MedPath

FLAMOD Advanced Drug Monograph

Published:Jun 16, 2025

Generic Name

FLAMOD

FLAMOD (Nebulized Recombinant Flagellin): A Comprehensive Report on a First-in-Class Host-Directed Adjunct Therapy for Drug-Resistant Respiratory Infections

Executive Summary

FLAMOD is an investigational, first-in-class immunomodulatory biologic being developed as an adjunct therapy for severe respiratory infections, specifically targeting the escalating global crisis of antimicrobial resistance (AMR). The active pharmaceutical ingredient is a recombinant form of flagellin, a potent bacterial protein, which is administered as an aerosol directly to the lungs via a vibrating mesh nebulizer. This localized delivery is a key strategic element, designed to maximize therapeutic effect at the site of infection while minimizing systemic side effects.

The drug's mechanism of action is fundamentally different from conventional antibiotics. Instead of directly targeting the pathogen, FLAMOD functions as a host-directed therapy by stimulating the innate immune system. As a specific agonist of Toll-like receptor 5 (TLR5), it triggers a rapid, localized, and controlled immune cascade within the lung tissue. This response enhances the body's natural defenses, leading to the recruitment of neutrophils and the production of antimicrobial peptides, thereby augmenting the efficacy of co-administered antibiotics.

The development of FLAMOD is a notable example of an academically driven, publicly funded initiative, spearheaded by the European Union-backed FAIR (Flagellin Aerosol therapy as an Immunomodulatory adjunct to the antibiotic treatment of drug-Resistant bacterial pneumonia) consortium. This multi-year, multi-institution collaboration has successfully advanced the program from concept through comprehensive preclinical validation in multiple animal models to its current stage: a Phase I clinical trial known as NEBUFLAG. This trial, which commenced in March 2025, is designed not only to assess safety but also to meticulously characterize the drug's pharmacodynamic effects in humans, a sophisticated approach for an early-stage study.

Supported by a robust preclinical data package and a strong, multi-layered intellectual property portfolio, FLAMOD represents a paradigm-shifting approach to treating drug-resistant pneumonia. If proven successful, it could offer a critically needed tool to restore the effectiveness of existing antibiotics and address one of the most pressing unmet medical needs in modern medicine.

Section 1: The Clinical Imperative for Novel Anti-Infective Strategies

1.1 The Global Burden of Respiratory Infections and the Shadow of Antimicrobial Resistance (AMR)

The development of FLAMOD is set against the backdrop of a severe and worsening global health crisis. Respiratory infections are the third leading cause of mortality worldwide, claiming an estimated three million lives annually.[1] Pneumonia, an acute inflammatory condition of the lung alveoli most often caused by bacteria or viruses, is a primary driver of this lethality, affecting individuals across the entire lifespan, from young children to the elderly.[4] For decades, the cornerstone of managing bacterial pneumonia has been antibiotic therapy. However, the efficacy of this entire class of drugs is being systematically eroded by the rise of antimicrobial resistance (AMR).

The World Health Organization (WHO) has identified AMR as one of the top global public health threats facing humanity. Projections indicate that if current trends continue, bacterial infections caused by drug-resistant pathogens could become the leading cause of death globally by the year 2050, surpassing even cancer.[5] This looming post-antibiotic era threatens to render common infections untreatable and routine medical procedures, such as surgery and chemotherapy, dangerously risky. The increasing prevalence of multidrug-resistant pathogens like

Pseudomonas aeruginosa, Klebsiella pneumoniae, and Streptococcus pneumoniae in hospital and community settings creates a profound and urgent unmet medical need for new therapeutic strategies.[9] It is this critical therapeutic gap that FLAMOD is designed to address, positioning it not merely as an incremental improvement but as a potentially vital tool in the global fight against AMR.[12]

1.2 The Rationale for Host-Directed Immunomodulatory Therapies

In response to the limitations of pathogen-directed drugs, a new therapeutic paradigm has emerged: host-directed therapy. FLAMOD is a leading example of this innovative approach. Unlike a conventional antibiotic that directly targets and kills a specific bacterium, FLAMOD is an immunomodulator designed to stimulate the patient's own innate immune system to fight the infection.[1] This strategy offers several fundamental advantages.

First, by targeting a conserved host pathway—the innate immune response—the therapy has the potential for broad-spectrum activity. The immune mechanisms mobilized by FLAMOD are not specific to a single bacterial species or resistance profile, suggesting it could be effective against a wide range of pathogens, including those resistant to multiple classes of antibiotics.[5] This circumvents the problem of pathogen-specific resistance mechanisms that render traditional antibiotics obsolete.

Second, FLAMOD is intended for use as an adjunct to, not a replacement for, standard-of-care antibiotics.[1] The goal is to enhance the effectiveness of first-line antibiotics, potentially restoring their utility against resistant strains and allowing for the use of lower or shorter courses of treatment, which could in itself help mitigate the future development of resistance. This adjunctive approach facilitates a more straightforward path to clinical integration.

