84012-15-7
Partial Thickness Wounds
Birch bark extract (DrugBank ID: DB16536) is a complex botanical substance that has achieved a remarkable transition from a traditional ethnobotanical remedy to a regulated, evidence-based pharmaceutical product. This monograph provides a comprehensive analysis of its scientific and clinical profile. The extract is a multi-component mixture, predominantly composed of pentacyclic triterpenoids, which collectively mediate its therapeutic effects. The culmination of modern scientific investigation into this traditional remedy is the marketing authorization of Filsuvez® (also known as Oleogel-S10), a topical oleogel formulation of the extract. This product is specifically indicated for the treatment of partial thickness wounds associated with two severe subtypes of epidermolysis bullosa (EB): dystrophic EB (DEB) and junctional EB (JEB), in patients aged 6 months and older.[1]
The approval of Filsuvez® in major regulatory jurisdictions, including the European Union and the United Kingdom, represents a significant milestone in botanical drug development. It serves as a successful case study for a development pipeline that has rigorously validated traditional knowledge through modern clinical science to address a substantial unmet medical need in a rare and devastating genetic disease. Unlike many plant-derived medicines that rely on the isolation of a single active molecule, the therapeutic agent in this case is the well-characterized, complex extract itself. This approach acknowledges the potential for synergistic interactions among multiple constituents and provides a powerful precedent for the development and registration of other multi-component botanical therapies, which have historically faced challenges navigating regulatory pathways designed for single chemical entities. This report will detail the journey of Birch bark extract from its botanical source to its clinical application, elucidating the chemistry, pharmacology, and clinical evidence that underpin its therapeutic use.
This report aims to provide an exhaustive, expert-level analysis of Birch bark extract (DB16536; CAS Number: 84012-15-7). The scope encompasses a multidisciplinary review of its botanical sourcing and phytochemistry, the intricate molecular pharmacology and mechanism of action, the pivotal clinical trial evidence supporting its efficacy, the unique pharmacokinetic challenges and formulation science, and a comprehensive safety and risk profile. The primary objective is to synthesize this information into a definitive scientific reference document that is valuable for pharmaceutical scientists, clinical researchers, dermatologists, pharmacognosists, and other professionals in the medical and life sciences fields.
The use of birch bark in medicine is not a recent discovery but is deeply rooted in centuries of traditional practice across multiple cultures. Historically, various parts of the birch tree, particularly the bark and leaves, have been employed in folk medicine for their purported diuretic, anti-inflammatory, and antimicrobial properties.[4] The most prominent traditional application, which directly foreshadowed its modern pharmaceutical use, was in the treatment of skin ailments, including wounds, sores, rashes, and burns.[7]
This ethnopharmacological history is well-documented. In North America, indigenous groups utilized the bark extensively; for instance, the Cree used the outer bark as a bandage for burns and in ointments for persistent scabs, while the Maliseet and Mi'kmaq applied it to infected cuts.[7] This knowledge was not confined to one continent; the medicinal consumption of birch extracts dates back to the Middle Ages in Europe, where the bark was applied to accelerate wound healing.[5] Its use is also noted in traditional systems as geographically diverse as Indian Ayurvedic medicine.[5]
The transition from traditional remedy to scientific inquiry began with the dawn of modern chemistry. A pivotal milestone occurred in 1788 when the German-Russian chemist Johann Tobias Lowitz first isolated and characterized a white, crystalline substance from the bark.[9] This compound, later named betulin, was identified as the primary constituent responsible for the bark's characteristic white color and was a foundational discovery in the field of natural product chemistry. This early isolation of a key active ingredient laid the groundwork for subsequent scientific investigation into the pharmacological properties of birch bark, ultimately leading to the highly specific and clinically validated therapeutic agent available today.
The pharmaceutical-grade Birch bark extract is derived from specific species of the Betula L. genus, belonging to the Betulaceae family.[12] The primary botanical sources are
Betula pendula Roth, commonly known as the silver birch, and Betula pubescens Ehrh., the white birch.[1] Hybrids of these two species are also utilized for production. The extract is obtained specifically from the outer, white layer of the bark (the cortex), which is particularly rich in the bioactive triterpenoid compounds.[1] While these Eurasian species are the main sources for the approved drug product, other species, such as the North American paper birch (
Betula papyrifera), also contain these compounds and are used in other commercial extracts, though the relative concentrations of key constituents can vary by species, geographical origin, and age of the tree.[13] This variability underscores the importance of standardized sourcing and processing for producing a consistent pharmaceutical-grade extract.
