parameter inhibitor-response curve to derive the half-maximal inhibitory concentration, IC50.
X-ray crystallography
The crystal structure of the transpeptidase domain of PBP2 derived from the cephalosporin reduced susceptibility strain 35/02 of N. gonorrhoeae (tPBP235/02) has been reported previously47. The protein was purified and concentrated to 13 mg/mL in Tris-HCl, pH 7.8, with 10% glycerol and 500 mM NaCl and then crystallized in the same way as reported previously. These crystals occupy the P212121 space group with cell dimensions a=50.5, b=61.2, and c=108.8 Å and one molecule in the asymmetric unit.
Complexes of tPBP235/02 with R,R-12 and R,R-15 were generated by soaking crystals with 1 μL of 10 mM boro-PBPi dissolved in PBS for 4 h, followed by flash-freezing in liquid nitrogen. To obtain the tPBP235/02/R,R-15 complex structure, a mixture of the S,R- and R,R-diastereomers was used for soaking and only R,R-15 in the electron density was observed. Diffraction data were collected at a wavelength of 1.00 Å on an Eiger-16M detector at the SER-CAT 22-ID beamline at the Advanced Photon Source in Argonne, IL, USA. 200° of data were collected in 0.25° oscillations with an exposure time of 0.2 s/frame and a crystal-to-plate distance of 200 mm. Data were processed using HKL200073 and structures solved by refinement against the apo structure of tPBP235/02. Bound inhibitors were modeled into the |Fo|-|Fc| difference electron density map, followed by iterative cycles of model building and refinement using COOT 0.9.8.9574 and PHENIX 1.18.2–387475. The crystallographic data collection and model refinement statistics are found in Table S14.
Pharmacokinetic analysis of boro-PBPi 18 in mice
18 was formulated at the maximum concentration of 100 mg/mL in PBS with pH adjusted between 6 and 7 by NaOH. The pharmacokinetic study was conducted at the BioDuro-Sundia DMPK group (Jiangsu, China) using female BALB/c mice (7–9 weeks old). 18 was administered once intravenously at 3 mg/kg or subcutaneously at 10, 30, 100, 300 and 1,000 mg/kg (3 mice per group). Blood microsamples (30 μL each) were collected at the timepoints of 0.0083, 0.25, 0.5, 1, 2, 4, 8 and 24 h post-dose. The plasma of each blood sample was prepared using K2EDTA and centrifugation. The concentration of boro-PBPi in the plasma was measured using the LC-MS/MS system with a Waters™ Acquity Ultra Performance LC and a Sciex API 6500 controlled by Analyst Software 1.7.1. Each plasma sample was diluted in 5% trichloroacetic acid in water followed by centrifugation. The supernatant was diluted 2-fold with 50% acetonitrile and loaded onto an Avantor® ACE 5 C4 column (50 mm × 2.1 mm). The mobile phase (MP) A was 5 mM ammonium acetate and 0.05% formic acid in water and MP B was 0.1% formic acid in acetonitrile. 18 was eluted in a linear gradient from 5% to 95% MP B over 1.2 min at a 0.6 mL/min flow rate and the elution was subjected to the MS/MS analysis using multiple reaction monitoring with a positive ion mode at 611.10/429.00. Buspirone at 5 ng/mL and tolbutamide at 50 ng/mL in 5% trichloroacetic acid were used as an internal standard. The lower limit of detection for 18 was 2 ng/mL.