The development of FLAMOD is built on the recognition that the innate immune system, particularly within the specialized environment of the respiratory tract, represents a powerful but historically underexploited target for drug discovery in infectious disease.[7] The strategic decision to pursue a host-directed immunomodulatory agent like FLAMOD is therefore a direct response to the scientific and clinical realities of the AMR crisis. This positions the drug program as a potential case study for how public and academic initiatives can tackle critical health challenges that may be neglected by market forces more traditionally focused on chronic, rather than acute, diseases. The narrative of FLAMOD is thus inextricably linked to the global AMR threat, a linkage that provides a powerful rationale for its continued development and a compelling value proposition for regulatory bodies, funding agencies, and potential future commercial partners.

Section 2: FLAMOD - Pharmaceutical Profile and Mechanism of Action

2.1 Drug Substance and Formulation

The investigational drug FLAMOD is a highly defined biologic agent, the product of precise molecular engineering and formulation science.

The active pharmaceutical ingredient is a recombinant form of flagellin, a protein that is the principal component of bacterial flagella.[15] Specifically, the molecule is FliCΔ174-400, derived from the FliC flagellin of

Salmonella enterica serovar Typhimurium.[9] The "Δ174-400" designation is of critical importance; it signifies that the central, hypervariable region of the native protein, which constitutes its primary antigenic domain, has been deleted through recombinant DNA technology. This molecular engineering is a key design feature intended to reduce the molecule's intrinsic antigenicity. By removing the main target for the adaptive immune system, the risk of patients developing neutralizing antibodies against the drug is minimized. This is a crucial consideration for any therapeutic biologic, as it allows for the possibility of repeated administrations over the course of treatment without a loss of efficacy due to immune clearance.[9]

The clinical-grade (GMP) drug substance is manufactured by the Statens Serum Institut in Denmark, using a well-established E. coli expression system where the protein is produced in inclusion bodies and subsequently purified.[22] The final drug product is a highly soluble protein formulated in a specific buffer solution consisting of 10 mM phosphate, 145 mM NaCl at a pH of 6.5, and containing 0.02% (w/v) polysorbate 80.[19] The inclusion of polysorbate 80, a non-ionic surfactant, is vital for stabilizing the protein structure and preventing aggregation or denaturation during the high-shear stress process of aerosolization. The specificity and importance of this formulation are underscored by its protection under a dedicated patent, WO2023275292, which adds a significant layer to the overall intellectual property estate of the program.[21]

2.2 Detailed Mechanism of Action: A TLR5-Mediated Cascade

FLAMOD exerts its therapeutic effect by acting as a potent and specific agonist for Toll-like receptor 5 (TLR5).[9] TLR5 is a pattern recognition receptor (PRR), a key component of the innate immune system responsible for detecting conserved molecular structures from microbes. In the respiratory tract, TLR5 is expressed on the surface of airway epithelial cells, which are the principal drivers of the initial response, as well as on resident immune cells such as alveolar macrophages.[9]

The mechanism of action unfolds through a well-defined signaling cascade:

  1. Binding and Activation: When FLAMOD is delivered to the airways, the flagellin protein binds to TLR5 on the surface of lung cells.
  2. Signal Transduction: This binding event initiates a MyD88-dependent intracellular signaling pathway, which is the canonical pathway for most TLRs. This cascade culminates in the activation and nuclear translocation of the master inflammatory transcription factor, NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells).[9]
  3. Transcriptional Response: Once in the nucleus, NF-κB orchestrates the transcription of a wide array of genes involved in host defense. This results in the rapid production and secretion of a suite of immune mediators, creating a potent pro-inflammatory and antimicrobial environment localized to the lung tissue.
  4. Effector Molecule Production: The specific mediators produced include pro-inflammatory cytokines such as interleukin-6 (IL-6), interleukin-1 beta (IL-1β), and granulocyte-macrophage colony-stimulating factor (GM-CSF); chemokines designed to recruit immune cells, such as CXCL1, CXCL2, CXCL5, and CCL20; and directly antimicrobial peptides like β-defensins.[9]
  5. Cellular Recruitment: The ultimate effector phase of this response is the robust recruitment and activation of neutrophils, the primary phagocytic cells of the innate immune system, into the airways. This influx of neutrophils is critical for engulfing and destroying invading bacteria, thereby enhancing pathogen clearance.[9]

A crucial feature of this TLR5-mediated response is that it is both potent and transient. The signaling pathway includes strong negative feedback mechanisms that ensure the inflammatory response is self-limiting, peaking within hours and decaying shortly thereafter, which allows the tissue to return to homeostasis and prevents excessive, damaging inflammation.[9]

2.3 Pharmacological Rationale for Aerosol Delivery

FLAMOD is being developed not as a standalone drug but as an integrated drug-device combination product. It is specifically designed for administration as an aerosol via a vibrating mesh nebulizer, with the Aerogen Solo device being used in the clinical program.[1] This delivery route is a cornerstone of the therapeutic strategy.

The primary rationale for aerosol delivery is to achieve high local drug concentrations directly at the site of infection—the lung epithelium—while minimizing systemic absorption and exposure.[1] This approach is intended to maximize the localized immune-stimulating effect where it is needed most, thereby enhancing therapeutic efficacy, while simultaneously avoiding the potential for systemic side effects that could arise from widespread immune activation. Preclinical studies have provided strong validation for this strategy, demonstrating that nebulization leads to a more rapid resolution of systemic pro-inflammatory markers compared to other routes of administration, confirming a superior safety profile for this targeted approach.[13]

The integrated nature of this program—combining a specifically engineered biologic, a patented stabilizing formulation, and a validated delivery device—represents a sophisticated and forward-thinking development strategy. A competitor would face the challenge of not only developing a new flagellin-like molecule but also a new formulation and delivery system capable of safely and effectively aerosolizing it. This multi-layered approach creates a high barrier to entry and establishes a strong intellectual property moat, significantly enhancing the value and potential market exclusivity of the FLAMOD program.

Section 3: Comprehensive Preclinical Evaluation of FLAMOD

The clinical development of FLAMOD is underpinned by a comprehensive and strategically designed preclinical program. These studies, conducted across multiple animal models and against various pathogens, have not only established the drug's proof-of-concept but have also provided critical insights that have directly shaped the clinical trial design.

3.1 Proof-of-Concept in Murine Models of Pneumonia

The foundational evidence for FLAMOD's therapeutic potential was generated in mouse models of bacterial pneumonia. These studies were instrumental in establishing the core principle of using flagellin as an adjunct to antibiotic therapy.

In a key study, mice were infected with a multidrug-resistant strain of Pseudomonas aeruginosa. Prophylactic intranasal administration of flagellin was shown to restore the antimicrobial efficacy of the antibiotic gentamicin, which was otherwise ineffective against this resistant strain.[9] This provided the first direct evidence that stimulating the host's innate immunity could overcome established antibiotic resistance.

Similarly, in models of pneumonia caused by antibiotic-resistant Streptococcus pneumoniae, flagellin demonstrated clear therapeutic synergy when combined with the antibiotic amoxicillin.[9] This synergy was a crucial finding, as it validated the "adjunct therapy" concept that forms the basis of FLAMOD's intended clinical use.

A pivotal study published in 2024 directly compared nebulized delivery to the intranasal route used in earlier experiments.[13] This study yielded two critical findings for the program's translation to the clinic. First, it revealed that while nebulization resulted in dramatically lower deposition of the drug in the lungs (less than 1% of the administered dose) compared to intranasal delivery (approximately 40%), it still induced a comparable local innate immune response. This suggests that FLAMOD is highly potent at its target site and that only a small amount is needed to trigger the desired biological effect. Second, and equally important, the nebulized route led to an accelerated resolution of systemic pro-inflammatory responses. This confirmed that localized aerosol delivery provides a superior safety profile by minimizing systemic effects, a key de-risking step for human trials. The study concluded by confirming that nebulized flagellin, when used as an adjunct to amoxicillin, was effective against resistant pneumococcal pneumonia in mice, providing the final piece of preclinical validation for the chosen drug-device combination.

3.2 Validation in a Physiologically Relevant Porcine Model

To bridge the gap between small animal models and human clinical trials, the FAIR consortium conducted studies in pigs, a large animal model with a respiratory system more analogous to that of humans. These studies provided invaluable data on safety and refined the therapeutic strategy.

A 2025 study was the first to assess FLAMOD delivered by nebulization in pigs. The results were highly encouraging, demonstrating that a single nebulization of the drug was safe and did not cause any adverse clinical effects.[21] Pharmacodynamic analysis confirmed that the mechanism of action observed in mice was conserved in this large animal model. The study documented immune cell infiltration, specifically granulocyte recruitment, into the lung tissue and demonstrated broad activation of immune-related genes throughout the respiratory tract, from the nose to the deep lung.[21]

A second porcine study, also from 2025, investigated the critical question of therapeutic timing.[19] This experiment yielded a crucial, program-defining result. When FLAMOD was used therapeutically—administered after infection as an adjunct to a subtherapeutic dose of penicillin G—it significantly improved the clearance of the pathogen

Actinobacillus pleuropneumoniae and was associated with a trend toward fewer lung lesions. However, when the same dose of FLAMOD was administered prophylactically 24 hours before the bacterial challenge, it failed to confer protection and was, in fact, associated with more severe lung lesions.