The production of Birch bark extract involves sophisticated extraction and purification processes designed to isolate and concentrate the desired triterpenoid fraction. A common laboratory and industrial method is solvent reflux extraction, using solvents or solvent mixtures such as ethanol or a combination of chloroform, dichloromethane, and methanol, followed by recrystallization to purify the final product.[11] The efficiency of this process can be enhanced through physical methods; for example, the application of ultrasonic treatment during reflux can disrupt the bark's cellular structure, thereby improving both the extraction yield and the purity of the final betulin-rich extract.[11]
More advanced, "green chemistry" techniques such as supercritical CO2 fluid extraction have also been developed. This method uses carbon dioxide under high pressure and temperature as a solvent, offering a more environmentally friendly alternative to organic solvents. The resulting product is a dry triterpene extract, which serves as the active pharmaceutical ingredient (API) in formulations like Filsuvez®.[9]
Birch bark extract is a complex mixture of natural compounds. While it is overwhelmingly dominated by one class of molecules, a full phytochemical analysis reveals the presence of several groups, including terpenoids, steroids (e.g., β-sitosterol), tannins, flavonoids (e.g., kaempferol), essential oils, and hydrocarbons.[4] The primary drivers of the extract's pharmacological activity, however, are the pentacyclic triterpenoids of the lupane skeletal type, which can constitute over 95% of the final composition in some high-purity extracts.[1]
The biological activity and pharmaceutical development of Birch bark extract are fundamentally governed by the physicochemical properties of its constituent triterpenoids. A central characteristic of these molecules is their high lipophilicity and consequently poor aqueous solubility. This property is a double-edged sword: it allows for interaction with cellular membranes but presents a significant challenge for drug delivery and bioavailability. The development pathway of the approved drug product can be understood as a direct response to these intrinsic chemical properties. The decision to formulate the extract as an oleogel—a suspension in sunflower oil—is not an arbitrary choice but a rational and necessary strategy to create a stable, deliverable formulation for these highly lipophilic active compounds.[2] This direct link between the fundamental chemistry of the constituents and the design of the final pharmaceutical product highlights a core principle of formulation science. The key properties of the most abundant and pharmacologically significant triterpenoids are summarized in Table 1.
Table 1: Physicochemical Properties of Key Triterpenoids in Birch Bark Extract
| Property | Betulin | Betulinic Acid | Lupeol |
|---|---|---|---|
| IUPAC Name | (1R,3aS,5aR,5bR,7aR,9S,11aR,11bR,13aR,13bR)-3a-(hydroxymethyl)-5a,5b,8,8,11a-pentamethyl-1-prop-1-en-2-yl-1,2,3,4,5,6,7,7a,9,10,11,11b,12,13,13a,13b-hexadecahydrocyclopenta[a]chrysen-9-ol 19 | (1R,3aS,5aR,5bR,7aR,9S,11aR,11bR,13aR,13bR)-9-hydroxy-5a,5b,8,8,11a-pentamethyl-1-prop-1-en-2-yl-1,2,3,4,5,6,7,7a,9,10,11,11b,12,13,13a,13b-hexadecahydrocyclopenta[a]chrysen-3a-carboxylic acid 21 | lup-20(29)-en-3-ol 14 |
| Molecular Formula | C30H50O2 19 | C30H48O3 21 | C30H50O 13 |
| Molecular Weight | 442.72 g/mol 19 | 456.7 g/mol 21 | 426.7 g/mol 13 |
| CAS Number | 473-98-3 11 | 472-15-1 9 | 545-47-1 13 |
| Appearance | White crystalline powder; off-white powder 11 | White to off-white solid powder 22 | White powder 13 |
| Melting Point | 251-257 °C 11 | 295-318 °C (dec.) 22 | 215-216 °C 13 |
| Solubility | Insoluble in water; soluble in ethanol, chloroform, benzene, diethyl ether, DMSO 11 | Insoluble in water; soluble in DMSO, ethanol 22 | Insoluble in water 13 |
The mechanism of action of Birch bark extract in promoting wound healing is complex and, while not yet fully elucidated, has been shown to involve a sophisticated, biphasic modulation of the healing process.[1] This mechanism departs significantly from simple anti-inflammatory or pro-proliferative actions. Instead, it appears to orchestrate the distinct phases of wound healing, beginning with a controlled inflammatory response and transitioning to a robust regenerative phase. This dual action represents a more intelligent approach to wound management than mere suppression of inflammation.
