MedPath

Epetraborole Advanced Drug Monograph

Published:Oct 30, 2025

Generic Name

Epetraborole

Drug Type

Small Molecule

Chemical Formula

C11H16BNO4

CAS Number

1093643-37-8

Epetraborole (DB11744): A Comprehensive Monograph on a Novel Boron-Based Antibiotic

Executive Summary of Epetraborole

Epetraborole is a first-in-class, orally bioavailable, investigational small molecule antibiotic distinguished by its boron-containing benzoxaborole chemical structure.[1] It represents a significant innovation in the field of antimicrobial therapy, primarily due to its novel mechanism of action. The compound is a potent and selective inhibitor of bacterial leucyl-tRNA synthetase (LeuRS), an essential enzyme in protein synthesis.[1] Epetraborole functions via a unique Oxaborole tRNA-Trapping (OBORT) mechanism, forming a stable adduct with transfer RNA for leucine (tRNALeu) within the enzyme's editing site, which effectively halts bacterial protein production—a mode of action distinct from all currently approved antibiotic classes.[3]

The drug exhibits a broad spectrum of activity, with notable potency against several difficult-to-treat pathogens. Its development has primarily focused on nontuberculous mycobacteria (NTM), including Mycobacterium avium complex (MAC) and the highly drug-resistant Mycobacterium abscessus, as well as the Gram-negative pathogen Burkholderia pseudomallei, the causative agent of the severe tropical disease melioidosis.[1]

Epetraborole's clinical development has been marked by both significant promise and notable setbacks. Initially developed by GlaxoSmithKline (GSK) for Gram-negative infections, including complicated urinary tract and intra-abdominal infections, this line of investigation was discontinued following a Phase II trial that revealed the rapid emergence of on-treatment bacterial resistance.[5] Subsequently, AN2 Therapeutics acquired the asset and strategically pivoted its development toward NTM lung disease, a rare and progressive condition with a high unmet medical need. This strategy was bolstered by strong preclinical data and resulted in the granting of multiple favorable regulatory designations from the U.S. Food and Drug Administration (FDA) and European authorities, including Orphan Drug, Qualified Infectious Disease Product (QIDP), and Fast Track status.[8]

The cornerstone of the NTM program was the pivotal EBO-301 Phase 2/3 clinical trial in patients with treatment-refractory MAC lung disease. However, in 2024, this trial was terminated. While the study demonstrated a positive signal on a patient-reported outcome (PRO) measure, indicating that patients felt better, it failed to meet its key microbiological endpoint of sputum culture conversion.[11] This critical failure to demonstrate a bacteriological cure in a severely ill patient population led to the discontinuation of the trial.

Despite this significant setback, the development of Epetraborole is not entirely concluded. Its potential remains under active evaluation for other indications where its unique attributes may offer a distinct advantage. These include the notoriously difficult-to-treat M. abscessus lung disease, where its exceptional in vitro potency may overcome therapeutic challenges, and acute melioidosis, where it could be used in combination with standard-of-care antibiotics to reduce the disease's high mortality rate.[3] The developer, AN2 Therapeutics, has concurrently shifted its corporate focus to leverage its broader boron chemistry platform for other therapeutic areas while exploring non-dilutive funding pathways to potentially continue the targeted development of Epetraborole.[11]

Molecular Profile and Physicochemical Properties

Chemical Identity and Nomenclature

Epetraborole is a synthetic organic small molecule that has been systematically cataloged across major chemical and pharmacological databases to ensure unambiguous identification for research, regulatory, and clinical purposes. Its primary identifiers are:

  • DrugBank ID: DB11744 [16]
  • CAS Number: 1093643-37-8 (for the free base form).[17] The compound has also been formulated as hydrochloride (CAS 1234563-16-6) and R-mandelate (CAS 1234563-15-5) salts.[20]
  • Synonyms and Developmental Codes: The compound is widely known by its former developmental codes, including GSK2251052 (from its time at GlaxoSmithKline), AN3365, and BRII-658.[2]
  • IUPAC Name: The formal chemical name under International Union of Pure and Applied Chemistry nomenclature is 3-oxy]propan-1-ol.[2]
  • Other Key Identifiers: Additional identifiers include its FDA Unique Ingredient Identifier (UNII) 6MC93Z2DF9, ChEMBL ID CHEMBL3549142, and NCI Thesaurus Code C166950.[17]

Structural Analysis and Boron Chemistry

Epetraborole's chemical architecture is central to its novel mechanism of action. It is classified as an aminomethylbenzoxaborole, a class of boron-heterocyclic compounds.[2] Its two-dimensional structure is unambiguously represented by the following:

  • SMILES (Simplified Molecular Input Line Entry Specification): NC[C@H]1OB(O)c2c(OCCCO)cccc12 [20]
  • InChIKey (International Chemical Identifier Key): FXQIIDINBDJDKL-SNVBAGLBSA-N [20]

The potent and specific activity of Epetraborole is not accidental but rather the result of rational drug design. Specific structural modifications to the core benzoxaborole scaffold were engineered to optimize interactions with the bacterial LeuRS enzyme and its tRNA substrate.[5] Two modifications are particularly critical:

  1. The 3-aminomethyl group: This substituent was added to the benzoxaborole core to enhance interactions with the amino acid editing domain of the Escherichia coli LeuRS enzyme.[5]
  2. The 7-O-propanol substituent: This side chain introduces a novel point of contact, allowing the molecule to interact with the phosphate backbone of the tRNALeu molecule itself.[5]

The integration of a boron atom into the heterocyclic ring system is the key chemical feature that enables this unique biological activity. Boron's distinct chemistry allows for the formation of a stable, reversible covalent bond with the diol group of the terminal adenosine ribose on the tRNA molecule, an interaction not readily achievable with traditional carbon-based molecules.[3] This targeted chemical design, while creating a highly potent inhibitor, may also contribute to a very specific and therefore potentially narrow pathway for the development of resistance. If the primary mechanism of action relies on such precise molecular interactions, mutations at the specific binding site on the target enzyme can readily abrogate the drug's effect, a phenomenon that was later observed in clinical settings.[5]

Physicochemical and "Drug-Likeness" Properties

Epetraborole's physicochemical properties are consistent with those of an orally bioavailable drug candidate, aligning with its intended route of administration for chronic infections like NTM lung disease. The compound is also classified as a phenol ether, which describes the aromatic ring substituted with an ether group.[16] A summary of its key properties is provided in Table 1.