This finding, while seemingly negative, provided an invaluable piece of strategic intelligence. It demonstrated that ill-timed immune priming could be detrimental, potentially by creating an excessive inflammatory environment that the subsequent infection could exploit. This result effectively ruled out a prophylactic indication, preventing a potentially costly and unsuccessful clinical trial and focusing the entire program on the adjunct therapeutic strategy, which had been consistently successful across all preclinical models. This demonstrates a mature, hypothesis-driven approach to preclinical development, where studies are designed not just to confirm efficacy but to proactively identify and mitigate risks before entering human trials.

3.3 Preclinical Safety and Toxicology Overview

The collective preclinical data supports a favorable safety profile for FLAMOD. The recombinant flagellin itself is not classified as a hazardous substance according to standard safety data sheets.[34] The broader FAIR project explicitly included formal modeling of toxicological doses as part of its five-year work plan to prepare for the first-in-human trial, indicating a rigorous and systematic approach to safety evaluation.[1] The consistent observation across both mouse and pig models is that the immune response triggered by FLAMOD is transient and localized, with minimal systemic spillover, a safety profile that is significantly enhanced by the use of nebulized delivery.[13]

Study ReferenceAnimal ModelPathogenIntervention(s)Key Efficacy/Pharmacodynamic FindingsKey Safety/Strategic Findings
Maia AR, et al. Antimicrob Agents Chemother. 2024 9MouseMultidrug-resistant Pseudomonas aeruginosaProphylactic intranasal flagellin + gentamicinFlagellin restored the efficacy of gentamicin against a resistant strain, decreasing bacterial load and inflammation.Established proof-of-concept for flagellin to overcome antibiotic resistance.
Baldry M, et al. Antimicrob Agents Chemother. 2024 13MouseAntibiotic-resistant Streptococcus pneumoniaeNebulized flagellin + amoxicillin vs. Intranasal flagellinNebulized route induced a comparable local immune response (cytokine/neutrophil influx) to intranasal route despite <1% lung deposition. Combination therapy was effective against resistant pneumonia.Nebulization is a viable and potent delivery route. Confirmed superior safety profile with accelerated resolution of systemic inflammation via nebulization.
Baldry M, et al. bioRxiv. 2025 21PigN/A (Healthy)Single nebulization of FLAMODConfirmed safety with no adverse clinical effects. Induced robust immune cell infiltration and immune gene activation throughout the porcine respiratory tract.Validated the drug's mechanism of action and safety in a large, translationally relevant animal model.
Fleurot I, et al. bioRxiv. 2025 19PigActinobacillus pleuropneumoniaeTherapeutic FLAMOD + Penicillin G vs. Prophylactic FLAMODTherapeutic adjunct strategy significantly improved bacterial clearance. Prophylactic strategy failed to protect and was associated with worse lung lesions.Critically important finding that focused the clinical development strategy exclusively on the adjunct therapeutic setting, de-risking future trials.

Section 4: The Clinical Pathway: FAIR Project and the NEBUFLAG Trial

4.1 The FAIR Consortium: A Model for Academically Driven, EU-Funded Development

The journey of FLAMOD from a laboratory concept to a clinical-stage asset is a testament to a unique and successful development model driven by public funding and academic collaboration. The entire program is encompassed within the FAIR project, an acronym for Flagellin Aerosol therapy as an Immunomodulatory adjunct to the antibiotic treatment of drug-Resistant bacterial pneumonia.[5]

FAIR is a large-scale research initiative funded by a €10 million grant from the European Union's Horizon 2020 program, with a project timeline spanning from January 2020 to June 2025.[7] What distinguishes this project is its predominantly academic and non-profit structure, a departure from the typical industry-led drug development path.[1] The consortium represents a pan-European collaboration of eleven partners across seven countries, each contributing specialized expertise:

  • Sponsor and Lead: The French National Institute of Health and Medical Research (Inserm) serves as the project's official sponsor and coordinator.[7]
  • Core Development: The Center for Infection and Immunity of Lille (CIIL) and its researchers, notably Dr. Jean-Claude Sirard, are credited with the foundational discovery and development of FLAMOD.[1]
  • Clinical Execution: The Tours University Hospital in France is the sole clinical site for the Phase I trial.[1]
  • Manufacturing: The Statens Serum Institut in Denmark is responsible for producing the GMP-grade biologic drug substance.[15]
  • Device Partner: Aerogen, a commercial entity, provides the specialized vibrating mesh nebulizer technology essential for the drug's delivery.[15]
  • Dissemination and Advocacy: The European Respiratory Society (ERS) and European Lung Foundation (ELF) are key partners responsible for disseminating findings and engaging with patient and healthcare communities.[15]

This collaborative structure has enabled the project to overcome the significant financial and logistical hurdles that often prevent promising academic discoveries from reaching clinical evaluation—a phenomenon often referred to as the "valley of death" in drug development.[11]

4.2 The NEBUFLAG Phase I Clinical Trial

The culmination of the FAIR project's five years of preclinical work is the NEBUFLAG trial, the first-in-human study of FLAMOD.[1]

Trial Identity and Status: The study is formally titled "Phase I Single-blind Clinical Trial to Evaluate the Safety and Local Immune Activation of a Toll-like Receptor 5 Agonist (FLAMOD) Administered by Aerosol".[24] It is a monocentric trial being conducted exclusively at the Clinical Investigation Centre of the Tours University Hospital in France, with Professor Antoine Guillon serving as the Principal Investigator.[1] The trial began recruiting in March 2025 and is projected to conclude by late 2026.[2]

Study Design: The NEBUFLAG trial employs a sophisticated design for a Phase I study. It is a single-blind, randomized, placebo-controlled, dose-escalation study that will enroll an estimated 60 healthy adult volunteers.[24] The trial is structured in two distinct stages:

  1. Start-up Phase: A traditional dose-escalation phase to ensure the safety of single, ascending doses of FLAMOD.
  2. Dose-finding Phase: This stage utilizes the data gathered to inform several dose-activity models, allowing for a data-driven selection of the optimal dose for subsequent cohorts, rather than simply escalating to the highest tolerable dose.

Objectives and Endpoints: The trial's objectives go far beyond a standard safety assessment, incorporating a deep investigation into the drug's mechanism of action in humans.

  • Primary Endpoint: The primary objective is to evaluate the safety and tolerability of a single aerosolized dose of FLAMOD. This is measured by the proportion of participants experiencing Serious Adverse Events (SAEs), critical safety events, laboratory abnormalities, and cytokine release syndrome, as defined by CTCAE v5.0.[24]
  • Secondary Endpoints: The secondary endpoints are designed to provide a rich pharmacodynamic dataset, effectively making this a human proof-of-mechanism study. Key endpoints include:
  • Determination of the Immune Activating Dose (IAD): A primary goal is to identify the lowest dose of FLAMOD that reliably triggers an immune response. This will be assessed using systemic biomarker surrogates such as blood levels of IL-6, C-reactive protein (CRP), procalcitonin (PCT), and neutrophils.[24]
  • Characterization of the Local Immune Response: The trial will perform multiomic analyses (proteomics and transcriptomics) on samples collected directly from the nasal mucosa via nasosorption and curettage. This will provide a detailed molecular picture of the local immune-enhancing effects, focusing on key mediators like IL-6, IL-8, and CCL20, as well as broader changes in gene expression pathways.[24]
  • Characterization of the Systemic Immune Response: Corresponding multiomic analyses will be performed on blood samples to create a comprehensive profile of the systemic immune signature induced by the drug.[24]

This intense focus on defining an "Immune Activating Dose" based on biological activity, rather than merely a "Maximum Tolerated Dose" based on toxicity, is a hallmark of modern, rational drug development. It reflects a sophisticated strategy to select a Phase II dose that is most likely to be effective in patients. The comprehensive biomarker data generated will be invaluable for guiding future trials, potentially identifying patient stratification markers or response monitoring tools, thereby significantly de-risking the program's transition into more complex and costly patient studies.

Participant Criteria: The trial enrolls a tightly controlled population of healthy, non-smoking adults between the ages of 18 and 65. The extensive exclusion criteria are designed to eliminate confounding factors, such as pre-existing pulmonary disease, immune compromise, recent infections or vaccinations, or use of medications that could alter immune responses (e.g., steroids, anticoagulants). This ensures that any observed effects can be confidently attributed to the investigational drug.[6]

ParameterDescription
Trial Name / AcronymNEBUFLAG / Phase I Single-blind Clinical Trial to Evaluate the Safety and Local Immune Activation of a Toll-like Receptor 5 Agonist (FLAMOD) Administered by Aerosol
PhasePhase I
SponsorInstitut National de la Santé Et de la Recherche Médicale (Inserm) 4
Lead Clinical SiteClinical Investigation Centre, Tours University Hospital, France 1
Principal InvestigatorProfessor Antoine Guillon, MD 1
Study DesignSingle-blind, randomized, placebo-controlled, single-dose, dose-escalation, monocentric 24
Population & SizeHealthy volunteers, 18-65 years old; N=60 (estimated) 24
Intervention ArmsFLAMOD (recombinant flagellin) administered by aerosol vs. Placebo administered by aerosol 24
Primary EndpointSafety and tolerability (assessed via incidence of AEs, SAEs, lab abnormalities, cytokine release syndrome) 24
Key Secondary EndpointsDetermination of Immune Activating Dose (IAD) via systemic biomarkers (IL-6, CRP, neutrophils); Local immune profiling (proteomics/transcriptomics of nasal samples); Systemic immune profiling (proteomics/transcriptomics of blood samples) 24
Current StatusActive, Recruiting 24
Estimated TimelineStart: March 2025; Estimated Completion: Late 2026 2

Section 5: Strategic Analysis, Future Outlook, and Intellectual Property

5.1 Competitive Landscape and Key Differentiators

FLAMOD is being developed in a therapeutic area of immense need but with a unique competitive positioning. As a first-in-class host-directed immunomodulatory adjunct, its primary "competitor" is not another drug with a similar mechanism, but rather the current, and increasingly inadequate, standard of care: antibiotic monotherapy. The key differentiating attributes that define its value proposition are rooted in its novel scientific approach.

  • Novel Mechanism of Action: The most significant differentiator is FLAMOD's ability to circumvent conventional bacterial resistance mechanisms. By stimulating a broad-based host immune response rather than targeting a specific bacterial enzyme or structure, its efficacy is theoretically independent of a pathogen's antibiotic susceptibility profile.[1]
  • Localized Delivery for Enhanced Safety: The drug-device combination, delivering FLAMOD directly to the lungs via nebulization, is a critical feature. This strategy is designed to concentrate the immune-stimulating effect at the site of infection while minimizing systemic exposure, a concept strongly supported by preclinical data showing an improved safety profile over other delivery routes.[1]
  • Adjunctive Strategy for Clinical Integration: FLAMOD is designed to complement, not replace, existing antibiotics. This approach is strategically sound, as it aims to restore the efficacy of established, familiar drugs, which may facilitate easier adoption into clinical practice compared to a completely novel standalone therapy.[2]
  • Broad-Spectrum Potential: Because it targets a universal host pathway (TLR5 signaling), FLAMOD has the potential to be effective against a wide variety of bacterial pathogens. This offers a significant advantage over narrow-spectrum antibiotics that are only effective against specific bacteria.[5]

5.2 Intellectual Property Portfolio

A robust intellectual property (IP) portfolio is a cornerstone of any biotechnology program, and the developers of FLAMOD have established a strong, multi-layered position. This IP estate is a core asset that underpins the program's long-term value.

  • Use Patents: A foundational family of patents, including WO2009156405, WO2011161491, WO2015011254, and US8889149B2, covers the core concept of using flagellin as a biologic to prevent and treat infectious diseases, particularly in the respiratory tract. These patents list key researchers like Dr. Jean-Claude Sirard as inventors, securing the fundamental therapeutic application.[14]
  • Formulation Patent: Critically, the IP strategy extends beyond the general use of the biologic. Patent WO2023275292 specifically protects the aerosol formulation used for the pulmonary delivery of FLAMOD.[14] This is a highly valuable asset, as the ability to create a stable, deliverable formulation for a biologic is a significant technical challenge.

This dual IP strategy, protecting both the molecule's use and its specific delivery formulation, creates a formidable barrier to competition. A potential competitor would need to not only invent a new, non-infringing flagellin-like molecule but also independently develop and validate a new formulation and delivery system. This significantly raises the technical and financial bar for entry, thereby enhancing and extending the potential market exclusivity for FLAMOD.

5.3 Future Clinical Development and Regulatory Path

The immediate future of the FLAMOD program hinges on the outcomes of the ongoing NEBUFLAG Phase I trial. A successful data readout, demonstrating a favorable safety profile and clear evidence of dose-dependent immune activation, will be the next major value inflection point for the program.

Following a successful Phase I, the logical clinical development path would proceed to Phase II trials in patient populations. These studies would likely focus on indications with the highest unmet need, such as ventilator-associated pneumonia (VAP) or community-acquired pneumonia caused by documented drug-resistant bacteria. The rich biomarker data being generated in NEBUFLAG will be critical for designing these trials, allowing for the rational selection of a dose, schedule, and potentially a patient subpopulation most likely to benefit.

The transition to larger, global Phase II and III trials will require financial and operational resources that typically exceed the capacity of an academic consortium. Therefore, a key strategic imperative for the FAIR project leadership will be to secure a partnership with a mid-to-large-sized pharmaceutical or biotechnology company. The comprehensive preclinical and Phase I data package being meticulously assembled is clearly designed to support such a transaction, providing a potential partner with a de-risked asset with a well-defined mechanism and a clear clinical path forward.

5.4 Concluding Assessment

FLAMOD represents a highly innovative and scientifically rigorous program that directly addresses the critical global threat of antimicrobial resistance. It is a first-in-class asset distinguished by its novel host-directed mechanism, its localized delivery system, and its potential for broad-spectrum adjunctive activity.

The development of FLAMOD to date has been characterized by a mature and sophisticated translational strategy. The preclinical program was well-designed, yielding not only proof-of-concept but also critical insights that have de-risked and focused the clinical plan. The ongoing NEBUFLAG Phase I trial is exceptionally well-conceived, aiming to generate a rich biomarker dataset that will be invaluable for guiding late-stage development. This is complemented by a strong, multi-layered intellectual property portfolio that provides a durable competitive advantage.