Contrary to the conventional paradigm of treating wounds with anti-inflammatory agents, Birch bark extract initiates its effect by transiently enhancing the acute inflammatory response.[7] This initial phase is critical for preparing the wound bed for healing. The extract, and particularly its main component betulin, has been shown to temporarily upregulate the expression of several key pro-inflammatory mediators in human keratinocytes, including cytokines like Interleukin-6 (IL-6) and Interleukin-8 (IL-8), as well as the enzyme cyclooxygenase-2 (COX-2).[18] This controlled inflammatory burst serves a vital physiological purpose: it orchestrates the recruitment of immune cells such as macrophages and granulocytes to the wound site, which are essential for clearing cellular debris, removing dead tissue, and preventing bacterial infection.[18]
The molecular pathway underlying this effect involves post-transcriptional regulation. Studies have demonstrated that the extract and betulin increase the messenger RNA (mRNA) levels of COX-2 and IL-6 not by increasing gene transcription, but by stabilizing the existing mRNA molecules, thus prolonging their functional lifespan and leading to greater protein production. This mRNA stabilization process is mediated through the activation of the p38 Mitogen-Activated Protein Kinase (MAPK) signaling pathway. Activation of p38 MAPK, in turn, is believed to promote the translocation of the RNA-binding protein HuR (Human antigen R) from the nucleus to the cytoplasm, where it binds to and stabilizes the mRNA of pro-inflammatory mediators.[18] This transient, controlled pro-inflammatory action kickstarts the healing cascade in a productive manner.
Following the initial inflammatory phase, the extract's action shifts to promoting the proliferative and remodeling phases of wound healing, primarily by acting on keratinocytes, the main cells of the epidermis.[1] The extract induces keratinocyte differentiation, a process necessary for building a new, functional skin barrier. This is evidenced by the increased expression of key differentiation markers such as keratin 10 (KRT10) and involucrin (INV).[1]
Crucially, the extract significantly enhances the migration of keratinocytes, enabling them to move into the wound bed and close the defect more rapidly.[7] This pro-migratory effect is underpinned by the extract's ability to induce profound changes in the cellular architecture. It promotes the rearrangement of the actin cytoskeleton to form structures essential for cell motility, such as filopodia (slender, finger-like projections) and lamellipodia (broad, sheet-like extensions at the leading edge of a migrating cell). This process is dependent on the activation of the Rho family of GTPases, which are master regulators of the actin cytoskeleton.[18]
Interestingly, this second phase of action highlights the synergistic nature of the extract's composition. While betulin is the primary driver of the initial inflammatory modulation, the pro-migratory effects are potently elicited not only by betulin but also by the minor constituents lupeol and erythrodiol. These compounds have been shown to be effective at inducing these cytoskeletal changes even at low, nanomolar concentrations.[18] This observation provides a strong rationale for the use of the whole extract as the API; the combination of multiple active compounds, each potentially targeting different facets of the healing process (inflammation versus migration), leads to a more comprehensive and robust therapeutic effect than could be achieved by any single isolated molecule. Porcine ex-vivo wound healing models have shown that while pure betulin has a beneficial effect, it is less pronounced than that of the total extract, further supporting this concept of chemical synergy.[18]
Beyond the specific biphasic mechanism in wound healing, the individual triterpenoid constituents of Birch bark extract possess a wide range of other pharmacological activities that have been extensively studied in preclinical models.