Table 1: Epetraborole Key Identifiers and Physicochemical Properties

PropertyValueSource(s)
Molecular Formula$C_{11}H_{16}BNO_{4}$17
Molecular Weight237.06 g/mol[4, 16, 19]
Topological Polar Surface Area (TPSA)84.9 Ų[4, 17]
Hydrogen Bond Acceptors54
Hydrogen Bond Donors34
Rotatable Bonds54
XLogP-0.834
Lipinski's Rule-of-Five Violations04

An analysis of these properties reveals that Epetraborole adheres to Lipinski's Rule-of-Five, a widely used guideline for predicting the "drug-likeness" of a molecule in terms of its potential for oral absorption and intestinal permeability.[4] The lack of any violations supports its development as an oral agent.

Mechanism of Action and Antimicrobial Spectrum

The LeuRS Target: A Novel Approach to Antibiosis

Epetraborole's antibacterial effect stems from its highly specific and potent inhibition of bacterial leucyl-tRNA synthetase (LeuRS).[1] LeuRS is a member of the aminoacyl-tRNA synthetase family of enzymes, which are essential for life in all organisms. These enzymes perform the critical first step of protein synthesis: catalyzing the covalent attachment of a specific amino acid (in this case, leucine) to its cognate transfer RNA (tRNA) molecule.[1] By targeting this fundamental process, Epetraborole effectively shuts down the production of all proteins within the bacterium, leading to a cessation of growth and, ultimately, cell death. The half-maximal inhibitory concentration ($IC_{50}$) of Epetraborole against the LeuRS enzyme has been measured at 0.31 µM, indicating high potency.[22]

The Oxaborole tRNA-Trapping (OBORT) Mechanism

The molecular mechanism by which Epetraborole inhibits LeuRS is known as the Oxaborole tRNA-Trapping (OBORT) mechanism, a sophisticated process that exploits the enzyme's natural function.[3] The mechanism can be described in a stepwise fashion:

  1. Enzyme Entry: Epetraborole enters the active site of the bacterial LeuRS enzyme.
  2. Adduct Formation: Inside the enzyme, the boron atom of Epetraborole forms a stable, covalent bond with the 2'- and 3'-hydroxyl groups of the terminal adenosine ribose of an uncharged tRNALeu molecule. This creates a novel, tripartite Epetraborole-tRNALeu-adenosine adduct.[3]
  3. Trapping in the Editing Site: This newly formed adduct is then trapped within the editing domain of the LeuRS enzyme. The editing domain is a proofreading site that normally functions to remove incorrectly charged amino acids from tRNA, ensuring the fidelity of protein synthesis.[3]
  4. Inhibition of Synthesis: The physical presence of the trapped adduct in the editing site sterically hinders the enzyme's primary synthetic site. This blockage prevents the enzyme from catalyzing the attachment of leucine to other tRNALeu molecules, effectively shutting down the entire leucylation process and halting bacterial protein synthesis.[3]

This mechanism is novel among antibiotics and represents a new paradigm for inhibiting bacterial growth, enabled specifically by the unique chemical properties of boron.[3]

Spectrum of Activity: A Focus on Difficult-to-Treat Pathogens

Epetraborole has demonstrated a broad spectrum of antibacterial activity, but its clinical development has strategically focused on pathogens for which new therapeutic options are urgently needed. A comparative summary of its in vitro potency is presented in Table 2.

Table 2: Summary of Epetraborole In Vitro Activity (MIC50/MIC90) Against Key Pathogens

PathogenMIC50 (µg/mL)MIC90 (µg/mL)Source(s)
Mycobacterium abscessus-0.1226
Mycobacterium avium complex (MAC)2426
Mycobacterium tuberculosis23228
Burkholderia pseudomalleiData not availableData not available-

Nontuberculous Mycobacteria (NTM)

  • Mycobacterium abscessus (Mab): Epetraborole exhibits exceptional potency against M. abscessus, a pathogen notorious for its high intrinsic and inducible drug resistance.[5] It is active against all subspecies, clinical isolates, and both the smooth (S) and rough (R) morphotypes.[5] The minimum inhibitory concentration required to inhibit 90% of isolates (MIC90) was reported to be a remarkably low 0.12 µg/mL against a panel of 147 international clinical isolates.[26] Furthermore, it effectively inhibits the growth of M. abscessus residing within macrophages and has demonstrated superior efficacy to comparator agents like clarithromycin in murine infection models.[5]
  • Mycobacterium avium complex (MAC):* The drug also shows potent in vitro activity against MAC, the most common cause of NTM lung disease.[1] Studies on clinical isolates from both the United States and Japan have consistently found MIC50 and MIC90 values of 2 µg/mL and 4 µg/mL, respectively.[26] Crucially, its activity is retained against isolates that are resistant to clarithromycin, a cornerstone of current MAC therapy.[27] In chronic mouse lung infection models, Epetraborole monotherapy was highly effective, and its addition to the standard-of-care (SOC) regimen significantly enhanced bacterial killing.[31]

Gram-Negative Pathogens

  • Burkholderia pseudomallei (Melioidosis):* Epetraborole has demonstrated potent activity against B. pseudomallei, the causative agent of melioidosis, a severe and often fatal infection.[1] Preclinical studies in murine models have shown robust, dose-dependent efficacy against a wide range of genetically and geographically diverse strains, validating its potential as a new therapeutic agent for this neglected tropical disease.[13]
  • Other Gram-Negative Bacteria: The drug's initial development targeted multidrug-resistant Gram-negative pathogens, such as those in the Enterobacteriaceae family.[19] However, this avenue was abandoned after clinical trials revealed the rapid emergence of resistance, rendering it unsuitable for these indications.[5]

Mycobacterium tuberculosis (Mtb)

Epetraborole is active in vitro against M. tuberculosis, with MIC values against clinical isolates ranging from 0.25 to 64 mg/L and a high MIC90 of 32 mg/L.[28] Despite this activity, its development for tuberculosis (TB) was not pursued. This decision underscores a critical principle in antibiotic development: clinical viability is determined not by raw potency alone, but by the achievable therapeutic window. Preclinical pharmacokinetic/pharmacodynamic (PK/PD) modeling and simulations predicted that the drug exposures (Area Under the Curve, or AUC) required to meet the therapeutic target against the high MICs of Mtb would likely be unattainable at doses that are safe for human administration.[28] This analysis effectively closed the door on its development for TB and provides a crucial framework for understanding its strategic redirection toward pathogens with much greater susceptibility, such as M. abscessus, where the therapeutic target can be reached with a much lower, safer dose.

Preclinical and Clinical Pharmacology: Pharmacokinetics and Pharmacodynamics

Pharmacokinetics (PK): ADME Profile

The pharmacokinetic profile of Epetraborole has been characterized through multiple Phase 1 clinical trials in healthy volunteers, defining its absorption, distribution, metabolism, and excretion (ADME) properties. A summary of key human PK parameters is presented in Table 3.

Table 3: Summary of Human Pharmacokinetic Parameters from Phase 1 Studies

ParameterValueDose/ConditionsSource(s)
Terminal Half-Life (T1/2)~6–11 hours500 mg oral QD26
Volume of Distribution (Vd)445 ± 91 L500 mg oral QD26
Systemic Clearance (CL)22–24 L/hOral or IV26
Time to Max Concentration (Tmax)Variable, increased with foodOral26
Accumulation Ratio (AUC)0.99–1.24Repeat Dosing26
  • Absorption and Formulation: Epetraborole has been developed in both oral tablet and intravenous (IV) formulations.[1] The oral 500 mg once-daily (QD) dose has been the focus for NTM lung disease and was found to be generally well-tolerated.[1] Administration with food has a mild effect, causing a decrease in the maximum plasma concentration ($C_{max}$) and an increase in the time to reach it ($T_{max}$), but with only a minor change in total drug exposure (AUC).[26]
  • Distribution: A key feature of Epetraborole is its extensive distribution into tissues, evidenced by a large apparent volume of distribution ($Vd$) of approximately 445 L.[1] This high degree of tissue penetration is advantageous for an antibiotic intended to treat infections caused by intracellular pathogens like mycobacteria, which reside within host cells.
  • Metabolism: The primary metabolic pathway for Epetraborole is the oxidation of its propanol side chain to form a carboxylic acid, resulting in the major circulating metabolite, M3, which is microbiologically inactive.[1] This conversion is believed to be mediated by alcohol dehydrogenase (ADH) enzymes rather than the cytochrome P450 (CYP) system.[1] A dedicated Phase 1 study in healthy Japanese subjects, who have a high prevalence of genetic variants in the ADH1B enzyme, confirmed that these different genotypes did not have a clinically meaningful impact on the exposure of either Epetraborole or its M3 metabolite, supporting a consistent dosing strategy across populations.[26]
  • Excretion: The drug and its metabolite are primarily eliminated via the kidneys. Following administration, approximately 90% of the total dose (Epetraborole + M3) is recovered in the urine, with a smaller fraction (~8%) found in the feces.[26] The systemic clearance is relatively low (22–24 L/h), which is consistent with minimal first-pass metabolism after oral dosing.[26]
  • Linearity and Accumulation: Pharmacokinetic studies have demonstrated that Epetraborole exhibits linear kinetics across a wide range of oral and IV doses (250–4000 mg). With repeat daily dosing, there is minimal drug accumulation, with an AUC accumulation ratio near 1.0 (0.99–1.24), which indicates that plasma concentrations at steady-state are predictable and do not increase unexpectedly over time.[26]

Pharmacodynamics (PD): The AUC/MIC Driver

Pharmacodynamic studies are crucial for defining the relationship between drug exposure and antimicrobial effect, which in turn informs optimal dosing regimens. For Epetraborole, preclinical models have consistently identified the ratio of the 24-hour area under the concentration-time curve to the minimum inhibitory concentration (AUC0-24/MIC) as the primary PK/PD index driving its efficacy.[28] This means that the total amount of drug exposure over a 24-hour period, relative to the pathogen's susceptibility, is the most important determinant of bacterial killing. This is characteristic of drugs with concentration-dependent killing and a moderate-to-prolonged post-antibiotic effect. For M. tuberculosis, the optimal AUC/MIC target for bacterial killing in the hollow fiber system model was determined to be 327.1.[28]

Drug-Drug Interaction (DDI) Potential

Given that Epetraborole is intended for use in complex, multi-drug combination regimens for diseases like NTM and melioidosis, a low potential for drug-drug interactions is a critical attribute. Comprehensive in vitro studies were conducted to assess the DDI profile of both Epetraborole and its main metabolite, M3.[1]

  • CYP Enzyme Interactions: The studies revealed a favorable profile. Epetraborole is a poor substrate for the major CYP enzymes, meaning its metabolism is unlikely to be affected by CYP inhibitors or inducers. Furthermore, neither Epetraborole nor M3 were found to be potent reversible or time-dependent inhibitors of these enzymes. Epetraborole was identified as a weak inducer of CYP2B6 and CYP3A4, but not CYP1A2.[1]
  • Transporter Interactions: Epetraborole was found to be a substrate only for the organic cation transporter 2 (OCT2), an uptake transporter primarily in the kidneys, though this is not considered a major pathway for its clearance. At concentrations significantly higher than those achieved clinically, Epetraborole showed weak inhibition of several efflux (P-glycoprotein [P-gp], BCRP) and uptake (OATP1B1/3, OCT1/2) transporters.[1]

Overall, this low potential for clinically significant pharmacokinetic DDIs is a major strategic asset for the molecule. It significantly de-risks its use in combination with the complex antibiotic cocktails required to treat NTM lung disease and melioidosis, as it is unlikely to dangerously alter the concentrations of co-administered drugs.

The DrugBank database also lists a number of theoretical pharmacodynamic interactions. The most prominent of these is a potential increased risk of methemoglobinemia when Epetraborole is combined with various local anesthetics (e.g., benzocaine, articaine, bupivacaine) and other agents.[16] Additionally, a theoretical increased risk of bleeding is noted when combined with vitamin K antagonist anticoagulants (e.g., acenocoumarol, phenprocoumon), and it may decrease the therapeutic efficacy of certain vaccines (e.g., BCG, typhoid) and gut-modifying agents (e.g., lactulose).[16]

Clinical Development Program: A Historical and Current Perspective

The clinical development of Epetraborole has followed a complex and challenging trajectory, characterized by an early-stage failure, a strategic pivot to a rare disease indication, and a subsequent late-stage setback. This history provides valuable context for its current status and future potential. A summary of its clinical trial history is presented in Table 4.

Table 4: Overview of Epetraborole Clinical Trial History and Status

IndicationPhaseStatusKey Rationale / OutcomeSource(s)
Complicated Urinary Tract Infections (cUTI)IIDiscontinuedDiscontinued due to rapid emergence of on-treatment drug resistance.[5, 7, 22]
Complicated Intra-abdominal Infections (cIAI)IIDiscontinuedProgram suspended along with other Gram-negative indications. Trial NCT01381562 was terminated.[7, 38]
Treatment-Refractory MAC Lung DiseaseII/IIITerminatedFailed to meet key microbiological endpoint (sputum culture conversion) despite a positive signal on a patient-reported outcome measure.[11, 39]
Melioidosis (B. pseudomallei Infections)IActiveStrong preclinical rationale and high unmet need due to high mortality with standard of care.7

Early Development for Gram-Negative Infections and Discontinuation

Epetraborole was originally developed by GlaxoSmithKline (GSK) as a novel antibiotic for serious Gram-negative bacterial infections, including complicated urinary tract infections (cUTI) and complicated intra-abdominal infections (cIAI).[5] However, this development program was halted following a Phase II clinical trial in patients with cUTI. The trial revealed a clinically unacceptable rate of rapid emergence of drug resistance in bacteria during the course of treatment.[5] This finding was a major setback, demonstrating that despite its novel mechanism, the drug was not suitable for treating these types of acute infections where a high frequency of resistance could lead to treatment failure. Consequently, the entire Gram-negative infection program was suspended.[7]

Strategic Repurposing for Nontuberculous Mycobacterial (NTM) Lung Disease

Following the discontinuation of its Gram-negative program, the rights to Epetraborole were acquired by AN2 Therapeutics, a company focused on rare infectious diseases.[3] AN2 executed a strategic pivot, repurposing the drug for the treatment of NTM lung disease. This decision was based on several compelling factors:

  1. Strong Preclinical Efficacy: Extensive preclinical data demonstrated potent activity against key NTM pathogens, particularly MAC and the highly drug-resistant M. abscessus.[5]
  2. High Unmet Medical Need: NTM lung disease is a chronic, progressive illness for which current treatments are long, poorly tolerated, and often ineffective, especially in the refractory setting.[31]
  3. Favorable Regulatory Pathway: By targeting a rare disease, the company was able to secure multiple valuable regulatory designations, including Orphan Drug, QIDP, and Fast Track status, which are designed to expedite the development and review of drugs for serious conditions.[8]

In-Depth Analysis of the EBO-301 Phase 2/3 Trial in MAC Lung Disease

The centerpiece of AN2's strategy was the EBO-301 trial (NCT05327803), a pivotal Phase 2/3, randomized, double-blind, placebo-controlled study designed to evaluate the efficacy and safety of oral Epetraborole (500 mg once daily) when added to an Optimized Background Regimen (OBR) in patients with treatment-refractory MAC lung disease (TR-MAC-LD).[1]

The trial deliberately enrolled a very challenging patient population with severe, advanced disease, long disease duration, high rates of cavitary lung disease, and significant baseline resistance to existing antimycobacterial drugs.[12] While this "highest unmet need" strategy can accelerate development, it also carries substantial risk.

In February 2024, AN2 announced a voluntary pause in the enrollment of the Phase III portion of the trial after a blinded aggregate data review suggested potentially lower-than-expected efficacy.[10] Subsequently, upon unblinding the topline results from the 80-patient Phase 2 portion of the study, the company announced the termination of the entire trial.[11]

The results of the Phase 2 study were nuanced and are summarized in Table 5. The trial technically met its primary objective, which was to validate a novel patient-reported outcome (PRO) instrument called MACrO2. The data showed a numerically higher clinical response rate based on this PRO in the Epetraborole arm (39.5%) compared to the placebo arm (25.0%). While not statistically significant (p=0.19), this signal was supported by a nominally statistically significant improvement in a secondary PRO measure, the Quality of Life–Bronchiectasis (QOL-B) Respiratory Domain score (p=0.0365).[11] This indicated that patients receiving Epetraborole were, on average, experiencing a tangible improvement in their respiratory symptoms.

However, this symptomatic improvement did not translate to a microbiological benefit. The trial failed to meet its key secondary endpoint of sputum culture conversion (SCC) by month 6. The SCC rates were low and virtually identical between the two arms: 13.2% in the Epetraborole group versus 10.0% in the placebo group (p=0.64).[11] The inability of Epetraborole to clear the mycobacteria from patients' lungs, despite making them feel better, was the ultimate reason for the trial's termination.

This outcome suggests that achieving a complete microbiological cure in such an advanced, ultra-refractory patient population with extensive lung damage and high bacterial burden may have been an unrealistic goal. The drug may have been effective at reducing the overall bacterial load to a degree that alleviated symptoms (the PRO signal), but not enough to achieve full culture negativity against a backdrop of scarred lung tissue and a partially effective OBR. This result provides a critical lesson for the design of future NTM clinical trials.

Table 5: Key Efficacy Outcomes from the EBO-301 Phase 2 Study (Month 6)

EndpointEpetraborole + OBR Arm (N=39)Placebo + OBR Arm (N=41)Treatment Difference (95% CI)p-valueSource(s)
MACrO2 PRO Response Rate15/38 (39.5%)10/40 (25.0%)13.9% (-6.8, 33.8)0.186312
Sputum Culture Conversion (SCC)5/38 (13.2%)4/40 (10.0%)3.4% (-12.0, 19.8)0.636612
QOL-B Resp. Domain LS Mean Change7.200.306.90 (0.45, 13.36)0.036512

Investigational Pathway for Melioidosis

Epetraborole remains in active, albeit early-stage, clinical development for the treatment of melioidosis.[7] The rationale for this indication is strong, driven by the disease's high mortality rate (often exceeding 40% even with treatment), the limitations of the current standard of care (prolonged courses of IV ceftazidime or meropenem followed by oral trimethoprim-sulfamethoxazole), and its designation as a high-priority biothreat agent.[1] Preclinical data have been highly encouraging, demonstrating potent in vivo efficacy.[13] A Phase 1 clinical trial has already established that a 2000 mg IV dose of Epetraborole in humans can achieve the AUC exposure (~110 µg·h/mL) that was identified as effective in murine models, and that daily IV doses up to 4000 mg for 14 days were well tolerated.[33]

Comprehensive Safety and Tolerability Profile

Overview of Human Safety Data

The safety and tolerability of Epetraborole have been evaluated across multiple Phase 1 studies in healthy volunteers and in the Phase 2/3 EBO-301 study in patients with TR-MAC-LD.[12] Across this program, the drug has consistently demonstrated a generally favorable and manageable safety profile. Oral doses up to 1,000 mg daily and IV doses up to 4,000 mg daily have been administered and were well tolerated.[33] Critically, the decision to terminate the pivotal EBO-301 trial was based solely on insufficient efficacy and was not driven by any safety concerns.[11] This manageable safety profile remains one of the drug's key assets.

Adverse Event Profile from Clinical Trials

Phase 1 Studies (Healthy Volunteers)

In studies involving healthy volunteers, Epetraborole was generally well tolerated.[46] The most frequently reported treatment-emergent adverse events (TEAEs) were mild and primarily gastrointestinal in nature. In a 28-day dose-ranging study, the most common TEAEs in the Epetraborole group were nausea (23.1%), headache (17.9%), and diarrhea (10.3%).[10] No severe or serious adverse events were reported in these studies.[46] A Phase 1 study conducted exclusively in healthy Japanese subjects was notable for having no reported TEAEs whatsoever.[37]

EBO-301 Phase 2 Study (TR-MAC Patients)

In the more complex and co-morbid patient population of the EBO-301 trial, the incidence of TEAEs was high overall, but was broadly similar between the Epetraborole and placebo groups (94.9% vs. 85.4%, respectively).[12] As expected, TEAEs considered by investigators to be related to the study drug were more frequent in the active treatment arm (76.9%) compared to the placebo arm (24.4%).[12]

An adverse event of special interest that emerged from the trial was anemia or a decrease in hemoglobin. This was the most frequently reported drug-related TEAE, occurring in 35.9% of patients receiving Epetraborole versus 0% of patients receiving placebo.[12] The clinical course of this anemia was predictable and manageable: hemoglobin values declined gradually after treatment initiation, stabilized by the third month of therapy, and returned to or toward baseline levels after the drug was discontinued. This side effect was generally tolerated by this elderly patient population and was considered monitorable.[12]

Other common TEAEs reported in at least 10% of patients in the Epetraborole arm included dizziness (17.9%), headache (10.3%), COVID-19 (10.3%), diarrhea (10.3%), pyrexia (fever, 10.3%), and back pain (10.3%).[12] Gastrointestinal intolerance events were generally mild to moderate and infrequently led to discontinuation of the study drug.[12]

The Challenge of Antimicrobial Resistance

Mechanism of Resistance

The molecular basis for bacterial resistance to Epetraborole is well understood and highly specific. Resistance arises from missense mutations within the editing domain of the target enzyme, LeuRS, which is encoded by the leuS gene.[5] This has been experimentally confirmed through the selection of Epetraborole-resistant mutants of M. abscessus; sequencing of the leuS gene in these mutants invariably revealed mutations clustered within the region corresponding to the LeuRS editing domain.[5] These mutations alter the binding site, preventing the Epetraborole-tRNALeu adduct from being trapped and thereby allowing protein synthesis to continue.

Clinical Emergence of Resistance

The clinical relevance of this resistance mechanism was starkly demonstrated during the drug's early development. The Phase II trial in patients with cUTI was discontinued precisely because of the rapid emergence of resistant Gram-negative bacteria during the short course of therapy.[5] The frequency of spontaneous resistance to Epetraborole has been calculated to be in the range of $3.8 \times 10^{-8}$ to $8.1 \times 10^{-7}$ per colony-forming unit (CFU) for various Gram-negative species and M. abscessus, a frequency high enough to be a significant clinical concern, especially during monotherapy for acute infections.[5]

A Novel Mitigation Strategy: Exploiting the Resistance Mechanism

While resistance presents a major challenge, a fascinating and innovative strategy has emerged to potentially overcome it. The very mutations in the LeuRS editing domain that confer resistance to Epetraborole come at a significant fitness cost to the bacterium: the enzyme loses its critical proofreading or editing function.[29] This means the enzyme can no longer recognize and remove incorrectly charged amino acids from tRNALeu.

This acquired defect can be exploited therapeutically. The resistant bacteria become uniquely vulnerable to the misincorporation of amino acid analogs that mimic leucine, such as the non-proteinogenic amino acid norvaline.[29] When exposed to norvaline, the defective LeuRS in the resistant bacteria mistakenly attaches it to tRNALeu. This leads to the widespread incorporation of norvaline into newly synthesized proteins in place of leucine, causing protein misfolding, a cellular stress response, and ultimately, impaired bacterial growth or death.[29]

This concept has been validated experimentally. The co-administration of Epetraborole and norvaline was shown to have significantly improved efficacy in a murine model of M. abscessus infection compared to Epetraborole alone. Furthermore, supplementing culture media with norvaline was shown to reduce the in vitro emergence of Epetraborole-resistant mutants.[29] This strategy effectively reframes the problem of resistance, turning the bacterium's escape mechanism into a "Trojan horse" that creates a new, targetable vulnerability—a concept known as synthetic lethality. This approach holds significant promise for ensuring the durable efficacy of Epetraborole, particularly in the context of long-term therapy for chronic infections like NTM.

Strategic Analysis and Future Outlook

Post-Mortem on the EBO-301 Trial: Why Did a PRO Signal Not Translate to Microbiological Success?

The failure of the EBO-301 trial in TR-MAC-LD, despite a positive signal in patient-reported outcomes, warrants a careful strategic analysis. The outcome was likely the result of a confluence of factors related to trial design and the inherent challenges of the disease state, rather than a simple failure of the drug itself. The decision to target an ultra-refractory patient population—while sound from a regulatory and unmet need perspective—placed an extremely high bar for success.[39] These patients presented with advanced, often cavitary, lung disease, extensive scarring, and high rates of baseline resistance to the drugs in their optimized background regimen.[12] In this context, achieving complete and sustained microbiological eradication (sputum culture conversion) is a formidable challenge for any new agent. The observed disconnect between symptomatic improvement (the PRO signal) and microbiological cure suggests that Epetraborole may exert a bacteriostatic or slowly bactericidal effect in vivo. This effect could be sufficient to reduce the overall mycobacterial load and associated inflammation, leading to patients feeling better, but insufficient to fully clear the infection from damaged and scarred lung tissue. Therefore, the trial's failure may be more indicative of a mismatch between the drug's capabilities and an exceedingly difficult clinical challenge—a "bridge too far"—than a definitive verdict on its potential utility in NTM disease.

Evaluation of Epetraborole's Viability in Future Indications

Despite the setback in MAC, Epetraborole possesses a unique profile that may be well-suited for other serious infections with high unmet needs.

The Case for Mycobacterium abscessus

M. abscessus lung disease arguably represents the most promising future indication for Epetraborole. The strategic rationale is built on several strong pillars:

  1. Exceptional Potency: The in vitro activity of Epetraborole against M. abscessus is substantially greater than against MAC, with an MIC90 of just 0.12 µg/mL.[26] This significantly increases the probability of achieving the target AUC/MIC ratio at safe and well-tolerated doses, improving the likelihood of clinical and microbiological success.
  2. Dire Unmet Need: Current treatment for M. abscessus is arduous, requiring prolonged courses of multi-drug intravenous therapy that are associated with significant toxicity and yield cure rates of 50% or less.[5] An effective, well-tolerated oral agent would be a paradigm-shifting advance for patients.
  3. Novel Mechanism and Resistance Mitigation: Its unique mechanism of action is a major advantage against a pathogen with high levels of intrinsic and inducible resistance to other antibiotic classes.[29] Furthermore, the innovative strategy of combining Epetraborole with norvaline to prevent and overcome resistance provides a potential pathway to achieve durable efficacy.[29]

The Case for Melioidosis

Melioidosis represents a high-risk, high-reward opportunity. The primary therapeutic goal in this acute, life-threatening infection would be to reduce the unacceptably high mortality rate seen with current standard-of-care antibiotics.[1] The case for its development rests on:

  1. Strong Preclinical Rationale: Potent, strain-independent in vivo efficacy has been clearly demonstrated in relevant animal models of the disease.[13]
  2. Achievable Therapeutic Exposure: Phase 1 human pharmacokinetic data indicate that the effective AUC target identified in preclinical models is achievable in humans using a well-tolerated intravenous dosing regimen.[33]

Recommendations for Future Clinical Development

The path forward for Epetraborole requires a carefully considered clinical and corporate strategy. AN2 Therapeutics' decision to pivot its primary focus to its broader boron chemistry platform is a prudent corporate strategy to diversify and de-risk its pipeline.[11] However, Epetraborole remains a valuable asset that should not be abandoned. The pursuit of non-dilutive funding to advance its development in targeted niche indications is a sound approach.[14]

  • For M. abscessus: A meticulously designed Phase II proof-of-concept study should be initiated. To maximize the probability of success, this trial should consider enrolling treatment-naïve or less refractory patients than the EBO-301 MAC trial. The study should prospectively evaluate the combination of Epetraborole with norvaline alongside an optimized background regimen. The primary endpoint should be a robust microbiological outcome, such as sputum culture conversion, supported by key secondary clinical endpoints, including PROs.
  • For Melioidosis: A Phase II trial should be designed to evaluate the addition of intravenous Epetraborole to the current standard of care (e.g., ceftazidime or meropenem) in patients with severe melioidosis. The primary endpoint for such a study should be a clinical outcome that reflects the urgent goal of treatment: a reduction in 28-day or 90-day all-cause mortality.