The academic-led, EU-funded model has proven remarkably successful in advancing FLAMOD from a scientific concept to a clinical-stage asset, a notable achievement. While the program remains in early-stage development, it possesses all the hallmarks of a high-potential therapeutic. Its future trajectory will be determined by the successful completion and data readout from the NEBUFLAG trial (expected after 2026) and the subsequent ability of the consortium to forge a strategic partnership to carry the program through late-stage development and commercialization. FLAMOD is a compelling and scientifically sound program that warrants close monitoring by all stakeholders in the anti-infective space.

Works cited

  1. FLAMOD Enters Phase I Trial: A New Innovation Against Respiratory Infections, accessed June 16, 2025, https://www.ciil.fr/actualites/news-details-1?tx_news_pi1%5Baction%5D=detail&tx_news_pi1%5Bcontroller%5D=News&tx_news_pi1%5Bnews%5D=289&cHash=0a244d1ad79e0bd8d269fa32e8825062
  2. FAIR project reaches new milestone as phase I clinical trial of innovative new drug begins, accessed June 16, 2025, https://www.fair-flagellin.eu/component/content/article/8-news/78-fair-project-reaches-new-milestone-as-phase-i-clinical-trial-of-innovative-new-drug-begins
  3. Farmaco innovativo contro le infezioni respiratorie. Avviata sperimentazione clinica in Francia - insalutenews.it, accessed June 16, 2025, https://www.insalutenews.it/in-salute/farmaco-innovativo-contro-le-infezioni-respiratorie-avviata-sperimentazione-clinica-in-francia/
  4. [RECHERCHE DE VOLONTAIRES] Le CHRU de Tours au cœur d'une innovation thérapeutique contre les infections respiratoires, accessed June 16, 2025, https://www.chu-tours.fr/recherche-de-volontaires-le-chru-de-tours-au-coeur-dune-innovation-therapeutique-contre-les-infections-respiratoires/
  5. fair-flagellin.eu, accessed June 16, 2025, https://fair-flagellin.eu/
  6. NEBUFLAG - CHRU de Tours, accessed June 16, 2025, https://www.chu-tours.fr/wp-content/uploads/2025/04/CP_Recherche-Projet-FAIR-Essai-clinique-I.pdf
  7. FAIR project: investigating treatments for antibiotic-resistant pneumonia - ERS, accessed June 16, 2025, https://www.ersnet.org/news-and-features/news/fair-project-investigating-treatments-for-antibiotic-resistant-pneumonia/
  8. Event - CEPR, accessed June 16, 2025, https://cepr-inserm.fr/category/event/
  9. Preventive nasal administration of flagellin restores antimicrobial ..., accessed June 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11064517/
  10. Bacteria Antibiotics and Immunity - - Center for Infection & Immunity of Lille, accessed June 16, 2025, https://www.ciil.fr/teams/bacteria-antibiotics-and-immunity
  11. Jean-Claude Sirard: Towards a Drug of Bacterial Origin to Fight Pneumonia - Inserm, accessed June 16, 2025, https://www.inserm.fr/en/news/jean-claude-sirard-towards-a-drug-of-bacterial-origin-to-fight-pneumonia/
  12. FLAGELLIN AEROSOL THERAPY AS AN IMMUNOMODULATORY ADJUNCT TO THE ANTIBIOTIC TREATMENT OF DRUG-RESISTANT BACTERIAL PNEUMONIA | H2020 | CORDIS, accessed June 16, 2025, https://cordis.europa.eu/project/id/847786/reporting
  13. Targeted delivery of flagellin by nebulization offers optimized respiratory immunity and defense against pneumococcal pneumonia - PubMed, accessed June 16, 2025, https://pubmed.ncbi.nlm.nih.gov/39480071/
  14. Targeted delivery of flagellin by nebulization offers optimized respiratory immunity and defense against pneumococcal pneumonia - PMC, accessed June 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11619323/
  15. FLAMOD against respiratory infections: phase I clinical trial of innovative new drug begins, accessed June 16, 2025, https://firstwordpharma.com/story/5943497
  16. Project - FAIR Website, accessed June 16, 2025, https://fair-flagellin.eu/project
  17. FAIR Project Reaches Key Milestone with Launch of Phase I Clinical Trial - CEPR, accessed June 16, 2025, https://cepr-inserm.fr/2025/04/29/fair-project-reaches-key-milestone-with-launch-of-phase-i-clinical-trial/
  18. FLAMOD against respiratory infections | PULSE International, accessed June 16, 2025, https://pulsepakistan.com/flamod-against-respiratory-infections/
  19. (PDF) Flagellin aerosol administration improves the efficacy of antibiotic treatment in Actinobacillus pleuropneumoniae infected pigs - ResearchGate, accessed June 16, 2025, https://www.researchgate.net/publication/390019360_Flagellin_aerosol_administration_improves_the_efficacy_of_antibiotic_treatment_in_Actinobacillus_pleuropneumoniae_infected_pigs
