ABSTRACT
The World Health Organization (WHO) has warned that our current arsenal of antibiotics is not innovative enough to face impending infectious diseases, especially those caused by multidrug-resistant Gram-negative pathogens. Although the current preclinical pipeline is well stocked with novel candidates, the last U.S. Food and Drug Administration (FDA)-approved antibiotic with a novel mechanism of action against Gram-negative bacteria was discovered nearly 60 years ago. Of all the antibiotic candidates that initiated investigational new drug (IND) applications in the 2000s, 17% earned FDA approval within 12 years, while an overwhelming 62% were discontinued in that time frame. These “leaks” in the clinical pipeline, where compounds with clinical potential are abandoned during clinical development, indicate that scientific innovations are not reaching the clinic and providing benefits to patients. This is true for not only novel candidates but also candidates from existing antibiotic classes with clinically validated targets. By identifying the sources of the leaks in the clinical pipeline, future developmental efforts can be directed toward strategies that are more likely to flow into clinical use. In this review, we conduct a detailed failure analysis of clinical candidates with Gram-negative activity that have fallen out of the clinical pipeline over the past decade. Although limited by incomplete data disclosure from companies engaging in antibiotic development, we attempt to distill the developmental challenges faced by each discontinued candidate. It is our hope that this insight can help de-risk antibiotic development and bring new, effective antibiotics to the clinic.
KEYWORDS: Gram-negative, antibiotics, clinical trials, toxicity
INTRODUCTION
Bacterial resistance to antibiotics is a growing public health crisis; 1.27 million global deaths were attributed to multidrug resistance (MDR) in 2019 (1). Left unchecked, MDR could lead to 10 million global annual deaths in 2050 (2, 3). Modern medicine relies on antibiotics to control secondary bacterial infections from routine procedures like surgery and chemotherapy. These secondary infections may become untreatable due to antibiotic-resistant bacteria, escalating the risk of common medical procedures.
Of the most threatening MDR pathogens identified by the U.S. Centers for Disease Control and Prevention (CDC) (4) and the World Health Organization (WHO) (2), Gram-negative bacteria (GNB), including Klebsiella pneumoniae (of the Enterobacteriaceae family), Acinetobacter baumannii, and Pseudomonas aeruginosa, stand out as urgent unmet needs. In addition to their general intrinsic resistance to antibiotics, all three have developed critical resistance to the carbapenem class of antibiotics, leaving limited alternative treatment options (5, 6). Despite the growing threat of untreatable infections, the 2020 global antibiotic clinical pipeline contained only 23 candidates with GNB activity, none of which belonged to a new class (7). The high incidence of cross-resistance to existing antibiotics implies that the development of resistance to these new agents is closely trailing (8). While the success rate from phase 1 trials to U.S. Food and Drug Administration (FDA) approval for all antibacterial therapeutics between 2011 and 2020 was 16.3% (9), the last FDA-approved antibiotic with a novel mechanism of action against GNB was discovered nearly 60 years ago.
Clinical studies initiated in the 1980s and 1990s (largely cephalosporins, fluoroquinolones, and macrolides) had high success rates, with 40% of candidates obtaining market approval in a median time of 6 years (10). However, of the 61 antibiotics approved for use between 1980 and 2009, 43% have been withdrawn by the FDA, and the 6 antibiotics withdrawn due to safety issues were all fluoroquinolones (11). Moreover, the number of antibacterial investigational new drug (IND) applications filed with the FDA between 2010 and 2019 is the lowest that it has been in the past 4 decades (10). Despite the unique challenges of antibiotic discovery (12–15), 72% of candidates in the current global preclinical pipeline represent novel classes, with overlapping cellular targets and mechanisms of action that are distinct from those of antibiotics used in the clinic today (7, 16). The consequences of failure are unbearable for the small companies that drive antibiotic development and for the future of a society that so heavily depends on efficacious antibiotics.
Here, we profile antibiotic candidates with GNB activity that have fallen out of the clinical pipeline over the last decade and identify trends in their development. These vignettes are limited by the extent of information disclosure by the companies pursuing these candidates, but we hope to inform future discovery and development efforts by highlighting patterns in these failures. Stronger predictors of success may enable more diverse candidates from the preclinical pipeline to enter a de-risked clinical pipeline and emerge as FDA-approved therapeutics.
OVERVIEW OF THE CLINICAL DEVELOPMENT PIPELINE FOR GNB-ACTIVE ANTIBIOTICS (2010 TO 2020)
The clinical development pipeline for systemic GNB-active candidates over the past decade is detailed in Table 1. Despite the desperate need for antibiotics with novel targets and high target diversity in the preclinical pipeline, most candidates in clinical development are from clinically validated classes (Fig. 1), presumably due to the higher perceived risk of pursuing a non-clinically validated target. While half of all classes in development contain an antibiotic that has been approved in the past 10 years, the other half comprise unexploited antibiotic targets: tRNA synthetases, LpxC, and LptD.
TABLE 1.
Clinical development details of GNB-active antibiotic candidatesa
In the first column, candidates that are approved are shaded blue, those that are in current development are shaded green, and those that are discontinued are shaded orange. In the columns for approved or intended indications, approved or expected activity spectrum, and formulation, a shaded cell indicates that the category shown in the subcolumn heading is a characteristic of the corresponding antibiotic candidate in the first column. New characteristics reported after the date listed in the column labelled last status update are not reflected in this table. DCGI, Drugs Controller General of India; EMA, European Medicines Agency; FDA, U.S. Food and Drug Administration; NDA, new-drug application, filed after clinical trials; NMPA, Chinese National Medical Products Administration; ABSSSI, acute bacterial skin and skin structure infection; CF, cystic fibrosis; cIAI- complicated intra-abdominal infection; HABP, hospital-acquired bacterial pneumonia; uUTI, uncomplicated urinary tract infection; NCFB, non-cystic fibrosis bronchiectasis; NTM, nontubercular mycobacterium; VABP, ventilator-associated bacterial pneumonia; IV, intravenous; Breakthrough therapy, FDA designation to expedite the development and review of drugs; LPAD, limited population pathway for antibacterial and antifungal drugs designation given by the FDA to indicate a limited-usage recommendation; NTAP, new-technology add-on payment designation given by the Centers for Medicare and Medicaid Services as an incentive for hospitals; Orphan Drug, FDA designation given as an incentive; QIDP, qualified infectious disease product designation given by the FDA as an incentive; CRE, carbapenem-resistant Enterobacteriaceae; DBO, diazabicyclooctane class of BLIs with PBP-binding properties; ESBL, extended-spectrum β-lactamase; PBP, penicillin-binding protein.
FIG 1.
GNB-active clinical candidates by class and clinical trial status. Antibiotic classes that have undergone clinical development between 2010 and 2020 are represented as circles. Segments are colored according to the proportions of candidates in that class that have been approved, are currently in clinical development, or have been discontinued.
Although most discontinued candidates are first-time entrants into the clinical development pipeline, some candidates have traversed the pipeline as a different formulation (for example, inhalation therapies) or purposed for other indications (for example, label expansions). The remainder of this review profiles the journey of the 13 first-time entrants that have fallen out of the clinical pipeline. These select candidates target components of the outer membrane (OM), DNA replication, protein translation, and penicillin-binding proteins (PBPs) (Fig. 2A). The structural diversity (Fig. 2B) reflects the variety of mechanisms of action employed to inhibit GNB growth. Most of these candidates were discontinued after phase 1 (Fig. 2C) due to safety concerns (Fig. 2D).
FIG 2.
Profile of GNB-active clinical candidates discontinued between 2010 and 2020. (A) Cellular localizations of targets of discontinued candidates. Yellow boxes represent inhibitors of clinically unprecedented targets. (B) Chemical structures of discontinued candidates. Structures were retrieved from the following PubChem identifiers: 91824766 for murepavadin, 71466517 for ACHN-975, 405560444 for CB-182,804, 53323381 for SPR740, 135905457 for BAL30072, 56640741 for DS-8587, 11676981 for KPI-10, 46836890 for epetraborole, and 77843966 for cefiderocol. The TP-2758 structure was not found in PubChem and was instead replicated from the structure described previously by Sun et al. (49). The AIC499 structure was not found in PubChem and was instead replicated from the structure described previously by Freischem et al. (39). Structures of RC-01 and GSK3342830 are not disclosed. (C) Year and clinical trial stage at the time of discontinuation. Candidates appear in chronological order for each trial stage. (D) Reasons for discontinuation. Limited information was gathered from public press releases and published literature. (Created with BioRender.com.).
DISCONTINUED CANDIDATES WITH CLINICALLY VALIDATED TARGETS
β-Lactam derivatives.
The degradation of β-lactams by β-lactamases is a common resistance mechanism that has been partially addressed by structural optimization of the β-lactam scaffold, adjunctive administration of β-lactamase inhibitors (BLIs), and attachment of a siderophore for improved cellular uptake (17). Among the many attempts since 1980 to overcome resistance by attaching an iron-chelating group to a β-lactam (18, 19), cefiderocol was the first siderophore-antibiotic conjugate to gain FDA approval in 2019. No other clinical-stage siderophore–β-lactam conjugate (cefetecol, BAL30072, GSK3342830, and GT-1) has progressed past phase 1 trials.
(i) BAL30072.
BAL30072 is a siderophore-monobactam conjugate developed by Basilea Pharmaceutica (Basel, Switzerland) derived from tigemonam, with an appended dihydroxypyridinone moiety for iron chelation. Portions of the structure resemble those of aztreonam and avibactam. BAL30072 exhibits bactericidal activity against P. aeruginosa, Acinetobacter species, and Enterobacteriaceae and is stable to metallo-β-lactamases (20, 21). While most monobactams singularly inhibit PBP3, BAL30072 also engages the bifunctional PBP1a and -1b in Escherichia coli (20). Accordingly, while filamentation is usually observed in E. coli cells treated with monobactams targeting PBP3 (22), BAL30072 triggers spheroplasting prior to lysis (20). This spheroplasting phenotype is also elicited by some bicyclic β-lactams (23) and β-lactamase enhancers that target PBP2 (24).
Several in vitro studies indicate the synergy of BAL30072 in combination with meropenem or colistin against various MDR GNB clinical isolates (25–27). In vivo synergy was evaluated in soft tissue infection models of rats challenged with A. baumannii: while BAL30072 showed statistically significant activity, the addition of meropenem was not additive, synergistic, or antagonistic (25). This finding may be rationalized: both BAL30072 and carbapenems inhibit PBP2 in A. baumannii, limiting the pair’s success to mere additive effects. The synergy of these antibiotics might be exploited against Enterobacteriaceae or P. aeruginosa, where they have complementary PBP-binding profiles (27). In murine septicemia, the combination therapy offered protection against carbapenem-resistant P. aeruginosa and MDR A. baumannii, the former due to complementary PBP-binding profiles and the latter possibly due to complementary β-lactamase-binding profiles (27).
A 2010 phase 1 single-ascending-dose (SAD) study reported no serious adverse events at doses of up to 8 g. The multiple-ascending-dose (MAD) study established a maximum tolerated dose, limited by elevated alanine aminotransferase (ALT) enzyme levels (a measure of liver injury). In 2014, Basilea initiated another phase 1 MAD study of BAL30072, both alone and in combination with meropenem. When 2 g BAL30072 was administered as 1-h intravenous (i.v.) infusions every 8 h (6 g/day) or when 4 g of BAL30072 was administered as continuous 22-h infusions for 6 days, abnormally high ALT levels were observed in almost all healthy study subjects by as early as 3 days posttreatment, and the development of the i.v. formulation was ceased (28). In vitro studies revealed that BAL30072 inhibits the mitochondrial electron transport chain, β-oxidation, and glycolysis in HepG2 liver cells at concentrations of 100 to 200 μM, which is clinically relevant only after long-term exposure (28, 29). These findings were unexpected given positive toxicity studies in rats and marmosets dosed with BAL30072 for 4 weeks (28).
To assess utility for urinary tract infection (UTI), urinary concentrations of BAL30072 were analyzed in MAD study subjects (27, 29). Bactericidal activity against P. aeruginosa was weak in urine, presumably due to a low concentration of iron and consequent competition with native siderophores (29). Basilea also began preclinical studies of an inhalation formulation for the treatment of pulmonary infections in cystic fibrosis (CF) patients, which was stopped in 2016 due to a lack of confidence in the candidate’s success (30).
(ii) GSK3342830.
GlaxoSmithKline (GSK) (London, UK) and Shionogi (Osama, Japan) initiated a collaboration in 2010 to discover novel cephem antibiotics with GNB activity, yielding two promising cephalosporin-siderophore conjugates. In 2015, Shionogi retained rights to cefiderocol, which became the first siderophore-antibiotic conjugate to gain FDA approval (31), and GlaxoSmithKline retained rights to the catechol-cephem GSK3342830.
Phase 1 GSK3342830 trials began in 2017 (32). In the SAD component, pharmacokinetic (PK) properties were consistent with those of other cephalosporins, including cefiderocol, and no severe adverse events were detected at doses of up to 6 g (33). In the MAD study, 11 subjects received 1 g GSK3342830 as a single i.v. infusion on day 1, 3-times-a-day i.v. infusions on days 2 through 14, and a single i.v. infusion on day 15. Four participants discontinued the treatment due to headache, malaise, and/or fever, and 1 had high ALT levels leading to automatic discontinuation. The 6 subjects remaining in the study experienced malaise, headache, and fever with onset at between 9 and 10 days and a general decrease in platelet counts (33). While symptoms could be related to known off-target binding to the 5HT-3 serotonin receptor, this interaction seemed physiologically unlikely (33). GSK3342830 was discontinued following these results in 2018.
(iii) GT-1 and GT-055.
GT-1 (LCB10-0200) is a siderophore-cephalosporin conjugate developed by LegoChem Biosciences (Daejeon, South Korea) in a joint venture with Geom Therapeutics (San Francisco, CA, USA). The candidate features the same dihydroxypyridinone siderophore appendage as the one present in BAL30072 and a side chain similar to that of ceftazidime. GT-1 demonstrated efficacy against P. aeruginosa in murine models of systemic, thigh, respiratory tract, and urinary tract infections (34). Its activity spectrum also covers MDR Enterobacteriaceae and A. baumannii (35). The candidate was paired with GT-055 (LCB18-055), a diazabicyclooctane BLI with intrinsic activity against PBP2 (36, 37).
A phase 1 study was registered in Australia in 2019 (38). Only 8 participants were enrolled in this trial when it was terminated due to unspecified safety reasons, presumably hepatotoxicity.
(iv) AIC499.
AIC499 is a monobactam bearing high resemblance to aztreonam with notable activity against MDR A. baumannii and P. aeruginosa. Structural analysis shows hydrophobic interactions between the phenyl portion of the head group and PBP3, while the piperidine portion has a dynamic configuration with a lesser impact on binding yet beneficial PK/pharmacodynamic (PD) properties (39). The candidate was noted to have potent antibacterial activity when coadministered with a BLI, although the combination that AiCuris Anti-infective Cures GmbH (Wuppertal, Germany) pursued in clinical trials was unspecified. Phase 1 began in Austria in 2017, with phase 2 planned for complicated intra-abdominal infection (cIAI) and complicated urinary tract infection (cUTI). These results are unpublished, and the candidate was removed from the company’s pipeline in 2019 for undisclosed reasons.
Fluoroquinolone derivatives.
Fluoroquinolones began receiving FDA approval in the late 1960s for treating UTIs and respiratory tract infections, but the FDA has issued many side-effect warnings for these antibiotics since 2008. Reports of these adverse events during postmarketing surveillance led to the withdrawal of several fluoroquinolones. Second- and third-generation fluoroquinolones like ciprofloxacin, levofloxacin, and moxifloxacin are still used to treat GNB infections.
(i) DS-8587.
DS-8587 is a broad-spectrum fluoroquinolone synthesized by Daiichi Sankyo (Tokyo, Japan) with enhanced bactericidal activity against Acinetobacter baumannii. The candidate retains the core structure of post-second-generation fluoroquinolones, most closely resembling moxifloxacin; however, the fluorination of the cyclopropyl group, the C7 octahydrocyclopentapyrrole, and the methylated C8 distinguish the candidate from the newer-generation candidates that have other fused pyrrolidines at C7 and an ether or no functionality at C8. The dual-targeting compound has micromolar IC50 (50% inhibitory concentration) values for the A. baumannii ParC and GyrA enzymes, high potency against clinical isolates of A. baumannii with mutated ParC and GyrA domains, and low resistance frequency and efflux pump susceptibility (40). In murine calf muscle infection, efficacy was correlated with area under the concentration-versus-time curve (AUC)/MIC values, like other quinolones (41).
Daiichi Sankyo previously marketed three fluoroquinolones (ofloxacin, levofloxacin, and sitafloxacin), but DS-8587 development was discontinued in 2014 after phase 1 for unexplained reasons. Studies from 2017 revealed the in vivo efficacy of DS-8587 against Fusobacterium necrophorum, a pathogenic obligate GNB anaerobe, in murine liver abscess (42).
(ii) KPI-10.
KPI-10 (WQ3813) is a synthetic fluoroquinolone, bearing similarity to 4th-generation trovafloxacin, discovered by Wakunaga Pharmaceutical (Osaka, Japan). The broad-spectrum activity against Enterobacteriaceae, MDR Acinetobacter species, Neisseria gonorrhoeae, and notable Gram-positive organisms, including methicillin-resistant Staphylococcus aureus (MRSA) and Streptococcus pneumoniae (43–45), pointed toward the candidate’s utility in treating both community-acquired bacterial pneumonia (CABP) and UTI.
Kalidex Pharmaceuticals (Menlo Park, CA, USA) licensed the global development and commercialization rights to the candidate. Phase 1 of the oral formulation began in 2012. The SAD study demonstrated a favorable safety and PK profile, supporting a daily oral dosing regimen (46). Clinical development was discontinued for undisclosed reasons, and Kalidex reportedly ceased operation in 2016.
Tetracycline derivatives: (i) TP-2758.
Tetraphase (Watertown, MA, USA) optimized the convergent total synthesis of tetracycline to access analogs that are inaccessible by semisynthesis (47). This approach produced one clinically approved antibiotic (eravacycline) and two other phase 1 candidates (TP-271 and TP-6076). TP-2758, with a chiral 8-pyrrolidinyl substitution, was discovered while generating a series of novel 7-methoxy-8-heterocyclyl tetracycline analogs (48). Derivatives of tetracyclines, called glycylcyclines, were developed to combat the rise of tetracycline resistance. While most tetracyclines are orally dosed, glycylcyclines like tigecycline are restricted to i.v. dosing. TP-2758 was projected to become the first orally bioavailable glycycline.
TP-2758 was more potent than tigecycline against A. baumannii and Enterobacteriaceae, and both oral and i.v. dosing of TP-2758 significantly reduced the burden of infection in murine pyelonephritis induced by E. coli or MDR K. pneumoniae (49). Oral bioavailability values vary between animal species: while tetracycline has oral bioavailabilities of only 14.9% in rats and 6.7% in monkeys, it is >70% in humans (49). TP-2758 had oral bioavailabilities of 8.62% in rats and 30.4% in monkeys, implying higher oral bioavailability in humans than tetracycline (49). Phase 1 studies (50) for the oral formulation began in 2011, but results are unavailable. TP-2758 was removed from the company’s pipeline in 2013, and Tetraphase was acquired by La Jolla Pharmaceutical Company in 2020.
Polymyxin derivatives.
Polymyxins are cationic cyclic peptides (net charge of +5) thought to selectively disrupt and permeabilize the GNB OM to result in bactericidality, although evidence suggests that they may have more than one target (51). When polymyxins were first introduced to the clinic, they were quickly abandoned due to high incidences of dose-limiting nephrotoxicity and neurotoxicity (52). However, with the rise of MDR Gram-negative pathogens, this class has resurged in the clinic as a last-resort therapy (53). The two clinically administered polymyxins, polymyxin B (PMB) and colistin, are manufactured by fermentation as an impure, heterogeneous mix of related compounds. CB-182,804 was the first polymyxin to undergo clinical trials under the FDA’s oversight.
(i) CB-182,804.
BioSource Pharmaceuticals (Spring Valley, NY, USA) developed a semisynthetic route to substitute the N-terminal fatty acyl group that contributes to the toxicity of PMB by utilizing a deacylase enzyme from the microorganism Actinoplanes utahensis (54). After screening many urea-linked halophenyl functionalities for antimicrobial activity, the 2-chlorophenylurea derivative CB-182,804 emerged as a lead candidate. The candidate had bactericidal activity against E. coli, K. pneumoniae, P. aeruginosa, and A. baumannii. Cubist Pharmaceuticals (Lexington, MA, USA) obtained a provisional license for the candidate, and subsequent patents were filed jointly to further develop the strategy (55).
The MICs of CB-182,804 against 5,000 clinical isolates were only 2-fold higher than those of PMB, with observable cross-resistance (56). Similarly, in vivo efficacies in murine P. aeruginosa lung and A. baumannii thigh infection models were comparable for the two (57). However, the 50% effective concentration (EC50) values against a rat renal tubule cell line were >1,000 mg/L for CB-182,804 and 318 mg/L for PMB (57). In cynomolgus monkeys dosed at 6.6 mg/kg of body weight/day 3 times a day for 7 days, CB-182,804 showed limited renal tubular histological changes, whereas PMB exhibited renal tubular degeneration; at a higher dose of 9.9 mg/kg/day, CB-182,804 elicited only slight increases in blood urea nitrogen and serum creatinine, whereas PMB elicited severe signs of nephrotoxicity (58). CB-182,804 also demonstrated more favorable PK/PD parameters than PMB, including decreased serum protein binding, increased plasma clearance, increased volume of distribution, less systemic exposure, as well as a lower maximum concentration of drug in serum (Cmax) (58).
Clinical trials began in February 2009, but the development of this molecule ceased in 2010, presumably due to nephrotoxicity issues (59). Cubist was acquired by Merck Pharmaceuticals in 2015.
(ii) SPR741.
SPR741 (NAB741) is a fully synthetic PMB derivative that was designed to curtail nephrotoxicity issues associated with this class through a reduced positive charge (3+) and the removal of the highly lipophilic fatty acid side chain in PMB (60). In a rat model, renal clearance of SPR741 was 400-fold higher than that of colistin, suggesting improved safety-related PK properties (60). Despite having weak antibacterial activity, sub-MIC dosing of SPR741 enhances the permeation of other antibiotics through the OM (61). In vivo studies confirm this potentiation with expanded azithromycin coverage against MDR Enterobacteriaceae (62) and synergy with rifampicin against extremely drug-resistant (XDR) A. baumannii (63).
In a phase 1 drug-drug interaction study, i.v. dosing of other antibiotics (1.0 g of ceftazidime, 4.5 g of piperacillin-tazobactam, or 1.0 g of aztreonam) with 400 mg of SPR741 did not significantly affect the concentration-versus-time profile, clearance, or half-life of either drug (64). In the MAD study, 25% of subjects experienced decreased creatinine clearance across all drug dosage cohorts: 3 in the 600-mg, 1 in the 400-mg, 1 in the 150-mg, and 1 in the 50-mg cohorts (64). Of these 6 subjects, 5 had normal creatinine levels at day 16, while 1 from the 600-mg cohort had a moderate increase in the serum creatinine level above the baseline level that began on day 14. SPR741 was discontinued in January 2020 and replaced by SPR206, a different polymyxin analog from the potentiator platform. While SPR741 was developed as an antibiotic adjuvant, SPR206 has antibacterial activity as a standalone therapy and boasts potentially superior safety and efficacy profiles to those of SPR741.
DISCONTINUED CANDIDATES WITH CLINICALLY UNPRECEDENTED TARGETS
Murepavadin (LptD inhibitor).
Inspired by the antimicrobial host defense peptide protegrin I, Polyphor Ltd. (Allschwil, Switzerland) synthesized and screened a library of β-hairpin-shaped macrocyclic protein epitope mimetics for antimicrobial activity (65–68). While initial leads exhibited hemolysis of red blood cells and degradation by serum enzymes, optimization toward antibacterial activity yielded the clinical candidate murepavadin (POL-7080) (68, 69). Murepavadin reportedly targets the β-barrel protein LptD (69–71), an essential (72) surface-exposed OM protein that acts in a complex (73–75) to incorporate lipopolysaccharide (LPS) into the OM of GNB. The differential N-terminal lengths of LptD among GNB is thought to confer the specificity of murepavadin to the P. aeruginosa protein (69). In preclinical studies, murepavadin outperformed comparator antibiotics, including colistin, against even XDR P. aeruginosa clinical isolates (76, 77). Although oral bioavailability was low in rats, subcutaneous administration in humans yielded a bioavailability of 67 to 79% and a half-life of 5 to 8 h. The discovery and development of murepavadin has previously been reviewed (78).
Roche (Basel, Switzerland) obtained a license to develop and commercialize murepavadin in 2013. Six phase 1 studies explored the safety, tolerability, and PK of murepavadin: a combined SAD and MAD study in healthy male subjects (79), a multiple-dose study evaluating the penetration of murepavadin into the lungs (80), a drug-drug interaction investigation of murepavadin with colistin (81) and with amikacin (82), a thorough QT (TQT) (in reference to QT intervals measured by an electrocardiogram) study with SAD (83), and an SAD study of murepavadin in subjects with renal function impairment (84). Systemic exposure to murepavadin was increased in subjects with renal function impairment, indicating a need for dose adjustment based on the creatinine clearance rate (85). Despite Roche returning the murepavadin development license to Polyphor in 2015, two phase 2 studies were successfully completed: a 14-day dosage of murepavadin in subjects with acute exacerbation of non-cystic fibrosis bronchiectasis due to P. aeruginosa infection (86) and a MAD study of murepavadin coadministered with the standard of care (SOC) in subjects with ventilator-associated bacterial pneumonia (VABP) due to P. aeruginosa infection (87). In the latter study, clinical cure was achieved in 10 out of 12 (83%) patients with confirmed P. aeruginosa infection, and the 28-day all-cause mortality rate in this population was 9% (88).
Although murepavadin’s narrow spectrum of activity provides advantages as a treatment option, it complicated the phase 3 clinical trial design (89). While phases 1 and 2 tested murepavadin as a monotherapy, the ethics of phase 3 trials in pneumonia patients necessitated the coadministration of murepavadin with a broad-spectrum drug (89). The coadministered antibiotic needed to have no pseudomonal activity to avoid confounding the results of the trial. Ertapenem, a first-line therapy for CABP, was ultimately chosen for coadministration, and the appropriate dosing for hospital-acquired bacterial pneumonia (HABP)/VABP was determined (89).
Murepavadin underwent two separate phase 3 trials to test its efficacy in HABP/VABP infection due to P. aeruginosa (90, 91). The FDA-approved noninferiority study (PRISM-UDR) (90) compared murepavadin plus ertapenem to 1 β-lactam antibiotic to treat HABP/VABP driven by P. aeruginosa in clinical centers with a low incidence of MDR. The European Medicines Agency (EMA)-approved study (PRISM-MDR) (91), in contrast, compared murepavadin plus 1 antipseudomonal antibiotic to 2 antipseudomonal antibiotics in clinical centers with a high incidence of MDR to assess murepavadin efficacy over the SOC. Although a 25 to 40% incidence of kidney injury was anticipated based on the comparator arm, 56% of patients treated with murepavadin in the VABP study showed evidence of acute kidney injury (92). Polyphor terminated i.v. formulation development as of July 2019 due to nephrotoxicity concerns. Murepavadin was the only GNB-active clinical candidate in this decade to be discontinued after phase 3. Polyphor continued the preclinical development of an inhalation formulation of murepavadin, and clinical trial authorization was granted in the United Kingdom in December 2020.
ACHN-975 (LpxC inhibitor).
LpxC is a cytosolic zinc-dependent metalloenzyme that catalyzes the first committed step of lipid A biosynthesis. While many antibiotic discovery programs have pursued LpxC inhibitors (93), Achaogen’s (South San Francisco, CA, USA) structure-based discovery effort yielded the first LpxC inhibitor to advance into clinical trials. Like other previously patented LpxC inhibitors (94, 95), this synthetic compound contains a hydroxamic acid moiety that coordinates the catalytic Zn2+ and a long hydrophobic tail that interacts with the active-site tunnel.
While the genetic sequence of LpxC is highly conserved across GNB, the subtle structural differences in LpxC influence the potency and dynamics of inhibition (96). ACHN-975 exhibited optimal efficacy when the dose was administered once daily for P. aeruginosa but administered multiple times a day for E. coli and K. pneumoniae, so an intermittent high-dose regimen was established to treat respiratory P. aeruginosa infections (97). The possibility of resistance emergence set the minimum required dose: at concentrations 4-fold higher than the MIC, the frequency of resistance ranged from 10−7 to 10−10 in P. aeruginosa clinical isolates (97). However, ACHN-975 induces bradycardia in preclinical animal models (98), setting a maximum tolerated dose.
In 2012, a phase 1 SAD study to assess the candidate’s safety, tolerability, and PK in 50 healthy volunteers (99) was completed. The therapeutic window was deemed insufficient due to concentration-driven dose-limiting cardiovascular toxicity (transient hypotension without tachycardia), which occurred in the first subject who received an 18-mg/kg infusion (100). A 2013 MAD study (101) was prematurely terminated after enrolling four subjects. Participants encountered inflammation at the infusion site after repeat dosing of 4 mg/kg three times a day for 3 to 4 days.
In 2015, Achaogen began an optimization program focusing on P. aeruginosa (100). This pathogen was more sensitive to LpxC inhibition in in vivo models than Enterobacteriaceae species, and the structural features of P. aeruginosa LpxC seemed more amenable to curtailing drug toxicity (100). To investigate structure-toxicity relationships, a high-content assay in anesthetized rats was developed to assess maximum tolerated concentrations (100). Cardiovascular toxicity was attributed to a nonspecific effect of basic amines, so a new candidate was identified with a wider therapeutic window. With the removal of the amine, this new candidate was nonsolubilizable at 10- to 100-mg/mL concentrations using acidic pH (100). To overcome solubility issues and accommodate the anticipated dose of >1 g per day, the hydroxyl tail was converted to a phosphate prodrug. Surprisingly, this new prodrug, dosed in a simple aqueous formulation, demonstrated cardiovascular toxicity in the anesthetized rat model, even though the parent molecule, dosed in pH-adjusted hydroxypropyl-cyclodextrin, did not (100). Compounds and insights from these studies were passed on to Forge Therapeutics (San Diego, CA, USA) after Achaogen filed for bankruptcy in 2019.
RC-01 (LpxC inhibitor).
Fujifilm Toyama Chemical Co. Ltd. (Toyoma, Japan) screened compounds with malonamide, a derivative of the zinc-chelating hydroxamic acid, for LpxC activity. RC-01 (T-1228) was identified as a lead compound, exhibiting a subnanomolar IC50 against LpxC and bactericidal activity against P. aeruginosa and Enterobacteriaceae (102). In vitro exposure of RC-01 to GNB reduces the release of LPS (103), corroborating in vivo data from other LpxC inhibitors that decrease LPS-dependent stimulation of the host immune system, thereby attenuating bacterial virulence (104). In mouse models of P. aeruginosa-induced pneumonia and E. coli-induced UTI, the PK/PD parameter most highly correlated with efficacy was the AUC for the free, unbound fraction of the drug (fAUC)/MIC ratio (105). The frequency of resistance to RC-01 at 4× MIC was 10−7 to 10−8 (106).
In 2019, Recida Therapeutics (Menlo Park, CA, USA) licensed the development and commercialization rights for RC-01 outside Japan. LpxC-associated cardiovascular toxicity was unapparent with RC-01: at least 400 mg/kg/day was tolerated in 2-week repeated i.v. dosing in rats and dogs, with unreported fAUC and Cmax (107). Two formulations of RC-01 were pursued: an inhalation therapy for respiratory infections and i.v. therapy for systemic infections. The programs were prematurely terminated after enrolling 8 subjects in a phase 1 SAD study (108) for unspecified safety reasons. Recida soon after surrendered its business rights in California, and MicuRx was granted rights for investigational treatment with RC-01 in China.
Epetraborole (LeuRS inhibitor).
Epetraborole (GSK2251052; AN3365) is a bacteriostatic oxaborole-containing inhibitor (109) of leucyl-tRNA synthetase (LeuRS) that was discovered in a structure-based rational design screen led by Anacor Pharmaceuticals (Palo Alto, CA, USA). The only FDA-approved aminoacyl-tRNA synthetase inhibitor is mupirocin, which targets isoleucyl-tRNA synthetase for the treatment of Gram-positive infections (110–112). Mupirocin is restricted to topical use due to the rapid metabolism of its ester moiety and resistance emergence (113).
The mechanism of a benzoxaborole antifungal agent trapping the active conformation of the editing site of LeuRS inspired the rational design of epetraborole (114). Guided by crystallography, benzoxaborole analogs with extended coverage against A. baumannii were synthesized (114, 115). Screening against MDR clinical isolates demonstrated a 10−7 one-step resistance frequency at 4× MIC (115), coverage of anaerobic microorganisms (116, 117), and low MIC90s against P. aeruginosa (118). Mouse thigh infections highlighted the candidate’s efficacy against MDR GNB in vivo (115).
In 2009, Anacor initiated phase 1 trials for the i.v. formulation and reported favorable safety and PK properties in 72 subjects (119). In accordance with a 2007 alliance forged with GlaxoSmithKline (London, UK), GSK obtained an exclusive license for epetraborole in 2010. Phase 1 trials included SAD and MAD studies of oral formulations (120), a small-cohort mass balance study of the i.v. formulation (121), and serum and pulmonary PK of the i.v. formulation (122). Like mupirocin, epetraborole is highly metabolized in monkeys and humans: the oxidation of the propanol side chain by the polymorphic alcohol dehydrogenase generates an inactive carboxylic acid metabolite (123). Following a 1,500-mg i.v. infusion of the candidate in 6 human subjects, the candidate was found in systemic circulation and urinary excretions in its original form and, to a great extent, its oxidized form.
GSK initiated phase 2 trials for cUTI (124) and cIAI (125). In 3 of the 14 patients receiving epetraborole in the cUTI study, resistant isolates were recovered after only 1 day of treatment (126). Whole-genome sequencing revealed target-specific mutations in the LeuRS editing domain that conferred a low fitness cost (126). The emergence of these fit mutants suggests that either this specific mode of binding to LeuRS or general inhibition of LeuRS is unproductive for impeding bacterial growth. Due to resistance concerns, the cUTI study was terminated in 2012, and the cIAI study was terminated as a precaution, even though isolates from 3 of the 9 patients who received epetraborole in this study maintained baseline susceptibility to the drug candidate (126). GSK also assessed drug distributions in epithelial lining fluid and alveolar macrophages, which showed promise for efficacy under a pneumonia indication (127). GSK soon after returned licensing rights to Anacor, which was acquired by Pfizer in 2016.
DISCUSSION
A decade of leakiness in the GNB-active antibiotic clinical development pipeline is apparent from this review. The most prominent crack in the pipeline is the transition between phase 1 and phase 2. Data from AntibioticDB (128), a growing repository for antibiotics in global preclinical and clinical development from the 1960s to the present, show similar termination frequencies by clinical stage of development. In contrast, drugs from other therapeutic areas (including the “infectious disease” category) have the lowest success rate in the transition from phase 2 to phase 3 trials (129).
Both AntibioticDB and Hay et al. cite toxicology concerns (observable in phase 1) and lack of efficacy (after phase 1) as equally large determinants of failure for clinical candidates with disclosed discontinuation reasons. For the GNB-active candidates of this decade, however, halts over the past 10 years are largely attributable to safety issues in phase 1 trials; besides safety, three candidates were discontinued for unknown reasons, only one encountered resistance, one was replaced officially for commercial reasons, and none cited efficacy concerns (Fig. 2C).
Of the 13 discontinued candidates, 4 could have been first-in-class inhibitors, representing 3 novel targets: LptD, LpxC, and LeuRS. CB-182,804 was the first polymyxin to undergo clinical trials. Three of the four discontinued β-lactams attempted to follow the siderophore-antibiotic conjugation strategy successfully employed for cefiderocol. Overall, it is unclear whether novel targets are exceptionally failure prone given their small sample size. The poor safety profiles of these novel candidates may be due to the modalities of inhibiting new targets and/or the unanticipated toxicities of the novel chemical scaffolds. In the search for new antibiotics, the termination of first-in-class antibiotics is especially painful, as these new drugs provide hope for evading MDR.
Some of these discontinued clinical candidates do not strictly follow empirical guidelines for antibiotic design (130, 131). For example, while epetraborole was the only candidate terminated due to the emergence of resistance, the LpxC inhibitors ACHN-975 and RC-01 posed the same concerns for resistance due to their requisite high exposure and single-copy-single-enzyme-targeting mechanism (97). Additionally, ACHN-975 chelates the catalytic zinc of LpxC with hydroxamic acid, which is associated with the release of toxic metabolic by-products and off-target inhibition (100, 132–134). However, replacing the moiety impairs inhibitory potency and antibacterial activity with persisting toxicity (135, 136), underscoring the need for probing structure-toxicity relationships in new antibiotic classes. Conceivably, in vivo preclinical models are good predictors of antibacterial efficacy but poor predictors of safety, and alternate methods for assessing structure-toxicity relationships in vitro and in vivo should be developed.
The termination of some candidates was surprising considering the published toxicity data. Although hepatotoxicity was unapparent in preclinical models, BAL30072 treatment caused elevated ALT levels after only 3 days. In vitro nephrotoxicity is an unreliable predictor of clinical nephrotoxicity (137), which is especially problematic for polymyxins like CB-182,804 (138, 139). Despite decades of polymyxin use, structure-toxicity relationships of this class are still understudied; this gap in understanding coupled with the characteristic toxicity of this class may account for the dearth of analog development (140). Likewise, the long history of the β-lactam class, the similarity of siderophore-conjugated candidates to approved antibiotics, and the prior approval of one siderophore-conjugated antibiotic were insufficient to bring more siderophore-conjugated antibiotics to the clinic, and a better understanding of structure-toxicity relationships of the linker and iron chelator components may de-risk future development. The case of murepavadin highlights a latent nephrotoxicity concern that surfaced only in phase 3: phase 1 and 2 trials comprised 8 studies, in which 257 subjects received at least a single dose of murepavadin for up to 15 days, and the only 3 serious adverse events (SAEs) reported were fully reversible after discontinuation (78). As patients in phase 3 trials are typically sicker than the healthy subjects in phase 1, antibiotic toxicology must account for higher-acuity settings.
Can discontinued candidates be revived in the clinical pipeline? Polyphor has already initiated murepavadin clinical development by reformulating from i.v. to oral. An inhalation formulation could benefit pneumonia treatment candidates with dose-limiting toxicity by decreasing systemic exposure and increasing concentrations in lung tissues (141), and all approved inhaled antibiotics are reformulations of compounds initially dosed through the i.v. or oral route.
Another strategy for candidates with dose-limiting toxicity is coadministration in a synergistic combination therapy to expand their therapeutic window. While this strategy invites challenges pertaining to matching PK properties, it has been employed for several antibiotics: novel BLIs have successfully extended the spectrum of β-lactams to MDR GNB (142). In addition to binding β-lactamases, some potentiators inhibit cell growth by PBP-binding-dependent and -independent mechanisms (143, 144). Discontinued PBP-binding candidates could be explored further in combination with a BLI or as an adjuvant for other β-lactams of complementary PBP-binding and β-lactamase-binding properties. For example, the synergy of BAL30072 with meropenem compelled Basilea to pursue combination therapy in phase 1 trials despite the dose-limiting hepatotoxicity encountered in the previous MAD study of BAL30072 alone.
Similarly, antibiotic potency and/or spectrum of activity can be potentiated with polymyxins. Polymyxins have been investigated as potentiators for other classes of antibiotics without conclusive evidence of synergy in clinical treatments (145, 146). In vitro studies show evidence of polymyxin synergy with many antibiotics (147), including the addition of rifampicin to CB-182804 to improve potency and MDR coverage (56). SPR741 employed this potentiation strategy, although it was discontinued after phase 1 trials for commercial reasons.
Additionally, LpxC inhibitors have demonstrated synergy with antibiotics for which GNB activity is limited by the OM, like rifampicin and tetracycline (93). LpxC inhibition may contribute to A. baumannii clearance in vivo by enhancing bacterial opsonophagocytosis and reducing inflammation (104) despite the nonessentiality of LPS biosynthesis in this species and the resultant in vitro inefficacy of LpxC inhibitors. This antivirulence-based mechanism of action may reduce its likelihood of encountering resistance and extend the coadministered antibiotic’s spectrum of activity.
Finally, there is a critical yet latent misalignment of the antibiotic discovery pipeline with the clinical development pipeline (148). While antibiotic discovery typically focuses on identifying candidates corresponding to a particular MDR pathogen, cellular target, or chemical structure, late-stage clinical trials primarily test the candidate’s efficacy in the context of clinical indications. Even if a candidate fills an unmet need by targeting a critical MDR pathogen or demonstrating low cross-resistance, that coverage may be moot when tested at clinical trial sites with low incidences of MDR and compared to SOCs with high efficacy against susceptible pathogens (149). Since the rapid determination of an infection’s causative organism is usually infeasible, empirical treatment based on infection site is common. Recently, the FDA required an infection-site-specific indication while the EMA preferred a resistant-pathogen-specific indication for phase 3 trials of cefiderocol (31, 150). Such innovations in clinical trial design may enable the alignment of approved antibiotics with the unmet needs associated with antimicrobial resistance.
Structural, preclinical, and clinical data were inaccessible for several candidates. Considering that some public funding was critical for the early success of many candidates, we echo the call for broader data sharing (151). Although some public databases have compiled data, including ClinicalTrials.gov, the Pew Charitable Trusts, SPARK, and AntibioticDB, we should strive for completeness in archiving. As Achaogen, after declaring bankruptcy, shared its LpxC platform data with Forge Therapeutics, other abandoned data and learned lessons should be passed on.
In conclusion, the critical leak in the GNB-active antibiotic clinical development pipeline is between phase 1 and phase 2 and is largely attributable to safety issues. By sealing this rupture, we can increase the likelihood of FDA approval and de-risk investment in the antibiotic space. Given the complexities of antibiotic design from target validation and permeability to evasion of resistance mechanisms and nonconventional pharmacological properties, the low diversity of clinical trial termination reasons is notable. While safety presents a major challenge for antibiotic clinical development in this decade, solving this phase 1 issue may expose other issues in later clinical trials or after approval, like resistance or efficacy. Without innovations in preclinical predictive studies and clinical trial designs (148), the novel candidates in today’s preclinical pipeline that transition to clinical development in the next decade may face the same complications and consequences as those of the last. Alternatively, novel candidates with favorable in vivo profiles may be abandoned in the preclinical stage if the false-positivity rate of preclinical toxicity assays is too high. Finally, with many candidates withdrawn without public explanation as to why, it is challenging to learn from previous mistakes. Increased data sharing through existing mechanisms could reduce redundancy and accelerate future antibiotic development.
ACKNOWLEDGMENTS
O.S.R. is an employee of The Janssen Pharmaceutical Companies of Johnson & Johnson. R.T.C. is a former employee of Achaogen Inc. and a current employee of Revagenix Inc.
REFERENCES
- 1.Antimicrobial Resistance Collaborators. 2022. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 399:629–655. 10.1016/S0140-6736(21)02724-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.World Health Organization. 2017. Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics. World Health Organization, Geneva, Switzerland. [Google Scholar]
- 3.Review on Antimicrobial Resistance. 2014. Antimicrobial resistance: tackling a crisis for the health and wealth of nations.
- 4.US Centers for Disease Control and Prevention. 2019. Antibiotic resistance threats in the United States, 2019. US Centers for Disease Control and Prevention, Atlanta, GA. 10.15620/cdc:82532. [DOI] [Google Scholar]
- 5.Bassetti M, Peghin M, Vena A, Giacobbe DR. 2019. Treatment of infections due to MDR Gram-negative bacteria. Front Med (Lausanne) 6:74. 10.3389/fmed.2019.00074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Isler B, Doi Y, Bonomo RA, Paterson DL. 2019. New treatment options against carbapenem-resistant infections. Antimicrob Agents Chemother 63:e01110-18. 10.1128/AAC.01110-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Butler MS, Paterson DL. 2020. Antibiotics in the clinical pipeline in October 2019. J Antibiot (Tokyo) 73:329–364. 10.1038/s41429-020-0291-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Theuretzbacher U, Bush K, Harbarth S, Paul M, Rex JH, Tacconelli E, Thwaites GE. 2020. Critical analysis of antibacterial agents in clinical development. Nat Rev Microbiol 18:286–298. 10.1038/s41579-020-0340-0. [DOI] [PubMed] [Google Scholar]
- 9.Stinson V. 2022. New BIO report examines the state of antibacterial innovation. Biotechnology Innovation Organization, Washington, DC. https://www.bio.org/press-release/new-bio-report-examines-state-antibacterial-innovation. Accessed 4 March 2022. [Google Scholar]
- 10.Dheman N, Mahoney N, Cox EM, Farley JJ, Amini T, Lanthier ML. 2021. An analysis of antibacterial drug development trends in the United States, 1980–2019. Clin Infect Dis 73:e4444–e4450. 10.1093/cid/ciaa859. [DOI] [PubMed] [Google Scholar]
- 11.Outterson K., Powers J. H, Seoane-Vazquez E, Rodriguez-Monguio R, Kesselheim A. S.. 2013. Approval and withdrawal of new antibiotics and other antiinfectives in the U.S., 1980-2009. J. Law Med. Ethics. 41:688–696. [DOI] [PubMed] [Google Scholar]
- 12.Silver LL. 2011. Challenges of antibacterial discovery. Clin Microbiol Rev 24:71–109. 10.1128/CMR.00030-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.McDowell LL, Quinn CL, Leeds JA, Silverman JA, Silver LL. 2019. Perspective on antibacterial lead identification challenges and the role of hypothesis-driven strategies. SLAS Discov 24:440–456. 10.1177/2472555218818786. [DOI] [PubMed] [Google Scholar]
- 14.O’Shea R, Moser HE. 2008. Physicochemical properties of antibacterial compounds: implications for drug discovery. J Med Chem 51:2871–2878. 10.1021/jm700967e. [DOI] [PubMed] [Google Scholar]
- 15.Hughes D, Karlén A. 2014. Discovery and preclinical development of new antibiotics. Ups J Med Sci 119:162–169. 10.3109/03009734.2014.896437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Theuretzbacher U, Outterson K, Engel A, Karlén A. 2020. The global preclinical antibacterial pipeline. Nat Rev Microbiol 18:275–285. 10.1038/s41579-019-0288-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bush K. 2018. Past and present perspectives on β-lactamases. Antimicrob Agents Chemother 62:e01076-18. 10.1128/AAC.01076-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Möllmann U, Heinisch L, Bauernfeind A, Köhler T, Ankel-Fuchs D. 2009. Siderophores as drug delivery agents: application of the “Trojan Horse” strategy. Biometals 22:615–624. 10.1007/s10534-009-9219-2. [DOI] [PubMed] [Google Scholar]
- 19.Page MGP. 2013. Siderophore conjugates. Ann N Y Acad Sci 1277:115–126. 10.1111/nyas.12024. [DOI] [PubMed] [Google Scholar]
- 20.Page MGP, Dantier C, Desarbre E. 2010. In vitro properties of BAL30072, a novel siderophore sulfactam with activity against multiresistant Gram-negative bacilli. Antimicrob Agents Chemother 54:2291–2302. 10.1128/AAC.01525-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Higgins PG, Stefanik D, Page MGP, Hackel M, Seifert H. 2012. In vitro activity of the siderophore monosulfactam BAL30072 against meropenem-non-susceptible Acinetobacter baumannii. J Antimicrob Chemother 67:1167–1169. 10.1093/jac/dks009. [DOI] [PubMed] [Google Scholar]
- 22.Curtis NA, Eisenstadt RL, Turner KA, White AJ. 1985. Inhibition of penicillin-binding protein 3 of Escherichia coli K-12. Effects upon growth, viability and outer membrane barrier function. J Antimicrob Chemother 16:287–296. 10.1093/jac/16.3.287. [DOI] [PubMed] [Google Scholar]
- 23.Hanberger H, Nilsson LE, Kihlström E, Maller R. 1990. Postantibiotic effect of beta-lactam antibiotics on Escherichia coli evaluated by bioluminescence assay of bacterial ATP. Antimicrob Agents Chemother 34:102–106. 10.1128/AAC.34.1.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gutmann L, Vincent S, Billot-Klein D, Acar JF, Mrèna E, Williamson R. 1986. Involvement of penicillin-binding protein 2 with other penicillin-binding proteins in lysis of Escherichia coli by some beta-lactam antibiotics alone and in synergistic lytic effect of amdinocillin (mecillinam). Antimicrob Agents Chemother 30:906–912. 10.1128/AAC.30.6.906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Russo TA, Page MGP, Beanan JM, Olson R, Hujer AM, Hujer KM, Jacobs M, Bajaksouzian S, Endimiani A, Bonomo RA. 2011. In vivo and in vitro activity of the siderophore monosulfactam BAL30072 against Acinetobacter baumannii. J Antimicrob Chemother 66:867–873. 10.1093/jac/dkr013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hornsey M, Phee L, Stubbings W, Wareham DW. 2013. In vitro activity of the novel monosulfactam BAL30072 alone and in combination with meropenem versus a diverse collection of important Gram-negative pathogens. Int J Antimicrob Agents 42:343–346. 10.1016/j.ijantimicag.2013.05.010. [DOI] [PubMed] [Google Scholar]
- 27.Hofer B, Dantier C, Gebhardt K, Desarbre E, Schmitt-Hoffmann A, Page MGP. 2013. Combined effects of the siderophore monosulfactam BAL30072 and carbapenems on multidrug-resistant Gram-negative bacilli. J Antimicrob Chemother 68:1120–1129. 10.1093/jac/dks527. [DOI] [PubMed] [Google Scholar]
- 28.Paech F, Messner S, Spickermann J, Wind M, Schmitt-Hoffmann A-H, Witschi AT, Howell BA, Church RJ, Woodhead J, Engelhardt M, Krähenbühl S, Maurer M. 2017. Mechanisms of hepatotoxicity associated with the monocyclic β-lactam antibiotic BAL30072. Arch Toxicol 91:3647–3662. 10.1007/s00204-017-1994-x. [DOI] [PubMed] [Google Scholar]
- 29.Straubinger M, Blenk H, Naber KG, Wagenlehner FME. 2016. Urinary concentrations and antibacterial activity of BAL30072, a novel siderophore monosulfactam, against uropathogens after intravenous administration in healthy subjects. Antimicrob Agents Chemother 60:3309–3315. 10.1128/AAC.02425-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Taylor NP. 2016. Basilea drops inhaled antibiotic, expands cancer program in early-stage rejig. Fierce Biotech. https://www.fiercebiotech.com/biotech/basilea-drops-inhaled-antibiotic-expands-cancer-program-early-stage-rejig. Accessed 4 March 2022.
- 31.Echols R, Ariyasu M, Nagata TD. 2019. Pathogen-focused clinical development to address unmet medical need: cefiderocol targeting carbapenem resistance. Clin Infect Dis 69:S559–S564. 10.1093/cid/ciz829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.GlaxoSmithKline. 2016. A study to evaluate safety, tolerability and pharmacokinetics of ascending intravenous single dose and repeat dose of GSK3342830. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02751424 (identifier NCT02751424). Accessed 4 February 2021.
- 33.Tenero D, Farinola N, Berkowitz EM, Tiffany CA, Qian Y, Xue Z, Raychaudhuri A, Gardiner DF. 2019. Pharmacokinetics, safety, and tolerability evaluation of single and multiple doses of GSK3342830 in healthy volunteers. Clin Pharmacol Drug Dev 8:754–764. 10.1002/cpdd.637. [DOI] [PubMed] [Google Scholar]
- 34.Oh S-H, Park H-S, Kim H-S, Yun J-Y, Oh K, Cho Y-L, Kwak J-H. 2017. Antimicrobial activities of LCB10-0200, a novel siderophore cephalosporin, against the clinical isolates of Pseudomonas aeruginosa and other pathogens. Int J Antimicrob Agents 50:700–706. 10.1016/j.ijantimicag.2017.06.001. [DOI] [PubMed] [Google Scholar]
- 35.Nguyen LP, Pinto NA, Vu TN, Lee H, Cho YL, Byun J-H, D’Souza R, Yong D. 2020. In vitro activity of a novel siderophore-cephalosporin, GT-1 and serine-type β-lactamase inhibitor, GT-055, against Escherichia coli, Klebsiella pneumoniae and Acinetobacter spp. panel strains. Antibiotics (Basel) 9:267. 10.3390/antibiotics9050267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lee J, Cho Y-L, Shin S, Biek D. 2019. Penicillin binding protein (PBP) activity of beta-lactamase inhibitor GT-055, abstr P1187. Abstr 29th Eur Congr Clin Microbiol Infect Dis.
- 37.Sader HS, Duncan LR, Thompson J, Cho YL, Biek D, Flamm RK. 2018. Antimicrobial activity of the novel siderophore cephalosporin GT-1 tested alone and combined with the β-lactamase inhibitor GT-055 against molecularly characterized Enterobacteriaceae clinical isolates, abstr Sunday-571. Abstr ASM Microbe 2018.
- 38.Geom Therapeutics Australia Pty Ltd. 2018. A placebo controlled phase 1 study to determine the safety and tolerability of ascending doses of GT-1 (a new antibiotic) in healthy adult participants following intravenous infusion. ANZCTR. https://anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12618001980224 (registration number ACTRN12618001980224). Accessed 3 February 2021.
- 39.Freischem S, Grimm I, López-Pérez A, Willbold D, Klenke B, Vuong C, Dingley AJ, Weiergräber OH. 2021. Interaction mode of the novel monobactam AIC499 targeting penicillin binding protein 3 of Gram-negative bacteria. Biomolecules 11:1057. 10.3390/biom11071057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Higuchi S, Onodera Y, Chiba M, Hoshino K, Gotoh N. 2013. Potent in vitro antibacterial activity of DS-8587, a novel broad-spectrum quinolone, against Acinetobacter baumannii. Antimicrob Agents Chemother 57:1978–1981. 10.1128/AAC.02374-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Higuchi S, Kurosaka Y, Uoyama S, Yoshida K, Chiba M, Ishii C, Fujikawa K, Karibe Y, Hoshino K. 2014. Anti-multidrug-resistant Acinetobacter baumannii activity of DS-8587: in vitro activity and in vivo efficacy in a murine calf muscle infection model. J Infect Chemother 20:312–316. 10.1016/j.jiac.2014.01.011. [DOI] [PubMed] [Google Scholar]
- 42.Nagaoka K, Yanagihara K, Morinaga Y, Kurosaka Y, Hoshino K, Kohno S. 2017. In vivo antianaerobe activity of DS-8587, a new fluoroquinolone, against Fusobacterium necrophorum in a mouse model. J Infect Chemother 23:131–135. 10.1016/j.jiac.2016.09.007. [DOI] [PubMed] [Google Scholar]
- 43.Suzuki H, Hayashi N, Amago T, Takenaka H, Amano H, Itoh K, Kuramoto Y, Yazaki A. 2008. WQ-3810, a next generation respiratory fluoroquinolone with outstanding activity against QRSP. Abstr 48th Annu Intersci Conf Antimicrob Agents Chemother-Infect Dis Soc Am 46th Annu Meet.
- 44.Flamm RK, Biedenbach DJ, Sader HS, Konrardy ML, Jones RN. 2012. KPI-10 in vitro activity tested against pathogens commonly associated with community-acquired bacterial pneumonia (CABP) infections, abstr F-2051. Abstr 52nd Annu Intersci Conf Antimicrob Agents Chemother.
- 45.Kazamori D, Suzuki H, Amago T, Itoh K, Yazaki A. 2009. In vitro activity of WQ-3813 against nosocomial pathogens, abstr F1-1984. Abstr 49th Annu Intersci Conf Antimicrob Agents Chemother.
- 46.Eckburg PB, Ge Y, Jiang V, Kilfoil T, Talbot GH. 2012. Safety & pharmacokinetics of KPI-10, a novel fluoroquinolone, in healthy adults receiving single-dose oral administrations, abstr F-2047. Abstr 52nd Annu Intersci Conf Antimicrob Agents Chemother.
- 47.Sun C, Xiao X-Y. 2017. Fully synthetic tetracyclines: increasing chemical diversity to combat multidrug-resistant bacterial infections. Top Med Chem 26:55–95. 10.1007/7355_2017_11. [DOI] [Google Scholar]
- 48.Zhang W-Y, Sun C, Hunt D, He M, Deng Y, Zhu Z, Chen C-L, Katz CE, Niu J, Hogan PC, Xiao X-Y, Dunwoody N, Ronn M. 2016. Process development and scale-up of fully synthetic tetracycline TP-2758: a potent antibacterial agent with excellent oral bioavailability. Org Process Res Dev 20:284–296. 10.1021/acs.oprd.5b00404. [DOI] [Google Scholar]
- 49.Sun C, Deng Y, Hunt DK, Fyfe C, Chen C-L, Clark RB, Grossman TH, Sutcliffe JA, Xiao X-Y. 2018. Heterocyclyl tetracyclines. 2. 7-Methoxy-8-pyrrolidinyltetracyclines: discovery of TP-2758, a potent, orally efficacious antimicrobial against Gram-negative pathogens. J Antibiot (Tokyo) 71:287–297. 10.1038/ja.2017.86. [DOI] [PubMed] [Google Scholar]
- 50.Tetraphase Pharmaceuticals Inc. 2011. A phase 1, randomized, placebo-controlled, single-blind, multiple-dose, dose-escalation clinical study to assess the safety and pharmacokinetics of TP-2758 in normal healthy volunteers. EudraCT (number 2011-001204-35).
- 51.Trimble MJ, Mlynárčik P, Kolář M, Hancock REW. 2016. Polymyxin: alternative mechanisms of action and resistance. Cold Spring Harb Perspect Med 6:a025288. 10.1101/cshperspect.a025288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Koch-Weser J, Sidel VW, Federman EB, Kanarek P, Finer DC, Eaton AE. 1970. Adverse effects of sodium colistimethate. Manifestations and specific reaction rates during 317 courses of therapy. Ann Intern Med 72:857–868. 10.7326/0003-4819-72-6-857. [DOI] [PubMed] [Google Scholar]
- 53.Evans ME, Feola DJ, Rapp RP. 1999. Polymyxin B sulfate and colistin: old antibiotics for emerging multiresistant Gram-negative bacteria. Ann Pharmacother 33:960–967. 10.1345/aph.18426. [DOI] [PubMed] [Google Scholar]
- 54.Leese RA. July 2010. Antibiotic compositions for the treatment of Gram negative infections. World patent 2010075416.
- 55.Curran WV, Liu CM, Bombardier ACD, Leese RA, Hwang YS, Lippa BS, Zhang Y. April 2013. Antibiotic compositions for the treatment of Gram negative infections. US patent 8415307B1.
- 56.Quale J, Shah N, Kelly P, Babu E, Backer M, Rosas-Garcia G, Salamera J, George A, Bratu S, Landman D. 2012. Activity of polymyxin B and the novel polymyxin analogue CB-182,804 against contemporary Gram-negative pathogens in New York City. Microb Drug Resist 18:132–136. 10.1089/mdr.2011.0163. [DOI] [PubMed] [Google Scholar]
- 57.Arya A, Li T, Zhang S, Chuong L, Kang C, Zhang X, Mortin LI. 2010. Efficacy of CB-182,804, a novel polymyxin analog, in rat and mouse models of Gram-negative bacterial infections, abstr F-1627. Abstr 50th Annu Intersci Conf Antimicrob Agents Chemother.
- 58.Coleman S, Deats T, Pawliuk R, Chavan A, Oleson FB, Jr.. 2010. CB-182,804 is less nephrotoxic as compared to polymyxin B in monkeys following seven days of repeated intravenous dosing, poster F-1630/302. Abstr 50th Annu Intersci Conf Antimicrob Agents Chemother.
- 59.Blaskovich MAT, Pitt ME, Elliott AG, Cooper MA. 2018. Can octapeptin antibiotics combat extensively drug-resistant (XDR) bacteria? Expert Rev Anti Infect Ther 16:485–499. 10.1080/14787210.2018.1483240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vaara M, Siikanen O, Apajalahti J, Fox J, Frimodt-Møller N, He H, Poudyal A, Li J, Nation RL, Vaara T. 2010. A novel polymyxin derivative that lacks the fatty acid tail and carries only three positive charges has strong synergism with agents excluded by the intact outer membrane. Antimicrob Agents Chemother 54:3341–3346. 10.1128/AAC.01439-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Corbett D, Wise A, Langley T, Skinner K, Trimby E, Birchall S, Dorali A, Sandiford S, Williams J, Warn P, Vaara M, Lister T. 2017. Potentiation of antibiotic activity by a novel cationic peptide: potency and spectrum of activity of SPR741. Antimicrob Agents Chemother 61:e00200-17. 10.1128/AAC.00200-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Stainton SM, Abdelraouf K, Utley L, Pucci MJ, Lister T, Nicolau DP. 2018. Assessment of the in vivo activity of SPR741 in combination with azithromycin against multidrug-resistant Enterobacteriaceae isolates in the neutropenic murine thigh infection model. Antimicrob Agents Chemother 62:e00239-18. 10.1128/AAC.00239-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zurawski DV, Reinhart AA, Alamneh YA, Pucci MJ, Si Y, Abu-Taleb R, Shearer JP, Demons ST, Tyner SD, Lister T. 2017. SPR741, an antibiotic adjuvant, potentiates the in vitro and in vivo activity of rifampin against clinically relevant extensively drug-resistant Acinetobacter baumannii. Antimicrob Agents Chemother 61:e01239-17. 10.1128/AAC.01239-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Eckburg PB, Lister T, Walpole S, Keutzer T, Utley L, Tomayko J, Kopp E, Farinola N, Coleman S. 2019. Safety, tolerability, pharmacokinetics, and drug interaction potential of SPR741, an intravenous potentiator, after single and multiple ascending doses and when combined with β-lactam antibiotics in healthy subjects. Antimicrob Agents Chemother 63:e00892-19. 10.1128/AAC.00892-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Robinson JA, Shankaramma SC, Jetter P, Kienzl U, Schwendener RA, Vrijbloed JW, Obrecht D. 2005. Properties and structure-activity studies of cyclic β-hairpin peptidomimetics based on the cationic antimicrobial peptide protegrin I. Bioorg Med Chem 13:2055–2064. 10.1016/j.bmc.2005.01.009. [DOI] [PubMed] [Google Scholar]
- 66.Zerbe K, Moehle K, Robinson JA. 2017. Protein epitope mimetics: from new antibiotics to supramolecular synthetic vaccines. Acc Chem Res 50:1323–1331. 10.1021/acs.accounts.7b00129. [DOI] [PubMed] [Google Scholar]
- 67.Schmidt J, Patora-Komisarska K, Moehle K, Obrecht D, Robinson JA. 2013. Structural studies of β-hairpin peptidomimetic antibiotics that target LptD in Pseudomonas sp. Bioorg Med Chem 21:5806–5810. 10.1016/j.bmc.2013.07.013. [DOI] [PubMed] [Google Scholar]
- 68.Shankaramma SC, Athanassiou Z, Zerbe O, Moehle K, Mouton C, Bernardini F, Vrijbloed JW, Obrecht D, Robinson JA. 2002. Macrocyclic hairpin mimetics of the cationic antimicrobial peptide protegrin I: a new family of broad-spectrum antibiotics. Chembiochem 3:1126–1133. . [DOI] [PubMed] [Google Scholar]
- 69.Srinivas N, Jetter P, Ueberbacher BJ, Werneburg M, Zerbe K, Steinmann J, Van der Meijden B, Bernardini F, Lederer A, Dias RLA, Misson PE, Henze H, Zumbrunn J, Gombert FO, Obrecht D, Hunziker P, Schauer S, Ziegler U, Käch A, Eberl L, Riedel K, DeMarco SJ, Robinson JA. 2010. Peptidomimetic antibiotics target outer-membrane biogenesis in Pseudomonas aeruginosa. Science 327:1010–1013. 10.1126/science.1182749. [DOI] [PubMed] [Google Scholar]
- 70.Andolina G, Bencze L-C, Zerbe K, Müller M, Steinmann J, Kocherla H, Mondal M, Sobek J, Moehle K, Malojčić G, Wollscheid B, Robinson JA. 2018. A peptidomimetic antibiotic interacts with the periplasmic domain of LptD from Pseudomonas aeruginosa. ACS Chem Biol 13:666–675. 10.1021/acschembio.7b00822. [DOI] [PubMed] [Google Scholar]
- 71.Werneburg M, Zerbe K, Juhas M, Bigler L, Stalder U, Kaech A, Ziegler U, Obrecht D, Eberl L, Robinson JA. 2012. Inhibition of lipopolysaccharide transport to the outer membrane in Pseudomonas aeruginosa by peptidomimetic antibiotics. Chembiochem 13:1767–1775. 10.1002/cbic.201200276. [DOI] [PubMed] [Google Scholar]
- 72.Braun M, Silhavy TJ. 2002. Imp/OstA is required for cell envelope biogenesis in Escherichia coli. Mol Microbiol 45:1289–1302. 10.1046/j.1365-2958.2002.03091.x. [DOI] [PubMed] [Google Scholar]
- 73.Qiao S, Luo Q, Zhao Y, Zhang XC, Huang Y. 2014. Structural basis for lipopolysaccharide insertion in the bacterial outer membrane. Nature 511:108–111. 10.1038/nature13484. [DOI] [PubMed] [Google Scholar]
- 74.Dong H, Xiang Q, Gu Y, Wang Z, Paterson NG, Stansfeld PJ, He C, Zhang Y, Wang W, Dong C. 2014. Structural basis for outer membrane lipopolysaccharide insertion. Nature 511:52–56. 10.1038/nature13464. [DOI] [PubMed] [Google Scholar]
- 75.Wu T, McCandlish AC, Gronenberg LS, Chng S-S, Silhavy TJ, Kahne D. 2006. Identification of a protein complex that assembles lipopolysaccharide in the outer membrane of Escherichia coli. Proc Natl Acad Sci USA 103:11754–11759. 10.1073/pnas.0604744103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Sader HS, Dale GE, Rhomberg PR, Flamm RK. 2018. Antimicrobial activity of murepavadin tested against clinical isolates of Pseudomonas aeruginosa from the United States, Europe, and China. Antimicrob Agents Chemother 62:e00311-18. 10.1128/AAC.00311-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sader HS, Flamm RK, Dale GE, Rhomberg PR, Castanheira M. 2018. Murepavadin activity tested against contemporary (2016–17) clinical isolates of XDR Pseudomonas aeruginosa. J Antimicrob Chemother 73:2400–2404. 10.1093/jac/dky227. [DOI] [PubMed] [Google Scholar]
- 78.Martin-Loeches I, Dale GE, Torres A. 2018. Murepavadin: a new antibiotic class in the pipeline. Expert Rev Anti Infect Ther 16:259–268. 10.1080/14787210.2018.1441024. [DOI] [PubMed] [Google Scholar]
- 79.Wach A, Dembowsky K, Dale GE. 2018. Pharmacokinetics and safety of intravenous murepavadin infusion in healthy adult subjects administered single and multiple ascending doses. Antimicrob Agents Chemother 62:e02355-17. 10.1128/AAC.02355-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hoffmann-La Roche. 2014. A single center study to evaluate the penetration of RO7033877 into the lung in healthy volunteers. ClinicalTrials.gov (identifier NCT02165293).
- 81.Hoffmann-La Roche. 2014. A phase 1 clinical study in healthy volunteers to investigate the drug-drug interaction between multiple doses of RO7033877 and multiple doses of colistin methanesulfonate sodium (CMS). ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02156323 (identifier NCT02156323). Accessed 3 February 2021.
- 82.Polyphor Ltd. 2016. DDI study to investigate interaction between amikacin and POL7080. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02897869 (identifier NCT02897869). Accessed 3 February 2021.
- 83.Hoffmann-La Roche. 2014. A two-part, single-dose, randomized study to evaluate the safety of supra-therapeutic doses of RO7033877 and to investigate the effect of RO7033877 on the QTc interval. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02165332 (identifier NCT02165332). Accessed 3 February 2021.
- 84.Polyphor Ltd. 2013. Pharmacokinetics and safety of POL7080 in patients with renal impairment. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02110459 (identifier NCT02110459). Accessed 4 February 2021.
- 85.Dale GE, Halabi A, Petersen-Sylla M, Wach A, Zwingelstein C. 2018. Pharmacokinetics, tolerability, and safety of murepavadin, a novel antipseudomonal antibiotic, in subjects with mild, moderate, or severe renal function impairment. Antimicrob Agents Chemother 62:e00490-18. 10.1128/AAC.00490-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Polyphor Ltd. 2013. Safety, efficacy and PK/PD of POL7080 in patients with exacerbation of non-cystic fibrosis bronchiectasis. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02096315 (identifier NCT02096315). Accessed 4 February 2021.
- 87.Polyphor Ltd. 2013. Pharmacokinetics, safety and efficacy of POL7080 in patients with ventilator associated Pseudomonas aeruginosa pneumonia. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02096328 (identifier NCT02096328). Accessed 4 February 2021.
- 88.Armaganidis A, Frantzeskaki F, Diakaki C, Apostolopoulou O, Zakynthinos S, Ischaki E, Mandragos C, Katsenos C, Paraschos M, Patrani M, Amygdalou A, Giamarellos-Bourboulis EJ, Pistiki A, Tsaganos T, Antonakos N, Damoraki G, Ramirez P, de la Torre-Prados MV, Rodriguez A, Sommerville K, Wach A, Zwingelstein C, Beni L, Hooftman L, Dale GE, Torres A. 2017. Pharmacokinetic and efficacy analysis of murepavadin (POL7080) coadministered with standard-of-care (SOC) in a phase II study in patients with ventilator-associated pneumonia (VAP) due to suspected or documented Pseudomonas aeruginosa infection, abstr 2720. Abstr 27th Eur Congr Clin Microbiol Infect Dis.
- 89.Bader JC, Lakota EA, Dale GE, Sader HS, Rex JH, Ambrose PG, Bhavnani SM. 2019. Pharmacokinetic-pharmacodynamic evaluation of ertapenem for patients with hospital-acquired or ventilator-associated bacterial pneumonia. Antimicrob Agents Chemother 63:e00318-19. 10.1128/AAC.00318-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Polyphor Ltd. 2018. Pivotal study in nosocomial pneumonia suspected or confirmed to be due to Pseudomonas. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03582007 (identifier NCT03582007). Accessed 4 February 2021.
- 91.Polyphor Ltd. 2018. Pivotal study in VAP suspected or confirmed to be due to Pseudomonas aeruginosa. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03409679 (identifier NCT03409679). Accessed 4 February 2021.
- 92.Polyphor Ltd. 2019. Polyphor temporarily halts enrollment in the phase III studies of murepavadin for the treatment of patients with nosocomial pneumonia. Polyphor Ltd, Allschwil, Switzerland. https://www.polyphor.com/news/corporate-news-details/?newsid=1775911. [Google Scholar]
- 93.Erwin AL. 2016. Antibacterial drug discovery targeting the lipopolysaccharide biosynthetic enzyme LpxC. Cold Spring Harb Perspect Med 6:a025304. 10.1101/cshperspect.a025304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kalinin DV, Holl R. 2017. LpxC inhibitors: a patent review (2010-2016). Expert Opin Ther Pat 27:1227–1250. 10.1080/13543776.2017.1360282. [DOI] [PubMed] [Google Scholar]
- 95.Zhang J, Zhang L, Li X, Xu W. 2012. UDP-3-O-(R-3-Hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) inhibitors: a new class of antibacterial agents. Curr Med Chem 19:2038–2050. 10.2174/092986712800167374. [DOI] [PubMed] [Google Scholar]
- 96.Mdluli KE, Witte PR, Kline T, Barb AW, Erwin AL, Mansfield BE, McClerren AL, Pirrung MC, Tumey LN, Warrener P, Raetz CRH, Stover CK. 2006. Molecular validation of LpxC as an antibacterial drug target in Pseudomonas aeruginosa. Antimicrob Agents Chemother 50:2178–2184. 10.1128/AAC.00140-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Krause KM, Haglund CM, Hebner C, Serio AW, Lee G, Nieto V, Cohen F, Kane TR, Machajewski TD, Hildebrandt D, Pillar C, Thwaites M, Hall D, Miesel L, Hackel M, Burek A, Andrews LD, Armstrong E, Swem L, Jubb A, Cirz RT. 2019. Potent LpxC inhibitors with activity against multidrug-resistant Pseudomonas aeruginosa. Antimicrob Agents Chemother 63:e00977-19. 10.1128/AAC.00977-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bornheim L, McKinnell J, Fuchs-Knotts T, Boggs J, Kostrub CF. 2013. Preclinical safety evaluation of the novel LpxC inhibitor ACHN-975 in rat and monkey, abstr F-1231. Abstr 53rd Annu Intersci Conf Antimicrob Agents Chemother.
- 99.Achaogen Inc. 2012. A study to assess the safety, tolerability, and pharmacokinetics of ACHN-975 in healthy volunteers. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01597947 (identifier NCT01597947). Accessed 4 February 2021.
- 100.Cohen F, Aggen JB, Andrews LD, Assar Z, Boggs J, Choi T, Dozzo P, Easterday AN, Haglund CM, Hildebrandt DJ, Holt MC, Joly K, Jubb A, Kamal Z, Kane TR, Konradi AW, Krause KM, Linsell MS, Machajewski TD, Miroshnikova O, Moser HE, Nieto V, Phan T, Plato C, Serio AW, Seroogy J, Shakhmin A, Stein AJ, Sun AD, Sviridov S, Wang Z, Wlasichuk K, Yang W, Zhou X, Zhu H, Cirz RT. 2019. Optimization of LpxC inhibitors for antibacterial activity and cardiovascular safety. ChemMedChem 14:1560–1572. 10.1002/cmdc.201900287. [DOI] [PubMed] [Google Scholar]
- 101.Achaogen Inc. 2013. A multiple dose study to assess the safety, tolerability, and pharmacokinetics of ACHN-975 in healthy volunteers. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01870245 (identifier NCT01870245). Accessed 4 February 2021.
- 102.Shoji M, Suzumura Y, Fujiwara M, Kurosaki C, Nozaki Y, Mizunaga S. 2019. T-1228 (RC-01), a new LpxC inhibitor exhibiting potent activity against multi-drug resistant Gram-negative pathogens: design, synthesis and structure-activity relationship, abstr AAR-740. Abstr ASM Microbe 2019.
- 103.Fujiwara M, Nakagawa S, Mizunaga S, Eto M. 2019. Inhibition of LpxC activity and elimination of lipopolysaccharide (LPS) release by T-1228 (RC-01), abstr AAR-741. Abstr ASM Microbe 2019.
- 104.Lin L, Tan B, Pantapalangkoor P, Ho T, Baquir B, Tomaras A, Montgomery JI, Reilly U, Barbacci EG, Hujer K, Bonomo RA, Fernandez L, Hancock REW, Adams MD, French SW, Buslon VS, Spellberg B. 2012. Inhibition of LpxC protects mice from resistant Acinetobacter baumannii by modulating inflammation and enhancing phagocytosis. mBio 3:e00312-12. 10.1128/mBio.00312-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Nakagawa S, Fujiwara M, Eto M, Mizunaga S. 2019. PK/PD analysis of T-1228 (RC-01), a novel LpxC inhibitor, using murine pneumonia and urinary tract infection models, abstr AAR-751. Abstr ASM Microbe 2019.
- 106.Mizunaga S, Fujiwara M, Nakagawa S, Eto M. 2019. In vitro resistance development of T-1228 (RC-01), a novel LpxC inhibitor, compared with other agents, abstr AAR-744. Abstr ASM Microbe 2019.
- 107.Ge Y, Nozaki Y, Mochizuki S. 2019. Evaluation of the general preclinical toxicological profile of RC-01, a novel LpxC inhibitor, abstr AAR-748. Abstr ASM Microbe 2019.
- 108.Recida Therapeutics Inc. 2019. Single and multiple dose escalation trial of an intravenous antibiotic RC-01. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03832517 (identifier NCT03832517). Accessed 4 February 2021.
- 109.Baker SJ, Hernandez VS, Sharma R, Nieman JA, Akama T, Zhang Y-K, Plattner JJ, Alley MRK, Singh R, Rock F. 2010. Boron-containing small molecules. US patent 7816344B2.
- 110.Hudson IR. 1994. The efficacy of intranasal mupirocin in the prevention of staphylococcal infections: a review of recent experience. J Hosp Infect 27:81–98. 10.1016/0195-6701(94)90001-9. [DOI] [PubMed] [Google Scholar]
- 111.Sutherland R, Boon RJ, Griffin KE, Masters PJ, Slocombe B, White AR. 1985. Antibacterial activity of mupirocin (pseudomonic acid), a new antibiotic for topical use. Antimicrob Agents Chemother 27:495–498. 10.1128/AAC.27.4.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ochsner UA, Sun X, Jarvis T, Critchley I, Janjic N. 2007. Aminoacyl-tRNA synthetases: essential and still promising targets for new anti-infective agents. Expert Opin Investig Drugs 16:573–593. 10.1517/13543784.16.5.573. [DOI] [PubMed] [Google Scholar]
- 113.Walker ES, Vasquez JE, Dula R, Bullock H, Sarubbi FA. 2003. Mupirocin-resistant, methicillin-resistant Staphylococcus aureus: does mupirocin remain effective? Infect Control Hosp Epidemiol 24:342–346. 10.1086/502218. [DOI] [PubMed] [Google Scholar]
- 114.Rock FL, Mao W, Yaremchuk A, Tukalo M, Crépin T, Zhou H, Zhang Y-K, Hernandez V, Akama T, Baker SJ, Plattner JJ, Shapiro L, Martinis SA, Benkovic SJ, Cusack S, Alley MRK. 2007. An antifungal agent inhibits an aminoacyl-tRNA synthetase by trapping tRNA in the editing site. Science 316:1759–1761. 10.1126/science.1142189. [DOI] [PubMed] [Google Scholar]
- 115.Hernandez V, Crépin T, Palencia A, Cusack S, Akama T, Baker SJ, Bu W, Feng L, Freund YR, Liu L, Meewan M, Mohan M, Mao W, Rock FL, Sexton H, Sheoran A, Zhang Y, Zhang Y-K, Zhou Y, Nieman JA, Anugula MR, Keramane EM, Savariraj K, Reddy DS, Sharma R, Subedi R, Singh R, O’Leary A, Simon NL, De Marsh PL, Mushtaq S, Warner M, Livermore DM, Alley MRK, Plattner JJ. 2013. Discovery of a novel class of boron-based antibacterials with activity against Gram-negative bacteria. Antimicrob Agents Chemother 57:1394–1403. 10.1128/AAC.02058-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Mendes RE, Alley MRK, Sader HS, Biedenbach DJ, Jones RN. 2013. Potency and spectrum of activity of AN3365, a novel boron-containing protein synthesis inhibitor, tested against clinical isolates of Enterobacteriaceae and nonfermentative Gram-negative bacilli. Antimicrob Agents Chemother 57:2849–2857. 10.1128/AAC.00160-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Goldstein EJC, Citron DM, Tyrrell KL, Merriam CV. 2013. Comparative in vitro activities of GSK2251052, a novel boron-containing leucyl-tRNA synthetase inhibitor, against 916 anaerobic organisms. Antimicrob Agents Chemother 57:2401–2404. 10.1128/AAC.02580-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bouchillon SK, Hackel M, Hoban DJ, Hawser SP, Scangarella-Oman NE. 2011. GSK2251052, a novel boron-containing protein synthesis inhibitor, with comparative in vitro activity against Pseudomonas aeruginosa from a global population, poster E-129. Abstr 51st Annu Intersci Conf Antimicrob Agents Chemother.
- 119.Pfizer. 2009. Safety, pharmacokinetics (PK) and tolerability study of a novel drug for treatment of bacterial infections. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01015014 (identifier NCT01015014). Accessed 15 February 2021.
- 120.GlaxoSmithKline. 2010. A randomized, single blind, placebo controlled study to evaluate safety, tolerability, and pharmacokinetics of single oral doses and repeat escalating oral doses of GSK2251052 in healthy adult subjects. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01262885 (identifier NCT01262885). Accessed 4 February 2021.
- 121.GlaxoSmithKline. 2011. GSK2251052 mass balance in healthy adult subjects. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01475695 (identifier NCT01475695). Accessed 4 February 2021.
- 122.GlaxoSmithKline. 2011. An open-label, randomized, single period, parallel-cohort study to evaluate serum and pulmonary pharmacokinetics following single and multiple dose administration of intravenous GSK2251052 in healthy adult subjects. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01267968 (identifier NCT01267968). Accessed 4 February 2021.
- 123.Bowers GD, Tenero D, Patel P, Huynh P, Sigafoos J, O’Mara K, Young GC, Dumont E, Cunningham E, Kurtinecz M, Stump P, Conde JJ, Chism JP, Reese MJ, Yueh YL, Tomayko JF. 2013. Disposition and metabolism of GSK2251052 in humans: a novel boron-containing antibiotic. Drug Metab Dispos 41:1070–1081. 10.1124/dmd.112.050153. [DOI] [PubMed] [Google Scholar]
- 124.GlaxoSmithKline. 2011. GSK2251052 in complicated urinary tract infection. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01381549 (identifier NCT01381549). Accessed 4 February 2021.
- 125.GlaxoSmithKline. 2011. GSK2251052 in the treatment of complicated intra-abdominal infections. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01381562 (identifier NCT01381562). Accessed 4 February 2021.
- 126.O’Dwyer K, Spivak AT, Ingraham K, Min S, Holmes DJ, Jakielaszek C, Rittenhouse S, Kwan AL, Livi GP, Sathe G, Thomas E, Van Horn S, Miller LA, Twynholm M, Tomayko J, Dalessandro M, Caltabiano M, Scangarella-Oman NE, Brown JR. 2015. Bacterial resistance to leucyl-tRNA synthetase inhibitor GSK2251052 develops during treatment of complicated urinary tract infections. Antimicrob Agents Chemother 59:289–298. 10.1128/AAC.03774-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Tenero D, Bowers G, Rodvold KA, Patel A, Kurtinecz M, Dumont E, Tomayko J, Patel P. 2013. Intrapulmonary pharmacokinetics of GSK2251052 in healthy volunteers. Antimicrob Agents Chemother 57:3334–3339. 10.1128/AAC.02483-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Farrell LJ, Lo R, Wanford JJ, Jenkins A, Maxwell A, Piddock LJV. 2018. Revitalizing the drug pipeline: AntibioticDB, an open access database to aid antibacterial research and development. J Antimicrob Chemother 73:2284–2297. 10.1093/jac/dky208. [DOI] [PubMed] [Google Scholar]
- 129.Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. 2014. Clinical development success rates for investigational drugs. Nat Biotechnol 32:40–51. 10.1038/nbt.2786. [DOI] [PubMed] [Google Scholar]
- 130.Gajdács M. 2019. The concept of an ideal antibiotic: implications for drug design. Molecules 24:892. 10.3390/molecules24050892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Singh SB, Young K, Silver LL. 2017. What is an “ideal” antibiotic? discovery challenges and path forward. Biochem Pharmacol 133:63–73. 10.1016/j.bcp.2017.01.003. [DOI] [PubMed] [Google Scholar]
- 132.Dalvie D, Cosker T, Boyden T, Zhou S, Schroeder C, Potchoiba MJ. 2008. Metabolism distribution and excretion of a matrix metalloproteinase-13 inhibitor, 4-[4-(4-fluorophenoxy)-benzenesulfonylamino]tetrahydropyran-4-carboxylic acid hydroxyamide (CP-544439), in rats and dogs: assessment of the metabolic profile of CP-544439 in plasma and urine of humans. Drug Metab Dispos 36:1869–1883. 10.1124/dmd.108.022566. [DOI] [PubMed] [Google Scholar]
- 133.Sixto-López Y, Gómez-Vidal JA, de Pedro N, Bello M, Rosales-Hernández MC, Correa-Basurto J. 2020. Hydroxamic acid derivatives as HDAC1, HDAC6 and HDAC8 inhibitors with antiproliferative activity in cancer cell lines. Sci Rep 10:10462. 10.1038/s41598-020-67112-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Lee MS, Isobe M. 1990. Metabolic activation of the potent mutagen, 2-naphthohydroxamic acid, in Salmonella typhimurium TA98. Cancer Res 50:4300–4307. [PubMed] [Google Scholar]
- 135.Galster M, Löppenberg M, Galla F, Börgel F, Agoglitta O, Kirchmair J, Holl R. 2019. Phenylethylene glycol-derived LpxC inhibitors with diverse Zn2+-binding groups. Tetrahedron 75:486–509. 10.1016/j.tet.2018.12.011. [DOI] [Google Scholar]
- 136.Furuya T, Shapiro AB, Comita-Prevoir J, Kuenstner EJ, Zhang J, Ribe SD, Chen A, Hines D, Moussa SH, Carter NM, Sylvester MA, Romero JAC, Vega CV, Sacco MD, Chen Y, O’Donnell JP, Durand-Reville TF, Miller AA, Tommasi RA. 2020. N-Hydroxyformamide LpxC inhibitors, their in vivo efficacy in a mouse Escherichia coli infection model, and their safety in a rat hemodynamic assay. Bioorg Med Chem 28:115826. 10.1016/j.bmc.2020.115826. [DOI] [PubMed] [Google Scholar]
- 137.Huang JX, Blaskovich MA, Cooper MA. 2014. Cell- and biomarker-based assays for predicting nephrotoxicity. Expert Opin Drug Metab Toxicol 10:1621–1635. 10.1517/17425255.2014.967681. [DOI] [PubMed] [Google Scholar]
- 138.Gallardo-Godoy A, Muldoon C, Becker B, Elliott AG, Lash LH, Huang JX, Butler MS, Pelingon R, Kavanagh AM, Ramu S, Phetsang W, Blaskovich MAT, Cooper MA. 2016. Activity and predicted nephrotoxicity of synthetic antibiotics based on polymyxin B. J Med Chem 59:1068–1077. 10.1021/acs.jmedchem.5b01593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Roberts KD, Azad MAK, Wang J, Horne AS, Thompson PE, Nation RL, Velkov T, Li J. 2015. Antimicrobial activity and toxicity of the major lipopeptide components of polymyxin B and colistin: last-line antibiotics against multidrug-resistant Gram-negative bacteria. ACS Infect Dis 1:568–575. 10.1021/acsinfecdis.5b00085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Velkov T, Roberts KD, Thompson PE, Li J. 2016. Polymyxins: a new hope in combating Gram-negative superbugs? Future Med Chem 8:1017–1025. 10.4155/fmc-2016-0091. [DOI] [PubMed] [Google Scholar]
- 141.Wenzler E, Fraidenburg DR, Scardina T, Danziger LH. 2016. Inhaled antibiotics for Gram-negative respiratory infections. Clin Microbiol Rev 29:581–632. 10.1128/CMR.00101-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Papp-Wallace KM. 2019. The latest advances in β-lactam/β-lactamase inhibitor combinations for the treatment of Gram-negative bacterial infections. Expert Opin Pharmacother 20:2169–2184. 10.1080/14656566.2019.1660772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Verbist L. 1985. In-vitro activity of the combinations of ampicillin with mecillinam or with β-lactamase inhibitors against strains resistant to ampicillin. J Antimicrob Chemother 16:719–725. 10.1093/jac/16.6.719. [DOI] [PubMed] [Google Scholar]
- 144.Morinaka A, Tsutsumi Y, Yamada M, Suzuki K, Watanabe T, Abe T, Furuuchi T, Inamura S, Sakamaki Y, Mitsuhashi N, Ida T, Livermore DM. 2015. OP0595, a new diazabicyclooctane: mode of action as a serine β-lactamase inhibitor, antibiotic and β-lactam “enhancer.” J Antimicrob Chemother 70:2779–2786. 10.1093/jac/dkv166. [DOI] [PubMed] [Google Scholar]
- 145.Zusman O, Avni T, Leibovici L, Adler A, Friberg L, Stergiopoulou T, Carmeli Y, Paul M. 2013. Systematic review and meta-analysis of in vitro synergy of polymyxins and carbapenems. Antimicrob Agents Chemother 57:5104–5111. 10.1128/AAC.01230-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lenhard JR, Nation RL, Tsuji BT. 2016. Synergistic combinations of polymyxins. Int J Antimicrob Agents 48:607–613. 10.1016/j.ijantimicag.2016.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Rabanal F, Cajal Y. 2017. Recent advances and perspectives in the design and development of polymyxins. Nat Prod Rep 34:886–908. 10.1039/c7np00023e. [DOI] [PubMed] [Google Scholar]
- 148.Boucher HW, Ambrose PG, Chambers HF, Ebright RH, Jezek A, Murray BE, Newland JG, Ostrowsky B, Rex JH, Infectious Diseases Society of America. 2017. White paper: developing antimicrobial drugs for resistant pathogens, narrow-spectrum indications, and unmet needs. J Infect Dis 216:228–236. 10.1093/infdis/jix211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Rex JH, Talbot GH, Goldberger MJ, Eisenstein BI, Echols RM, Tomayko JF, Dudley MN, Dane A. 2017. Progress in the fight against multidrug-resistant bacteria 2005–2016: modern noninferiority trial designs enable antibiotic development in advance of epidemic bacterial resistance. Clin Infect Dis 65:141–146. 10.1093/cid/cix246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Bassetti M, Echols R, Matsunaga Y, Ariyasu M, Doi Y, Ferrer R, Lodise TP, Naas T, Niki Y, Paterson DL, Portsmouth S, Torre-Cisneros J, Toyoizumi K, Wunderink RG, Nagata TD. 2021. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): a randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect Dis 21:226–240. 10.1016/S1473-3099(20)30796-9. [DOI] [PubMed] [Google Scholar]
- 151.Kim W, Krause K, Zimmerman Z, Outterson K. 2021. Improving data sharing to increase the efficiency of antibiotic R&D. Nat Rev Drug Discov 20:1–2. 10.1038/d41573-020-00185-y. [DOI] [PubMed] [Google Scholar]