Pharmacokinetic analysis of boro-PBPi 21 in mice
21 was formulated at the maximum concentration at 30 mg/mL in PBS with pH adjusted between 6 and 7 by NaOH. The pharmacokinetic study was conducted at the BioDuro-Sundia DMPK group using CD-1 mice (7–9 weeks old). The pharmacokinetic study of 21 was conducted similarly to that of 18. The bioanalytical method to quantify 21 was the LC-MS/MS system with a SCIEX AD system and a SCIEX Triple Quad 7500 controlled by SCIEX OS 2.0.0.45330 software. Each sample prepared from plasma was loaded on to an ACQUITY UPLC® Peptide BEH C18 column (1.7 μm, 300 Å, 50 mm × 2.1 mm). The MP A was 1% formic acid in water and MP B is 1% formic acid in acetonitrile. 21 was eluted at a 0.5 mL/min flow rate in linear gradients from 5% to 55% of MP B over 0.1 min and to 95% of MP B over 0.8 min, and the elution was subjected to the MS/MS analysis using multiple reaction monitoring with a positive ion mode at 647.4/465.0. Tolbutamide at 0.5 ng/mL in 5% trichloroacetic acid was used as an internal standard. The lower limit of detection for 21 was 2 ng/mL.
The parameters derived from non-compartmental PK analysis were the plasma concentration at time 0 following IV administration (C0), plasma half-life (t1/2), maximum plasma concentration (Cmax), the area under the concentration vs. time curve from t = 0 to infinity (AUCinf), volume of distribution at steady-state (VSS), clearance (CL), time to maximum plasma concentration (Tmax), and bioavailability from the subcutaneous dose (FSC). In each of these instances, variability was related to concentrations below the lower limit of quantification (BLOQ) which restricted characterization of the terminal phase in at least one mouse per group (Table S3).
Efficacy assessment of boro-PBPi 18 in the FA1090 vaginal infection model
Efficacy analysis was performed by Eurofins Pharmacology Discovery Services (Taipei, Taiwan) through the NIAID preclinical services, using ovariectomized and 17β-estradiol-treated female BALB/c mice76. The facility and procedures have been accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International and assured from the Office of Laboratory Animal Welfare. The protocol was approved by the Institutional Animal Care and Use Committee. Groups of five immunocompetent female ovariectomized BALB/c mice aged 5–6 weeks were used for the efficacy study. Animals were received from BioLASCO Taiwan and acclimated for three days. After acclimation, an ovariectomy was performed on animals aged 4 weeks. The period of surgical recovery and acclimation was at least 7 days. Animals were treated with subcutaneous injection of meloxicam (20 mg/kg) if signs of pain or distress are observed during this period. Prior to infection, animals were subcutaneously injected with estradiol solution at 0.23 mg/mouse 2 days before infection (day −2) and on the day of infection (day 0). To minimize the indigenous vaginal bacteria, animals were treated twice daily (BID) with streptomycin (1.2 mg/mouse) and vancomycin (0.6 mg/mouse) by intraperitoneal injection along with trimethoprim sulfate at 0.4 mg/mL supplied in the drinking water. These antibiotic treatments started two days prior to infection and continued daily until the end of study. On day 0, animals were inoculated intravaginally with N. gonorrhoeae FA1090 (ATCC 700825, 0.02 mL [2.56 × 106 CFU] per mouse) under anesthesia by intraperitoneal injection of pentobarbital (80 mg/kg), followed by rinsing the vagina with 50 mM Hepes (pH 7.4, 30 μL). 18 was administered subcutaneously twice-a-day with a 12 h interval (q12h) starting at 2 h post-infection for one day (5 mice per group). At 2 or 26 h after infection, animals were sacrificed with CO2 asphyxiation for harvest of vaginal lavage fluid. For the vaginal infected animals, vaginal lavage was performed twice with 200 μL GC broth containing 0.05% saponin to recover vaginal bacteria. The lavage samples from each animal were pooled in a total volume of 500 μL. Bacterial burden in the lavage fluids was determined by performing 10-fold serial dilutions and plating 0.1 mL of each onto Chocolate agar plates. The colony forming unit value (CFU) per mL lavage fluid (CFU/mL) was calculated.