  20. Flagellin aerosol administration improves the efficacy of antibiotic treatment in Actinobacillus pleuropneumoniae infected pigs - bioRxiv, accessed June 16, 2025, https://www.biorxiv.org/content/10.1101/2025.03.17.643685v1.full.pdf
  21. Flagellin nebulization enhances respiratory immune responses in the porcine model, accessed June 16, 2025, https://www.biorxiv.org/content/10.1101/2025.03.17.643810v1
  22. Flagellin aerosol administration improves the efficacy of antibiotic treatment in Actinobacillus pleuropneumoniae infected pigs | bioRxiv, accessed June 16, 2025, https://www.biorxiv.org/content/10.1101/2025.03.17.643685v1.full-text
  23. Flagellin nebulization enhances respiratory immune responses in the porcine model, accessed June 16, 2025, https://www.researchgate.net/publication/390023087_Flagellin_nebulization_enhances_respiratory_immune_responses_in_the_porcine_model
  24. Phase I Single-blind Clinical Trial to Evaluate the Safet, accessed June 16, 2025, https://clinicaltrial.be/en/details/458131?per_page=20&only_recruiting=0&only_eligible=0&only_active=0
  25. Flagellin nebulization enhances respiratory immune responses in the porcine model | bioRxiv, accessed June 16, 2025, https://www.biorxiv.org/content/10.1101/2025.03.17.643810
  26. Jean-Claude Sirard PhD Microbiology - CV Science, accessed June 16, 2025, https://cvscience.aviesan.fr/cv/635/jean-claude-sirard
  27. FLA-PA Ultrapure | Flagellin from Pseudomonas aeruginosa | InvivoGen, accessed June 16, 2025, https://www.invivogen.com/fla-pa
  28. Neutrophil subsets enhance the efficacy of host-directed therapy in pneumococcal pneumonia - PubMed, accessed June 16, 2025, https://pubmed.ncbi.nlm.nih.gov/39592068/
  29. The GM-CSF Released by Airway Epithelial Cells Orchestrates the Mucosal Adjuvant Activity of Flagellin - PubMed, accessed June 16, 2025, https://pubmed.ncbi.nlm.nih.gov/33008952/
  30. Targeted delivery of flagellin by nebulization offers optimized respiratory immunity and defense against pneumococcal pneumonia | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed June 16, 2025, https://journals.asm.org/doi/10.1128/aac.00866-24
  31. Targeted delivery of flagellin by nebulization offers optimized respiratory immunity and defense against pneumococcal pneumonia | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed June 16, 2025, https://journals.asm.org/doi/abs/10.1128/aac.00866-24
  32. Flagellin nebulization enhances respiratory immune responses in the porcine model - Sciety, accessed June 16, 2025, https://sciety.org/articles/activity/10.1101/2025.03.17.643810
  33. Flagellin aerosol administration improves the efficacy of antibiotic treatment in Actinobacillus pleuropneumoniae infected pigs | bioRxiv, accessed June 16, 2025, https://www.biorxiv.org/content/10.1101/2025.03.17.643685v1
  34. safety data sheet - The Native Antigen Company, accessed June 16, 2025, https://thenativeantigencompany.com/wp-content/uploads/2018/12/SDS-Borrelia-Flagellin.pdf
  35. Launch of the NEBUFLAG study at Tours University Hospital - CEPR, accessed June 16, 2025, https://cepr-inserm.fr/2025/01/30/launch-of-the-nebuflag-study-at-tours-university-hospital/
  36. EU Projects - European Lung Foundation, accessed June 16, 2025, https://europeanlung.org/en/projects-and-campaigns/eu-projects/
  37. Phase I Single-blind Clinical Trial to Evaluate the Safety and Local Immune Activation of a Toll-like Receptor 5 Agonist (FLAMOD) Administered by Aerosol (NEBUFLAG) - ClinicalTrials.Veeva, accessed June 16, 2025, https://ctv.veeva.com/study/phase-i-single-blind-clinical-trial-to-evaluate-the-safety-and-local-immune-activation-of-a-toll-lik
  38. Development of the first-in-human trial for nebulisation of FLAMOD - Vienna 2024, accessed June 16, 2025, https://channel.ersnet.org/media-108537-development-of-the-first-in-human-trial-for-nebulisation-of-flamod
  39. US8889149B2 - Use of flagellin to prevent and treat gram negative bacterial infection - Google Patents, accessed June 16, 2025, https://patents.google.com/patent/US8889149B2/en
  40. (12) United States Patent - Googleapis.com, accessed June 16, 2025, https://patentimages.storage.googleapis.com/b2/4f/20/97e1ef36db5054/US8889149.pdf

Published at: June 16, 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.