The primary and approved therapeutic indication for Birch bark extract, formulated as the topical gel Filsuvez®, is for the treatment of partial thickness wounds in patients with epidermolysis bullosa (EB).[1] EB is a group of rare, inherited skin fragility disorders caused by mutations in genes encoding structural proteins that anchor the epidermis to the dermis. This genetic defect results in extremely fragile skin that blisters and erodes in response to minor friction or trauma, leading to chronic, painful, and difficult-to-heal open wounds.[2] The license for Birch bark extract is specific to two of the major subtypes: dystrophic EB (DEB), where the cleavage plane is below the basement membrane in the upper dermis, and junctional EB (JEB), where separation occurs within the basement membrane zone.[1]
Prior to the approval of Filsuvez®, the standard of care for EB was entirely supportive and palliative, focused on meticulous and painful daily wound care, dressing changes, and management of complications like infection and pain. There were no approved treatments that specifically targeted and accelerated the healing of EB wounds. Therefore, Birch bark extract addresses a significant unmet medical need for this patient population, offering the first therapy demonstrated to improve wound closure in this devastating condition.[2]
The regulatory approval of Birch bark extract gel was primarily based on the results of the EASE (Efficacy and Safety of Oleogel-S10 in Epidermolysis Bullosa) study, a large, robust, Phase III clinical trial.[2] The study employed a sophisticated and methodologically sound design well-suited for a rare and highly variable disease. It was a double-blind, randomized, vehicle-controlled trial with an intra-patient comparison. In this design, pairs of comparable wounds on the same patient were randomly assigned to receive either the active Birch bark extract gel (Oleogel-S10) or the control gel (the vehicle, consisting of 90% sunflower oil), allowing each patient to serve as their own control. This approach is powerful because it minimizes the impact of inter-patient variability in disease severity, genetic background, and other confounding factors, thereby increasing the statistical power to detect a true treatment effect even with the smaller patient populations typical of rare disease research. The study enrolled patients aged 6 months and older with DEB or JEB who had target wounds of a specific size and age.[1]
The primary endpoint of the trial was the proportion of patients who achieved the first complete closure of their target wound within a 45-day assessment period. The EASE study successfully met this primary endpoint, demonstrating that a statistically significantly larger proportion of wounds treated with Birch bark extract gel healed completely compared to those treated with the control gel.[2]
The trial also assessed several important secondary endpoints that further supported the clinical benefit of the treatment. These included the reduction in the total burden of wounds, measured by the change in the body surface area percentage (BSAP) covered by wounds, and changes in the EB Disease Activity and Scarring Index (EBDASI). The results suggested potential improvements in these areas, as well as a reduced frequency of required dressing changes. While the effect on other relevant outcomes like pain and itching was less clear from the primary trial phase, the overall evidence strongly supported the efficacy of the extract in accelerating wound closure.[2]
To assess the long-term effects of the treatment, patients who completed the 90-day double-blind phase of the EASE study were eligible to enter a 24-month open-label phase (OLP), during which all partial thickness wounds could be treated with Birch bark extract gel. The results from this extension phase provided crucial data on the durability of the treatment effect and its long-term safety profile.[35]
The OLP data demonstrated a sustained reduction in the overall wound burden over the 24-month period. Patients who were treated with the active gel throughout the entire study showed statistically significant reductions in total wound BSAP from their original baseline at 3, 12, and 24 months. Similarly, significant improvements in the EBDASI skin activity score were maintained over the long term.[35] These findings confirm that the wound healing benefits observed in the short-term trial are not transient but are sustained with continued use. Furthermore, treatment adherence during the OLP was exceptionally high (over 99%), suggesting good tolerability and acceptance of the therapy by patients and their caregivers.[35]
The evidence base for Birch bark extract's efficacy in wound healing is further supported by earlier-phase clinical studies and preclinical models. An open, prospective, randomized Phase II clinical trial involving 24 patients demonstrated that a birch bark preparation significantly accelerated the re-epithelialization of split-thickness skin graft donor sites, a standardized model of a partial thickness wound.[8]
In addition to formal trials, case reports have described the successful use of birch bark preparations in challenging clinical scenarios, including the treatment of severe necrotizing herpes zoster (shingles) in a patient who had failed conventional therapies, and in two patients suffering from second-degree burns.[7] These clinical observations are corroborated by data from porcine ex-vivo wound healing models, which showed positive wound healing effects and provided a platform for elucidating the molecular mechanisms of action that were later confirmed in human keratinocyte studies.[8] Early research has also suggested potential efficacy in treating actinic keratosis, a type of pre-cancerous skin growth caused by sun damage.[37]