Efficacy assessment of boro-PBPi 21 in the H041 vaginal infection model
In vivo efficacy of 21 was tested in female NCI BALB/c mice as described previously51. Using a simulation based on the mouse plasma protein binding (19%) and the PK profile in male CD-1 mice, four dosing regimens for 21 were selected to vary %fT>MIC from 20% to 100% in this efficacy study. Mice in diestrus or anestrus stage of estrous cycle were randomized into 6 groups and pretreated with 17-β estradiol as well as antibiotics (streptomycin and trimethoprim) to suppress the overgrowth of the commensal microbiota and to support N. gonorrhoeae colonization. The pretreated mice were vaginally infected with H041 (1–2 × 104 CFU/mouse) two days before treating the mice with 21 via subcutaneous injection, either once or alternate dosing regimens. Control groups were given ceftriaxone (CRO, 120 mg/kg SC, TID) or the vehicle control (PBS). Vaginal mucus was quantitatively cultured for N. gonorrhoeae for eight consecutive days post-treatment. A portion of the swab was also inoculated onto heart infusion agar (HIA) to monitor the presence of facultative aerobic commensal microbiota. Vaginal polymorphonuclear leukocyte (PMN) influx was assessed on each culture day by cytological examination of stained vaginal smears and reported as the percentage of PMNs among 100 vaginal cells. Efficacy was measured by comparing differences in the clearance rate and average bacterial burden over 8 days following treatment with 21, CRO, or the compound vehicle PBS.
In vitro safety and selectivity assays for boro-PBPi 21
The assays that examined plasma protein binding, cytotoxicity, hemolysis, mitochondrial toxicity, and chromosomal aberrations (micronucleus assay) as well as several in vitro inhibition assays (CYP450s and human proteases) or binding (hERG) assays for 21 were performed using established methods and controls that behaved as expected. The detailed description of the methods and raw data are found in Table S12.
Pharmacokinetic analysis of boro-PBPi 21 in Sprague Dawley rats
The PK study was performed in male Sprague Dawley (SD) rats after a single intravenous (IV) or intramuscular (IM) dose at QPS, LLC (Newark, DE). All dosing formulations of 21 were clear freshly prepared solutions. For IM dose preparation, 21 was dissolved in vehicle (45:55 vol/vol 1N NaOH:1.6× PBS) with vortex, sonication and stirring to achieve 75 mg/mL at pH ~6.8. A 0.6 mg/mL IV dose formulation was prepared by diluting the IM formulation with 1× PBS, pH 7.4 (final pH ~7.1) and filtered through a 0.22 μm filter (Millex®-GV). Male Sprague Dawley rats (weight ~270 g) were administered 21 via IM and IV routes. Blood samples (0.3 – 0.4 mL) were collected via tail vein at pre-dose (0), 0.083, 0.25, 0.5, 2, 4, 8 and 24 h post-dose from IV-dosed rats (n = 3) and pre-dose (0), 0.5, 1, 2, 4, 8 and 24 h pose-dose from IM-dosed rats (n = 3). All blood samples were collected into tubes containing K2EDTA on wet ice until centrifuged at 4°C, for 3 min, at 6,000 RPM (~3,800 × g) within 40 min of blood collection. Terminal blood samples were collected in 10 mL Vacutainer tubes and centrifuged at 4,000 RPM (~3,300 × g) for 15 min at 4°C. All plasma samples were snap frozen on dry ice and stored at approximately −70°C until bioanalysis to determine the concentration of 21 in plasma. The pharmacokinetic data (Figure S12) were evaluated using noncompartmental analysis (Phoenix® WinNonlin®, version 8.3, Certara USA Inc., Princeton, New Jersey) to determine IM bioavailability (FIM) in rats. Average concentrations from three rats per dose group were included. Following 3 mg/kg IV administration of 21 in rats, terminal half-life, R-squared, and adjusted R-squared values were 0.63 h, 0.98, and 0.98, respectively, while 30 mg/kg IM administration provided t½ = 0.95 h with R2 = 0.93, and adjusted R2 = 0.89. The FIM was calculated by dividing the dose-normalized area under the concentration-time curve (AUC) extrapolated to infinity based on the last observed concentration (AUCINF_D_obs in h*kg*ng/mL/mg) when administered IM by the AUCINF_D_obs when administered IV (1030.81/1558.40 = 66%).
Supplementary Material
Acknowledgements
We thank Robert Nicholas, Eric Brown, and Jean-Denis Docquier for providing strains. BioDuro-Sundia, Eurofins Panlabs Discovery Services Taiwan, Ltd., and QPS were the contract research organizations that supported this work, including compound synthesis, mouse PK, DMPK, in vivo efficacy, and rat PK. We also thank Michael Barbachyn, Alice Erwin, and Su Chiang (CARB-X) for supportive discussion, and our current and former colleagues at Venatorx for preliminary data generation and helpful discussions. Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Award Number R01 AI141239 and powered by CARB-X under Research Subaward Agreement Number 4500003206. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Diffraction data were collected at Southeast Regional Collaborative Access Team (SER-CAT) 22-ID beamline at the Advanced Photon Source, Argonne National Laboratory. SER-CAT is supported by its member institutions (https://www3.ser.aps.anl.gov/contact-us#TITLE_SER_CAT_Memberships), equipment grants (S10_RR25528, S10_RR028976 and S10_OD027000) from the National Institutes of Health, and funding from the Georgia Research Alliance. This research used resources of the Advanced Photon Source, a U.S. Department of Energy (DOE) Office of Science user facility operated for the DOE Office of Science by Argonne National Laboratory under Contract No. DE-AC02-06CH11357.
Footnotes
Competing interests
Authors T.U., A.L.Z., B.M., L.M.A., S.A.B., C.L.C., G.H.C., A.S.D., M.E., S.G.E.H., C.L.M., G.R., A.S., K.U., F.Y., D.C.P, C.J.B., D.M.D., and S.M.C. are current or former employees of Venatorx Pharmaceuticals, Inc. T.U., A.L.Z., S.A.B., C.L.C., G.H.C., C.L.M., D.C.P., C.J.B., D.M.D., and S.M.C. are co-inventors on a patent application covering molecules described in this manuscript.
Data availability
The data that support this study are included in this published article and its supplementary information or are available from the corresponding authors on reasonable request. Structural data have been deposited to the PDB with the accession codes of 9MD0 (tPBP235/02-12) and 9MCZ (tPBP235/02-15).
REFERENCES
- 1.Klein EY, et al. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc Natl Acad Sci U S A 115, E3463–E3470 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bush K. Past and Present Perspectives on beta-Lactamases. Antimicrob Agents Chemother 62, e01076–01018 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Naas T, et al. Beta-lactamase database (BLDB) - structure and function. J Enzyme Inhib Med Chem 32, 917–919 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bonomo RA. beta-Lactamases: A Focus on Current Challenges. Cold Spring Harbor perspectives in medicine 7, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bertonha AF, Silva CCL, Shirakawa KT, Trindade DM, Dessen A. Penicillin-binding protein (PBP) inhibitor development: A 10-year chemical perspective. Exp Biol Med (Maywood) 248, 1657–1670 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Papp-Wallace KM, Mack AR, Taracila MA, Bonomo RA. Resistance to novel beta-Lactam-beta-Lactamase inhibitor combinations: The “Price of Progress”. Infect Dis Clin North Am 34, 773–819 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Durand-Reville TF, et al. Rational design of a new antibiotic class for drug-resistant infections. Nature 597, 698–702 (2021). [DOI] [PubMed] [Google Scholar]
- 8.(WHO) WHO. Gonorrhoea (Neisseria gonorrhoeae infection). (2023).
- 9.(WHO) WHO. Multi-drug resistant gonorrhoea. (2023).
- 10.McKenna M. Can non-profits beat antibiotic resistance and soaring drug costs? Nature 626, 942–944 (2024).38418910 [Google Scholar]
- 11.Wi T, et al. Antimicrobial resistance in Neisseria gonorrhoeae: Global surveillance and a call for international collaborative action. PLoS Med 14, e1002344 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hook EW 3rd, Kirkcaldy RD. A brief history of evolving diagnostics and therapy for gonorrhea: lessons learned. Clin Infect Dis 67, 1294–1299 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Unemo M, Seifert HS, Hook EW 3rd, Hawkes S, Ndowa F, Dillon JR. Gonorrhoea. Nat Rev Dis Primers 5, 79 (2019). [DOI] [PubMed] [Google Scholar]
- 14.Frost KM, et al. Rolling the evolutionary dice: Neisseria commensals as proxies for elucidating the underpinnings of antibiotic resistance mechanisms and evolution in human pathogens. Microbiol Spectr 12, e0350723 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.CDC. Antibiotic resistance threats in the United States. U.S. Department of Health and Human Services, CDC, Atlanta, GA., (2019). [Google Scholar]
- 16.Tacconelli E, et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 18, 318–327 (2018). [DOI] [PubMed] [Google Scholar]
- 17.(WHO) WHO. WHO bacterial priority pathogens list, 2024: Bacterial pathogens of public health importance to guide research, development and strategies to prevent and control antimicrobial resistance.) (2024). [DOI] [PubMed]
- 18.St. Cyr S, et al. Update to CDC’s treatment guidelines for gonococcal infection. MMWR Morb Mortal Wkly Rep 69, 1911–1916 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ouk V, Pham CD, Wi T, van Hal SJ, Lahra MM, group ECw. The enhanced gonococcal surveillance programme, Cambodia. Lancet Infect Dis 23, e332–e333 (2023). [DOI] [PubMed] [Google Scholar]
- 20.Jensen JS, Unemo M. Antimicrobial treatment and resistance in sexually transmitted bacterial infections. Nat Rev Microbiol, (2024). [DOI] [PubMed] [Google Scholar]
- 21.Jerse AE, Bash MC, Russell MW. Vaccines against gonorrhea: current status and future challenges. Vaccine 32, 1579–1587 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Reimche JL, et al. Novel strain of multidrug non-susceptible Neisseria gonorrhoeae in the USA. Lancet Infect Dis 24, e149–e151 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lin X, et al. Dissemination and genome analysis of high-level ceftriaxone-resistant penA 60.001 Neisseria gonorrhoeae strains from the Guangdong Gonococcal antibiotics susceptibility Programme (GD-GASP), 2016–2019. Emerg Microbes Infect 11, 344–350 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lee K, et al. Clonal expansion and spread of the ceftriaxone-resistant Neisseria gonorrhoeae strain FC428, identified in Japan in 2015, and closely related isolates. The Journal of antimicrobial chemotherapy 74, 1812–1819 (2019). [DOI] [PubMed] [Google Scholar]
- 25.Lahra MM, et al. Cooperative Recognition of Internationally Disseminated Ceftriaxone-Resistant Neisseria gonorrhoeae Strain. Emerg Infect Dis 24, 735–740 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nakayama S, Shimuta K, Furubayashi K, Kawahata T, Unemo M, Ohnishi M. New ceftriaxone- and multidrug-resistant Neisseria gonorrhoeae strain with a novel mosaic penA gene isolated in Japan. Antimicrob Agents Chemother 60, 4339–4341 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Picker MA, et al. Notes from the field: first case in the united states of Neisseria gonorrhoeae harboring emerging mosaic penA60 allele, conferring reduced susceptibility to cefixime and ceftriaxone. MMWR Morb Mortal Wkly Rep 69, 1876–1877 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mlynarczyk-Bonikowska B, Majewska A, Malejczyk M, Mlynarczyk G, Majewski S. Multiresistant Neisseria gonorrhoeae: a new threat in second decade of the XXI century. Med Microbiol Immunol 209, 95–108 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kandinov I, et al. In vitro susceptibility to beta-lactam antibiotics and viability of Neisseria gonorrhoeae strains producing plasmid-mediated broad- and extended-spectrum beta-lactamases. Frontiers in microbiology 13, 896607 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hassain AS, Abdulrahman TR, Hassan JS. Occurrence of CTX-M I gene in beta lactam resistance Neisseria gonorrhoeae isolated form women with endocervical infection. J Popl Ther Clin Pharmacol 30, 49–53 (2023). [Google Scholar]
- 31.Callaway E. ‘Groundbreaking’: first treatment targeting ‘super-gonorrhoea’ passes trial. Nature 623, 236 (2023). [DOI] [PubMed] [Google Scholar]
- 32.Mullard A. GSK’s first-in-class antibiotic secures another phase III win, approaches regulatory run. Nature reviews Drug discovery, (2024). [DOI] [PubMed] [Google Scholar]
- 33.(WHO) WHO. 2021 Antibacterial agents in clinical and preclinical development: an overview and analysis. (2022).
- 34.Scangarella-Oman NE, et al. Microbiological analysis from a phase 2 randomized study in adults evaluating single oral doses of gepotidacin in the treatment of uncomplicated urogenital gonorrhea caused by Neisseria gonorrhoeae. Antimicrob Agents Chemother 62, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rubin DH, Mortimer TD, Grad YH. Neisseria gonorrhoeae diagnostic escape from a gyrA-based test for ciprofloxacin susceptibility and the effect on zoliflodacin resistance: a bacterial genetics and experimental evolution study. Lancet Microbe 4, e247–e254 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Abdellati S, et al. Gonococcal resistance to zoliflodacin could emerge via transformation from commensal Neisseria species. An in-vitro transformation study. Sci Rep 14, 1179 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.de Vries HJC, et al. Efficacy of ertapenem, gentamicin, fosfomycin, and ceftriaxone for the treatment of anogenital gonorrhoea (NABOGO): a randomised, non-inferiority trial. Lancet Infect Dis 22, 706–717 (2022). [DOI] [PubMed] [Google Scholar]
- 38.Hamrick JC, et al. VNRX-5133 (Taniborbactam), a broad-spectrum inhibitor of serine- and metallo-beta-lactamases, restores activity of cefepime in Enterobacterales and Pseudomonas aeruginosa. Antimicrob Agents Chemother 64, e01963–01919 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Liu B, et al. Discovery of Taniborbactam (VNRX-5133): A broad-spectrum serine- and metallo-beta-lactamase inhibitor for carbapenem-resistant bacterial infections. J Med Chem 63, 2789–2801 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chatwin CL, et al. Microbiological characterization of VNRX-5236, a broad-spectrum beta-lactamase inhibitor for rescue of the orally bioavailable cephalosporin ceftibuten as a carbapenem-sparing agent against strains of Enterobacterales expressing extended-spectrum beta-lactamases and serine carbapenemases. Antimicrob Agents Chemother 65, e0055221 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Trout RE, et al. Discovery of VNRX-7145 (VNRX-5236 etzadroxil): an orally bioavailable beta-lactamase inhibitor for enterobacterales expressing ambler class A, C, and D enzymes. J Med Chem 64, 10155–10166 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kumar V, Viviani SL, Ismail J, Agarwal S, Bonomo RA, van den Akker F. Structural analysis of the boronic acid beta-lactamase inhibitor vaborbactam binding to Pseudomonas aeruginosa penicillin-binding protein 3. PLoS One 16, e0258359 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Brem J, et al. Structural basis of metallo-beta-lactamase, serine-beta-lactamase and penicillin-binding protein inhibition by cyclic boronates. Nat Commun 7, 12406 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Krajnc A, et al. Bicyclic boronate VNRX-5133 inhibits metallo- and serine-beta-lactamases. J Med Chem 62, 8544–8556 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tomberg J, Unemo M, Ohnishi M, Davies C, Nicholas RA. Identification of amino acids conferring high-level resistance to expanded-spectrum cephalosporins in the penA gene from Neisseria gonorrhoeae strain H041. Antimicrob Agents Chemother 57, 3029–3036 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Singh A, Tomberg J, Nicholas RA, Davies C. Recognition of the beta-lactam carboxylate triggers acylation of Neisseria gonorrhoeae penicillin-binding protein 2. J Biol Chem 294, 14020–14032 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Singh A, et al. Mutations in penicillin-binding protein 2 from cephalosporin-resistant Neisseria gonorrhoeae hinder ceftriaxone acylation by restricting protein dynamics. J Biol Chem 295, 7529–7543 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Turner JM, Stratton CM, Bala S, Cardenas Alvarez M, Nicholas RA, Davies C. Ureidopenicillins are potent inhibitors of penicillin-binding protein 2 from multidrug-resistant Neisseria gonorrhoeae H041. ACS infectious diseases, (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jerse AE, Wu H, Packiam M, Vonck RA, Begum AA, Garvin LE. Estradiol-treated female mice as surrogate hosts for Neisseria gonorrhoeae genital tract infections. Frontiers in microbiology 2, 107 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hong LT, et al. International consensus recommendations for the use of prolonged-infusion beta-lactam antibiotics: endorsed by the American College of Clinical Pharmacy, British Society for Antimicrobial Chemotherapy, Cystic Fibrosis Foundation, European Society of Clinical Microbiology and Infectious Diseases, Infectious Diseases Society of America, Society of Critical Care Medicine, and Society of Infectious Diseases Pharmacists. Pharmacotherapy 43, 740–777 (2023). [DOI] [PubMed] [Google Scholar]
- 51.Connolly KL, Eakin AE, Gomez C, Osborn BL, Unemo M, Jerse AE. Pharmacokinetic data are predictive of in vivo efficacy for cefixime and ceftriaxone against susceptible and resistant Neisseria gonorrhoeae strains in the gonorrhea mouse model. Antimicrob Agents Chemother 63, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.CLSI. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically, 11th ed. Approved Guideline, CLSI document M07. Clinical and Laboratory Standards Institute, Wayne, PA. (2018). [Google Scholar]
- 53.Liao M, Gu WM, Yang Y, Dillon JA. Analysis of mutations in multiple loci of Neisseria gonorrhoeae isolates reveals effects of PIB, PBP2 and MtrR on reduced susceptibility to ceftriaxone. The Journal of antimicrobial chemotherapy 66, 1016–1023 (2011). [DOI] [PubMed] [Google Scholar]
- 54.Tomberg J, et al. Alanine 501 Mutations in penicillin-binding protein 2 from Neisseria gonorrhoeae: structure, mechanism, and effects on cephalosporin resistance and biological fitness. Biochemistry 56, 1140–1150 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Unemo M, Golparian D, Nicholas R, Ohnishi M, Gallay A, Sednaoui P. High-level cefixime- and ceftriaxone-resistant Neisseria gonorrhoeae in France: novel penA mosaic allele in a successful international clone causes treatment failure. Antimicrob Agents Chemother 56, 1273–1280 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Camara J, et al. Molecular characterization of two high-level ceftriaxone-resistant Neisseria gonorrhoeae isolates detected in Catalonia, Spain. The Journal of antimicrobial chemotherapy 67, 1858–1860 (2012). [DOI] [PubMed] [Google Scholar]
- 57.Vincent LR, et al. In Vivo-selected compensatory mutations restore the fitness cost of mosaic penA alleles that confer ceftriaxone resistance in Neisseria gonorrhoeae. mBio 9, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Liu H, Vidyaprakash E, Schmerer MW, Pham DC, St Cyr S, Kersh EN. A culture collection of 50 Neisseria gonorrhoeae isolates. Microbiol Resour Announc 9, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Organization WH. Target product profile for therapy of diagnosed uncomplicated gonorrhoea. World Health Organization, (2020). [Google Scholar]
- 60.van Breemen RB, Li Y. Caco-2 cell permeability assays to measure drug absorption. Expert Opin Drug Metab Toxicol 1, 175–185 (2005). [DOI] [PubMed] [Google Scholar]
- 61.Grams RJ, et al. The rise of boron-containing compounds: advancements in synthesis, medicinal chemistry, and emerging pharmacology. Chem Rev 124, 2441–2511 (2024). [DOI] [PubMed] [Google Scholar]
- 62.Fenton BA, Tomberg J, Sciandra CA, Nicholas RA, Davies C, Zhou P. Mutations in PBP2 from ceftriaxone-resistant Neisseria gonorrhoeae alter the dynamics of the beta3-beta4 loop to favor a low-affinity drug-binding state. J Biol Chem 297, 101188 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhou K, Chen SC, Yang F, van der Veen S, Yin YP. Impact of the gonococcal FC428 penA allele 60.001 on ceftriaxone resistance and biological fitness. Emerg Microbes Infect 9, 1219–1229 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Madden J, Outterson K. Trends in the global antibiotics market. Nature reviews Drug discovery 22, 174 (2023). [DOI] [PubMed] [Google Scholar]
- 65.Unemo M, et al. The novel 2016 WHO Neisseria gonorrhoeae reference strains for global quality assurance of laboratory investigations: phenotypic, genetic and reference genome characterization. The Journal of antimicrobial chemotherapy 71, 3096–3108 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Eyre DW, et al. Gonorrhoea treatment failure caused by a Neisseria gonorrhoeae strain with combined ceftriaxone and high-level azithromycin resistance, England, February 2018. Euro Surveill 23, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Cartwright CP, Stock F, Gill VJ. Improved enrichment broth for cultivation of fastidious organisms. J Clin Microbiol 32, 1825–1826 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.CLSI. Clinical Laboratory Standards Institute, Performance Standards for Antimicrobial Susceptibility Testing, 30st ed. CLSI; supplement M100. (2020). [Google Scholar]
- 69.Foerster S, Unemo M, Hathaway LJ, Low N, Althaus CL. Time-kill curve analysis and pharmacodynamic modelling for in vitro evaluation of antimicrobials against Neisseria gonorrhoeae. BMC Microbiol 16, 216 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Fedarovich A, Cook E, Tomberg J, Nicholas RA, Davies C. Structural effect of the Asp345a insertion in penicillin-binding protein 2 from penicillin-resistant strains of Neisseria gonorrhoeae. Biochemistry 53, 7596–7603 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Fedarovich A, Djordjevic KA, Swanson SM, Peterson YK, Nicholas RA, Davies C. High-throughput screening for novel inhibitors of Neisseria gonorrhoeae penicillin-binding protein 2. PLoS One 7, e44918 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods 9, 671–675 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Otwinowski Z, Minor W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol 276, 307–326 (1997). [DOI] [PubMed] [Google Scholar]
- 74.Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and development of Coot. Acta crystallographica Section D, Biological crystallography 66, 486–501 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Adams PD, et al. PHENIX: building new software for automated crystallographic structure determination. Acta crystallographica Section D, Biological crystallography 58, 1948–1954 (2002). [DOI] [PubMed] [Google Scholar]
- 76.Jerse AE. Experimental gonococcal genital tract infection and opacity protein expression in estradiol-treated mice. Infect Immun 67, 5699–5708 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.CLSI. Performance Standards for Antimicrobial Susceptibility Testing, 34th ed. CLSI supplement M100, Clinical and Laboratory Standards Institute;2024., (2024). [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data that support this study are included in this published article and its supplementary information or are available from the corresponding authors on reasonable request. Structural data have been deposited to the PDB with the accession codes of 9MD0 (tPBP235/02-12) and 9MCZ (tPBP235/02-15).