In conclusion, Epetraborole is a scientifically innovative antibiotic whose clinical journey has been a lesson in the complexities of drug development. While its path in MAC lung disease has been halted, its unique mechanism, potent activity against highly resistant pathogens, and manageable safety profile provide a strong rationale for its continued investigation in M. abscessus and melioidosis, where it still holds the potential to address profound unmet medical needs.

Works cited

  1. In Vitro Evaluation of Drug–Drug Interaction Potential of Epetraborole, a Novel Bacterial Leucyl-tRNA Synthetase Inhibitor - MDPI, accessed October 30, 2025, https://www.mdpi.com/1424-8247/17/1/120
  2. epetraborole | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed October 30, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=13079
  3. Development Pipeline - AN2 Therapeutics, accessed October 30, 2025, https://www.an2therapeutics.com/pipeline/
  4. epetraborole | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed October 30, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=refs&ligandId=13079
  5. Epetraborole Is Active against Mycobacterium abscessus ..., accessed October 30, 2025, https://journals.asm.org/doi/10.1128/aac.01156-21
  6. In Vitro Evaluation of Drug–Drug Interaction Potential of Epetraborole, a Novel Bacterial Leucyl-tRNA Synthetase Inhibitor - PMC - NIH, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10818931/
  7. Epetraborole - Pfizer - AdisInsight, accessed October 30, 2025, https://adisinsight.springer.com/drugs/800031276
  8. AN2 Therapeutics Announces Epetraborole Granted Orphan Drug ..., accessed October 30, 2025, https://investor.an2therapeutics.com/news-releases/news-release-details/an2-therapeutics-announces-epetraborole-granted-orphan-drug/
  9. AN2 Therapeutics Provides Update on Epetraborole Ex-U.S. Development Plan in Treatment-Refractory MAC Lung Disease - BioSpace, accessed October 30, 2025, https://www.biospace.com/an2-therapeutics-provides-update-on-epetraborole-ex-u-s-development-plan-in-treatment-refractory-mac-lung-disease
  10. Epetraborole Phase III enrolment pause sets back NTM late-stage pipeline, accessed October 30, 2025, https://www.clinicaltrialsarena.com/analyst-comment/epetraborole-phase-iii-enrolment-pause-sets-back-ntm-late-stage-pipeline/
  11. AN2 Therapeutics Stops Phase 2/3 Epetraborole Trial for MAC Lung Disease, accessed October 30, 2025, https://respiratory-therapy.com/products-treatment/pharmaceuticals/clinical-trials/an2-therapeutics-stops-phase-2-3-epetraborole-trial-mac-lung-disease/
  12. Results: Phase 2 Study (EBO-301) to Assess the ... - AN2 Therapeutics, accessed October 30, 2025, https://www.an2therapeutics.com/file.cfm/17/docs/69-poster_ebo-301_p2%20results_csu_ntm.pdf
  13. Epetraborole, a leucyl-tRNA synthetase inhibitor, demonstrates murine efficacy, enhancing the in vivo activity of ceftazidime against Burkholderia pseudomallei, the causative agent of melioidosis - PubMed Central, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10703401/
  14. AN2 Therapeutics Reports Data from the Phase 3 Portion of Previously Terminated EBO-301 Study and Outlines Future Milestones from Boron Chemistry Pipeline, accessed October 30, 2025, https://investor.an2therapeutics.com/news-releases/news-release-details/an2-therapeutics-reports-data-phase-3-portion-previously?mobile=1
  15. AN2 Therapeutics slides as MAC lung disease trial misses mark - FirstWord Pharma, accessed October 30, 2025, https://firstwordpharma.com/story/5956201
  16. Epetraborole: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 30, 2025, https://go.drugbank.com/drugs/DB11744
  17. Epetraborole | C11H16BNO4 | CID 46836890 - PubChem, accessed October 30, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Epetraborole
  18. epetraborole | Ligand page | IUPHAR/BPS Guide to, accessed October 30, 2025, https://www.guidetoimmunopharmacology.org/GRAC/LigandDisplayForward?ligandId=13079
  19. Epetraborole - TargetMol, accessed October 30, 2025, https://www.targetmol.com/compound/epetraborole
  20. Epetraborole | CAS# 1093643-37-8 | Biochemical - MedKoo Biosciences, accessed October 30, 2025, https://www.medkoo.com/products/44323
  21. Epetraborole | Bacterial | 1093643-37-8 | Invivochem, accessed October 30, 2025, https://www.invivochem.com/product/V102849
  22. Epetraborole (GSK-2251052) | LeuRS Inhibitor - MedchemExpress.com, accessed October 30, 2025, https://www.medchemexpress.com/epetraborole.html
  23. EPETRABOROLE - precisionFDA, accessed October 30, 2025, https://precision.fda.gov/ginas/app/ui/substances/e41a483f-05ad-4bc4-a961-0c5ec2920b06
  24. EPETRABOROLE - gsrs, accessed October 30, 2025, https://gsrs.ncats.nih.gov/ginas/app/beta/substances/6MC93Z2DF9
  25. Epetraborole by AN2 Therapeutics for Burkholderia pseudomallei Infections (Melioidosis): Likelihood of Approval - Pharmaceutical Technology, accessed October 30, 2025, https://www.pharmaceutical-technology.com/data-insights/epetraborole-an2-therapeutics-burkholderia-pseudomallei-infections-melioidosis-likelihood-of-approval/
  26. Company Overview - AN2 Therapeutics, accessed October 30, 2025, https://www.an2therapeutics.com/file.cfm/17/docs/eckburg_idweek_2023_epetraborole_pipeline_talk_pe12oct23.pdf