Table 2: Summary of Key Efficacy and Safety Endpoints from the EASE Clinical Trial
| Endpoint Type | Endpoint Description | Birch Bark Extract (Oleogel-S10) Result | Control Gel Result | Statistical Significance / Notes |
|---|---|---|---|---|
| Primary Efficacy | Proportion of patients with first complete target wound closure by Day 45 | Statistically significantly higher proportion | Lower proportion | p<0.05 2 |
| Secondary Efficacy | Change in Body Surface Area Percentage (BSAP) of wounds from baseline at 24 months | Mean reduction of -3.7% | N/A (OLP data) | p=0.003 vs baseline 35 |
| Secondary Efficacy | Change in EBDASI skin activity score from baseline at 24 months | Mean reduction of -3.0 | N/A (OLP data) | p=0.007 vs baseline 35 |
| Safety (OLP) | Patients reporting any Adverse Event (AE) | 77.1% | N/A (OLP data) | Most AEs were mild-to-moderate and related to underlying EB 35 |
| Safety (OLP) | Patients reporting Severe AEs | 18.0% | N/A (OLP data) | 35 |
| Safety (OLP) | Patients reporting Serious AEs | 24.4% | N/A (OLP data) | 35 |
| Safety (OLP) | Treatment-related withdrawals | 3 patients (out of 205) | N/A (OLP data) | 35 |
| Safety (OLP) | Target wound infections | Low incidence (n=7) | N/A (OLP data) | Majority were mild-to-moderate 35 |
A central challenge in the development of Birch bark extract for systemic applications is the inherent pharmacokinetic properties of its primary constituents. The pentacyclic triterpenes—betulin, lupeol, and betulinic acid—are large, rigid, and highly lipophilic molecules. This chemical nature results in extremely low solubility in aqueous environments like the gastrointestinal tract, which severely limits their absorption and systemic bioavailability when administered orally.[10] This poor solubility is the primary barrier to their use as conventional oral drugs. Furthermore, these compounds can be susceptible to degradation and oxidation, which can further reduce the amount of active substance that reaches systemic circulation.[38]
Recognizing this significant hurdle, considerable preclinical research has been dedicated to developing strategies to enhance the bioavailability of these triterpenes. These approaches can be broadly categorized into physical modification, chemical modification, and advanced formulation strategies.[38]
The development and success of the approved product, Filsuvez®, exemplifies a highly pragmatic and effective approach to overcoming the pharmacokinetic challenges of Birch bark extract. Instead of undertaking the complex and lengthy process of developing a systemically bioavailable oral drug, the developers focused on creating an optimized formulation for local, topical delivery, directly addressing the clinical need for wound healing. This strategy effectively sidesteps the systemic bioavailability problem for its approved indication.
Filsuvez® is a non-aqueous cutaneous gel composed of 10% dry Birch bark extract as the active pharmaceutical ingredient, suspended in a vehicle of 90% refined sunflower oil.[2] This oleogel formulation is a masterful example of aligning the properties of the API with the needs of the clinical indication. The sunflower oil vehicle provides an ideal lipophilic matrix for the triterpenes, ensuring they are solubilized, stable, and can be efficiently delivered directly to the target tissue—the wound bed.
The formulation also possesses advantageous physical properties. It is described as having thixotropic characteristics, meaning the viscous gel liquefies upon the application of shear stress (i.e., when it is spread on the skin) and returns to its gel state at rest. This property facilitates a gentle and easy application, which is critically important for patients with the extremely fragile skin characteristic of EB.[2] In some contexts, the triterpene extract itself can act as both an active ingredient and a stabilizer, allowing for very simple formulations that may not require traditional emulsifiers or preservatives.[9] This demonstrates that the formulation is not merely a passive carrier but an integral component of the therapeutic product, as critical to its clinical success as the active botanical extract itself.
The safety profile of Birch bark extract is highly dependent on the route of administration and the specific formulation. It is crucial to distinguish between the well-documented safety data for the approved topical gel used in clinical trials and the broader precautions associated with the systemic (oral) use of birch preparations in traditional or supplemental contexts.
Based on the robust data from the pivotal EASE Phase III trial and its 24-month open-label extension, the topical Birch bark extract gel is generally well-tolerated.[3] The overall tolerability profile was found to be similar to that of the control gel (vehicle), indicating that the extract itself does not add significant local irritation.[3]
The safety considerations for orally consumed birch preparations (e.g., teas, supplements) are substantially different from those for the topical gel due to systemic exposure to the extract's constituents.
This clear bifurcation in the safety profile is critical for clinical practice. The risks associated with the approved topical formulation are primarily local and manageable, whereas the risks of systemic use are related to diuretic and salicylate effects, requiring a different set of clinical considerations.
Table 3: Comprehensive Safety and Interaction Profile
| Route of Admin. | Finding Type | Description | