C30H47NO4S
224452-66-8
Staphylococcal impetigo, Streptococcal impetigo
Retapamulin is a semi-synthetic, small-molecule antibiotic and the first representative of the pleuromutilin class approved for human topical use.[1] Developed by GlaxoSmithKline, it is marketed under the brand names Altabax and Altargo for the treatment of uncomplicated bacterial skin infections.[1] Its primary clinical indication is for impetigo caused by susceptible strains of
Staphylococcus aureus (methicillin-susceptible only) and Streptococcus pyogenes.[1]
The therapeutic value of retapamulin stems from its novel mechanism of action. It selectively inhibits bacterial protein synthesis by binding to a unique site on the 50S ribosomal subunit, an interaction distinct from all other classes of ribosomal inhibitors.[4] This unique binding confers a low intrinsic potential for target-specific cross-resistance with other established antibiotic classes, a significant advantage in an era of increasing antimicrobial resistance.[4]
Clinical trials have established its efficacy. A 5-day, twice-daily regimen of 1% retapamulin ointment was demonstrated to be significantly superior to placebo in treating impetigo and non-inferior to both topical sodium fusidate and a 10-day course of oral cephalexin for certain skin infections.[7] Its safety profile is favorable, characterized predominantly by mild and infrequent local application site reactions, with minimal systemic absorption in adults and older children.[10]
Despite these strengths, retapamulin's clinical utility and market success have been constrained by several critical factors. A notable paradox exists between its excellent in vitro activity against methicillin-resistant S. aureus (MRSA) and its inadequate clinical efficacy against MRSA infections, leading to a contraindication for such use.[12] Furthermore, its clinical development program lacked a large-scale, head-to-head comparative trial against mupirocin, the widely accepted standard of care, creating an evidence gap that has positioned it as a more expensive alternative rather than a proven replacement.[3] These limitations, coupled with commercial considerations, ultimately led to the withdrawal of its marketing authorization in the European Union.[13] Retapamulin's lifecycle thus serves as an important case study on the complex interplay between scientific innovation, clinical evidence, and commercial viability in the modern antibiotic market.
Retapamulin is a small-molecule drug substance belonging to the pleuromutilin class of antibiotics.[14] It is systematically identified by a range of chemical and regulatory codes to ensure unambiguous reference in scientific literature, clinical practice, and regulatory documentation.
The physical and chemical properties of retapamulin dictate its formulation as a topical agent and influence its pharmacokinetic behavior.
Retapamulin was developed and originally marketed by the pharmaceutical company GlaxoSmithKline (GSK).[1] It is a semi-synthetic derivative of pleuromutilin, a natural diterpene antibiotic produced via fermentation of the basidiomycete fungus
Clitopilus passeckerianus.[5] While other pleuromutilin derivatives like tiamulin and valnemulin have been used in veterinary medicine, retapamulin was the first to be approved for human use.[21] It is known commercially by two primary brand names:
Altabax, used predominantly in the United States, and Altargo, used in the European Union and other international markets.[1]
The origin of retapamulin as a semi-synthetic compound derived from a fungal metabolite is a foundational element of its pharmacological profile. This distinct chemical lineage, separate from purely synthetic antibiotics or other natural product classes, results in a novel molecular structure. This structural novelty is the direct antecedent to its unique mechanism of action, which in turn provides the key therapeutic rationale for its development: a low probability of encountering pre-existing, target-specific cross-resistance with other antibiotic classes that have been in widespread clinical use for decades.[2] This attribute is particularly valuable in the context of globally rising antimicrobial resistance.
Table 1: Key Identifiers and Physicochemical Properties of Retapamulin
| Property | Value |
|---|---|
| Generic Name | Retapamulin |
| DrugBank ID | DB01256 |
| CAS Number | 224452-66-8 |
| IUPAC Name | (3aS,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-5-hydroxy-4,6,9,10-tetramethyl-1-oxodecahydro-3a,9-propano-3aH-cyclopentaannulen-8-yl${oct-3-yl]sulfanyl$}$acetate |
| Molecular Formula | C30H47NO4S |
| Molecular Weight | 517.76 g/mol |
| Physical Appearance | White to pale-yellow solid |
| Solubility | Insoluble in water; soluble in DMSO and ethanol |
| Brand Names | Altabax, Altargo |
Retapamulin exerts its antibacterial effect through the selective inhibition of bacterial protein synthesis, a mechanism common to many antibiotic classes. However, its specific mode of interaction with the bacterial ribosome is unique and defines its pharmacological class.[1]
Retapamulin binds with high potency (dissociation constant, Kd, of 3 nM) to a novel site on the 50S subunit of the bacterial ribosome.[16] This binding site is located within domain V of the 23S ribosomal RNA (rRNA) at the peptidyl transferase center (PTC), in proximity to ribosomal protein L3.[14] This location is distinct from the binding sites of other major classes of 50S inhibitors, such as macrolides, lincosamides, and streptogramins, which minimizes the potential for target-specific cross-resistance.[4] In contrast to its high affinity for prokaryotic ribosomes, retapamulin is ineffective at inhibiting eukaryotic protein synthesis, demonstrating its selective toxicity for bacteria.[16]
By occupying this unique ribosomal site, retapamulin disrupts protein synthesis through several concerted mechanisms, effectively halting bacterial proliferation [4]:
At clinically relevant concentrations, retapamulin's action is predominantly bacteriostatic, meaning it inhibits the growth and reproduction of bacteria without directly killing them.[3] This is sufficient for treating localized skin infections, where the host immune system can then clear the inhibited pathogens. However, at concentrations approximately 1,000 times higher than its Minimum Inhibitory Concentration (MIC), retapamulin can exhibit bactericidal (cell-killing) activity.[14] Such high concentrations are achievable at the site of topical application but not systemically.
The pharmacokinetic profile of retapamulin is characterized by low systemic absorption, which is central to its use as a topical agent and its overall safety profile.
Systemic absorption following topical application of the 1% ointment is minimal in most patient populations.[21]
Data regarding the tissue distribution of retapamulin in humans is not available.[7] For the small fraction of the drug that is systemically absorbed, it is highly bound (approximately 94%) to human plasma proteins.[21]
Systemically absorbed retapamulin is extensively metabolized in the liver. The primary metabolic pathway involves the Cytochrome P450 3A4 (CYP3A4) enzyme system, which catalyzes mono-oxygenation and di-oxygenation reactions to produce multiple inactive metabolites.[7]
The precise routes and extent of excretion of retapamulin and its metabolites in humans have not been determined.[1]
The drug's pharmacokinetic profile directly shapes its clinical utility and safety considerations. The minimal systemic absorption in adults and older children is the basis for its favorable systemic safety profile and its limitation to topical use for superficial infections only.[4] Concurrently, the finding of significantly higher systemic exposure in infants under 24 months, combined with its known metabolism by CYP3A4, creates a clinically relevant risk. This elevated exposure could lead to significant drug-drug interactions if co-administered with strong CYP3A4 inhibitors (e.g., ketoconazole), which could dangerously increase retapamulin plasma levels. This causal link between age-dependent pharmacokinetics and metabolic pathways is the direct scientific foundation for the explicit clinical warning against such co-administration in patients younger than 24 months.[10]
Retapamulin's antibacterial spectrum is focused on Gram-positive organisms, which are the most common causative pathogens in uncomplicated skin and skin structure infections.[4]
Retapamulin demonstrates high potency against its target pathogens in laboratory settings, as measured by MIC values. Global surveillance studies have shown that the concentration required to inhibit 90% of isolates (MIC90) for S. aureus and S. pyogenes is typically in the range of ≤0.03 to 0.12 μg/mL.[4]
A key microbiological feature of retapamulin is its retained activity against strains that have developed resistance to other antibiotic classes. In vitro, it remains potent against isolates of S. aureus that are resistant to methicillin (MRSA), mupirocin, and fusidic acid.[6] This resistance-breaking profile was a cornerstone of its developmental promise.