  27. 2135. Epetraborole in vitro Activity Against Mycobacterium avium ..., accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10679380/
  28. Epetraborole pharmacokinetics/pharmacodynamics in the hollow ..., accessed October 30, 2025, https://journals.asm.org/doi/10.1128/aac.00481-25
  29. Efficacy of epetraborole against Mycobacterium abscessus is increased with norvaline - NIH, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8535176/
  30. A Screening of the MMV Pandemic Response Box Reveals Epetraborole as A New Potent Inhibitor against Mycobacterium abscessus - MDPI, accessed October 30, 2025, https://www.mdpi.com/1422-0067/22/11/5936
  31. Epetraborole, a Novel Bacterial Leucyl-tRNA Synthetase Inhibitor, Demonstrates Potent Efficacy and - AN2 Therapeutics, accessed October 30, 2025, https://www.an2therapeutics.com/file.cfm/17/docs/1704-epetraborole-a-novel-leurs-inhibitor-demonstrates-potent-efficacy-and-improves-efficacy-of-soc-regimen-against-mac-in-a-chronic-mouse-lung-infection-model.pdf
  32. 1704. Epetraborole, a Novel Bacterial Leucyl-tRNA Synthetase Inhibitor, Demonstrates Potent Efficacy and Improves Efficacy of Standard of Care Regimen Against Mycobacterium avium complex in a Chronic Mouse Lung Infection Model - NIH, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9752932/
  33. Pharmacokinetics and In vivo Efficacy of Epetraborole against ..., accessed October 30, 2025, https://www.biorxiv.org/content/10.1101/2025.07.10.664055.abstract
  34. Pharmacokinetics and in vivo efficacy of epetraborole against Burkholderia pseudomallei, accessed October 30, 2025, https://www.researchgate.net/publication/396773781_Pharmacokinetics_and_in_vivo_efficacy_of_epetraborole_against_Burkholderia_pseudomallei
  35. Epetraborole pharmacokinetics/pharmacodynamics in the hollow fiber system model of Mycobacterium tuberculosis - ResearchGate, accessed October 30, 2025, https://www.researchgate.net/publication/395727164_Epetraborole_pharmacokineticspharmacodynamics_in_the_hollow_fiber_system_model_of_Mycobacterium_tuberculosis
  36. AN2 Therapeutics to Present New Data for Epetraborole at IDWeek 2023, accessed October 30, 2025, https://investor.an2therapeutics.com/news-releases/news-release-details/an2-therapeutics-present-new-data-epetraborole-idweek-2023/
  37. 2556. A Phase 1, Open-Label, Single Dose Study to Evaluate the Pharmacokinetics (PK), Safety, and Tolerability of Epetraborole Tablets and the Impact of Alcohol Dehydrogenase (ADH) Genotype on the PK of Epetraborole and Metabolite M3 in Healthy Japanese Adult Subjects | Open Forum Infectious Diseases | Oxford Academic, accessed October 30, 2025, https://academic.oup.com/ofid/article/10/Supplement_2/ofad500.2173/7446993
  38. NCT05327803 | Study of Epetraborole in Patients With Treatment-refractory MAC Lung Disease | ClinicalTrials.gov, accessed October 30, 2025, https://www.clinicaltrials.gov/study/NCT05327803
  39. Full article: Treatment of nontuberculous mycobacterial pulmonary disease requires a stepwise and multidisciplinary approach, accessed October 30, 2025, https://www.tandfonline.com/doi/full/10.1080/17476348.2025.2479615
  40. A Phase 2/3, Randomized, Double-blind, Placebo-controlled, Multicenter, Prospective Study to Assess the Efficacy, Safety, and Pharmacokinetics of Orally Administered Epetraborole in Patients With Treatment-refractory Mycobacterium Avium Complex Lung Disease - Froedtert, accessed October 30, 2025, https://www.froedtert.com/clinical-trials/phase-23-randomized-double-blind-placebo-controlled-multicenter-prospective-study
  41. AN2 Therapeutics halts Phase III enrolment for trial of epetraborole - Clinical Trials Arena, accessed October 30, 2025, https://www.clinicaltrialsarena.com/news/an2-therapeutics-trial-epetraborole/
  42. AN2 Therapeutics Updates on EBO-301 Phase 2/3 Study of Epetraborole for Treatment-Refractory MAC Lung Disease - Patsnap Synapse, accessed October 30, 2025, https://synapse.patsnap.com/article/an2-therapeutics-updates-on-ebo-301-phase-23-study-of-epetraborole-for-treatment-refractory-mac-lung-disease
  43. AN2 Provides Strategic Update for Phase 3 EBO-301 Trial in Treatment Refractory MAC Lung Disease, accessed October 30, 2025, https://investor.an2therapeutics.com/news-releases/news-release-details/an2-provides-strategic-update-phase-3-ebo-301-trial-treatment/
  44. Effective Therapeutic Options for Melioidosis: Antibiotics versus Phage Therapy - PMC, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9863960/
  45. AN2 Therapeutics Reports Phase 1b Data for Oral Epetraborole, accessed October 30, 2025, https://www.an2therapeutics.com/news/20220210/
  46. 1727. Phase 1b Dose-ranging Study Demonstrates Tolerability and Pharmacokinetics (PK) of Oral Epetraborole at the Predicted Therapeutic Dosage for Mycobacterium avium Complex (MAC) Lung Disease - PMC - NIH, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9752554/
  47. Efficacy of epetraborole against Mycobacteroides abscessus in a mouse model of lung infection | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed October 30, 2025, https://journals.asm.org/doi/10.1128/aac.00648-24
  48. Efficacy of epetraborole against Mycobacteroides abscessus in a mouse model of lung infection - PMC - NIH, accessed October 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11323969/

Published at: October 30, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.