While the potential for resistance development to retapamulin is considered low, several mechanisms have been identified.[6]
Despite these potential mechanisms, no development of resistance was observed during the clinical trial program for retapamulin.[21] This supports the premise that its unique mechanism of action makes it less susceptible to rapid resistance selection compared to some other topical agents.[2] However, a notable disconnect exists between this promising microbiological profile and its ultimate clinical application. While the development of new antibiotics is often driven by the need to overcome resistance to existing therapies, and retapamulin's
in vitro activity against mupirocin- and fusidic acid-resistant strains would seem to position it perfectly for this role, its approved indication is for general uncomplicated impetigo.[22] It was not specifically studied or approved for treating infections known to be caused by resistant pathogens. This failure to fully leverage its most significant microbiological strength in its clinical development strategy may have contributed to its perception as a niche alternative rather than an essential tool for combating resistance.
The clinical efficacy of retapamulin 1% ointment has been evaluated in a series of phase III trials for primary impetigo and secondarily infected skin lesions.
The superiority of retapamulin over placebo for the treatment of impetigo was firmly established in a randomized, double-blind, multicenter trial (Study 103469). In this study, patients aged 9 months and older received either retapamulin ointment twice daily for 5 days or a placebo ointment. The primary endpoint was clinical success at the end of therapy (day 7). In the intent-to-treat (ITT) population, the clinical success rate was 85.6% for the retapamulin group compared to 52.1% for the placebo group, a statistically significant difference (p<0.0001).[2] Microbiological success rates were similarly superior for retapamulin (91.2% vs. 50.9%).[2]
To establish its efficacy relative to an existing active treatment, retapamulin was compared to sodium fusidate 2% ointment in a randomized, observer-blinded, non-inferiority study. Patients received either retapamulin twice daily for 5 days or sodium fusidate three times daily for 7 days. The results demonstrated that retapamulin was non-inferior to sodium fusidate. In the per-protocol population, clinical efficacy was 99.1% for retapamulin versus 94.0% for sodium fusidate.[8] Another smaller study reported similar clinical success rates between retapamulin (84%) and fusidic acid (80%) in children with primary impetigo.[32]
The utility of retapamulin was extended to secondarily infected traumatic lesions (SITLs), such as small lacerations, sutured wounds, and abrasions. In two large, identical, randomized, double-blind trials, a 5-day course of topical retapamulin was compared to a 10-day course of oral cephalexin. The studies demonstrated that topical retapamulin was non-inferior to systemic cephalexin, with comparable clinical success rates at follow-up (e.g., 88.7% for retapamulin vs. 91.9% for cephalexin in one study).[3] These findings support the use of localized topical therapy as an effective alternative to systemic antibiotics for uncomplicated infections, thereby reducing the potential for systemic side effects and broader antibiotic pressure.[4]
A critical and defining limitation of retapamulin is the discrepancy between its laboratory performance and its clinical efficacy against methicillin-resistant Staphylococcus aureus (MRSA). As established, retapamulin demonstrates excellent in vitro potency against MRSA isolates, including those resistant to other topical agents like mupirocin.[6] This profile suggested it could be a valuable new weapon against this challenging pathogen.
However, clinical trial data did not support this potential. In studies of secondarily infected wounds, clinical success rates for infections caused by MRSA were found to be inadequate and significantly lower than for those caused by MSSA (68.6% vs. 92.2%, respectively).[12] In another unpublished study, retapamulin was significantly less effective than oral linezolid for treating MRSA skin infections (63.9% vs. 90.6% success).[12] This failure to translate promising
in vitro data into clinical success for one of the most significant resistant skin pathogens severely curtailed the drug's therapeutic niche. Consequently, prescribing information explicitly warns that retapamulin should not be used to treat infections known or suspected to be caused by MRSA.[12] This paradox effectively relegated the drug from a potential solution for difficult-to-treat infections to an alternative for more common, susceptible ones, placing it in direct competition with cheaper, established generic therapies.
Mupirocin is widely considered a first-line standard of care for the topical treatment of impetigo in many clinical guidelines.[3] A significant gap in retapamulin's clinical development program is the absence of a large, pivotal, head-to-head trial comparing it directly with mupirocin.[3] Such a trial would have been essential to definitively establish its place in therapy relative to the market leader.
The limited comparative data comes from a small, double-blind, community-based study in 57 children, which found retapamulin to be as effective as mupirocin based on wound size reduction and culture results. Notably, patients treated with retapamulin had a significantly lower (better) post-treatment score on the Skin Infection Rate Scale (SIRS) (p=0.015).[38]
Without more robust comparative data, retapamulin is generally positioned as an effective alternative to mupirocin, not a proven superior agent.[3] Its potential advantages lie in a more convenient dosing schedule (twice daily for 5 days vs. mupirocin's three times daily for up to 10-12 days) and a theoretically lower potential for resistance development.[3] However, these benefits are weighed against its significantly higher cost compared to generic mupirocin.[3] This lack of direct, compelling evidence of at least non-inferiority in a large trial made it difficult for clinicians and formulary committees to justify its routine use over the established, less expensive standard of care.
Table 2: Summary of Pivotal Phase III Clinical Trials for Retapamulin
| Study Reference | Indication | Patient Population (N) | Treatment Arms | Primary Endpoint | Key Efficacy Results (% Success) |
|---|---|---|---|---|---|
| Study 103469 7 | Impetigo | 210 | Retapamulin 1% BID for 5 days vs. Placebo BID for 5 days | Clinical success at end of therapy (Day 7) | 85.6% vs. 52.1% (p<0.0001) |
| Oranje et al. 2007 8 | Impetigo | 519 | Retapamulin 1% BID for 5 days vs. Sodium Fusidate 2% TID for 7 days | Clinical success (non-inferiority) | 99.1% vs. 94.0% (Per-protocol; non-inferiority met) |
| Study 030A/030B 7 | Secondarily Infected Traumatic Lesions | 1,904 (total) | Retapamulin 1% BID for 5 days vs. Oral Cephalexin 500 mg BID for 10 days | Clinical success at follow-up (non-inferiority) | 88.7% vs. 91.9% (Study 030A; non-inferiority met) |
Table 3: Comparative Profile: Retapamulin vs. Mupirocin and Fusidic Acid
| Feature | Retapamulin | Mupirocin | Fusidic Acid |
|---|---|---|---|
| Antibiotic Class | Pleuromutilin | Monoxycarbolic acid | Fusidane (steroid-like) |
| Mechanism of Action | Inhibits protein synthesis (unique site on 50S ribosome) | Inhibits isoleucyl-tRNA synthetase | Inhibits elongation factor G (EF-G) |
| Standard Dosing | Twice daily for 5 days | Three times daily for up to 10-12 days | Three to four times daily for 7 days |
| Spectrum (Clinical) | MSSA, S. pyogenes | MSSA, MRSA, S. pyogenes | MSSA, S. pyogenes |
| MRSA Activity | Ineffective in clinical trials | Clinically effective (resistance is a concern) | Resistance rates are high in many regions |
| Potential for Resistance | Considered low | Emerging resistance is a known concern | Widespread resistance reported globally |
| Cost Profile | Higher (Brand name) | Lower (Generic available) | Varies (Not available in US) |
Across a pooled analysis of clinical trials involving over 2,100 patients, retapamulin was found to be generally well-tolerated.[8]
Table 4: Summary of Reported Adverse Events (≥1% Incidence) from Pooled Clinical Trials
| Adverse Event | Retapamulin 1% Ointment (N=2,115) | Oral Cephalexin (N=819) | Placebo (N=71) |
|---|---|---|---|
| Application Site Irritation | 1.4% | N/A | 0% |
| Diarrhea | <2% | 1.7% | N/A |
| Application Site Pruritus | <2% | N/A | 1.4% |
| Headache | 1-2% | N/A | N/A |
| Nausea | 1% (adults) | N/A | N/A |
| Nasopharyngitis | 1-2% | N/A | N/A |
| 3 |
Drug interactions with topical retapamulin are limited due to its low systemic absorption. However, one clinically significant interaction has been identified and requires specific management, particularly in the pediatric population.
The small fraction of retapamulin that is systemically absorbed is primarily metabolized by the hepatic enzyme CYP3A4.[7] Consequently, co-administration with drugs that strongly inhibit this enzyme can lead to increased plasma concentrations of retapamulin. A clinical study in healthy adult males demonstrated that co-administration of oral ketoconazole (a strong CYP3A4 inhibitor) increased the systemic exposure (both
AUC and Cmax) of topically applied retapamulin by 81%.[10] Other strong CYP3A4 inhibitors include itraconazole, clarithromycin, ritonavir, and grapefruit juice.[25]
The clinical management of this interaction is nuanced and based directly on the age-dependent pharmacokinetic profile of retapamulin.
The concurrent use of retapamulin with other topical products on the same area of skin has not been studied and is therefore not recommended.[10]
Patients or caregivers should be instructed on the proper use of retapamulin ointment to ensure efficacy and safety.
Retapamulin was developed by GlaxoSmithKline and submitted to the U.S. Food and Drug Administration (FDA) for review.[2] The New Drug Application (NDA) was accepted for review on February 14, 2006.[46] On
April 12, 2007, the FDA approved retapamulin 1% ointment under the brand name Altabax for the topical treatment of impetigo.[1] At the time, its approval was notable as it represented the first new class of prescription topical antibacterial to be approved in the United States in nearly 20 years.[2] More recent reports indicate that the Altabax brand has since been discontinued in the U.S. market, suggesting a commercial withdrawal.[25]
Following its approval in the U.S., retapamulin was granted marketing authorisation in the European Union by the European Medicines Agency (EMA) on May 24, 2007, under the brand name Altargo.[1] The indication in the EU was for the short-term treatment of impetigo as well as infected small lacerations, abrasions, or sutured wounds.[44]
However, on February 25, 2019, the European Commission officially withdrew the marketing authorisation for Altargo in the EU.[13] This action was taken at the request of the marketing authorisation holder, Glaxo Group Ltd. The company cited purely
"commercial reasons" for its decision to permanently discontinue the product, and the withdrawal was not related to any new safety or efficacy concerns.[13]
The regulatory lifecycle of retapamulin, particularly its voluntary withdrawal from the major European market, serves as a compelling case study on the distinction between regulatory success and commercial success. Despite achieving regulatory approval based on sufficient evidence of safety and efficacy, retapamulin struggled to gain a significant foothold in the market. It was positioned as a niche alternative to well-established, inexpensive generic topical antibiotics like mupirocin and fusidic acid.[3] Several factors likely contributed to this outcome: its higher cost as a branded product [3]; the critical clinical limitation of being ineffective against MRSA infections [12]; and the strategic failure to generate head-to-head clinical trial data against the primary standard of care, mupirocin.[3] For a new antibiotic in a market with entrenched, low-cost options, demonstrating non-inferiority is often insufficient to drive adoption. Without a clear advantage in a difficult-to-treat population or proven superiority over the standard of care, its novel mechanism alone was not enough to ensure commercial viability.
Retapamulin represents a significant scientific achievement in antibacterial drug development, marking the successful introduction of the pleuromutilin class into human medicine. Its unique mechanism of action, targeting a novel site on the bacterial ribosome, provided a sound scientific basis for its development, offering a low potential for cross-resistance with other antibiotic classes and potent in vitro activity against key skin pathogens.
Clinically, its strengths are well-documented. It demonstrated clear superiority over placebo and non-inferiority to active comparators like sodium fusidate and oral cephalexin in treating uncomplicated superficial skin infections. Furthermore, its favorable safety profile, characterized by minimal systemic absorption and predominantly local side effects, combined with a convenient twice-daily, 5-day dosing regimen, offered tangible advantages for patient compliance.
However, the trajectory of retapamulin also highlights critical challenges that can impede the success of a new antimicrobial agent. The most significant of these was the "MRSA paradox"—the stark failure to translate excellent in vitro activity against this formidable pathogen into meaningful clinical efficacy. This single limitation profoundly constrained its therapeutic potential, preventing it from filling a crucial unmet need in treating resistant infections. Compounding this was the absence of robust, direct comparative data against mupirocin, the established and less expensive standard of care. This evidence gap left clinicians and healthcare systems with little compelling reason to adopt a more costly alternative for routine infections.
In conclusion, retapamulin is a clinically effective and safe topical antibiotic whose scientific novelty did not fully translate into sustained market success. Its story underscores a crucial reality in modern pharmaceutical development: regulatory approval is only the first hurdle. To thrive in a competitive therapeutic area with established, low-cost generics, a new agent must demonstrate a clear and compelling value proposition—be it through superior efficacy, an improved safety margin, or, most critically for an antibiotic, reliable effectiveness in a resistant patient population where other options fail. The commercial withdrawal of retapamulin from major markets suggests that, despite its scientific merits, it was ultimately unable to clear this high bar.
Published at: September 27, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.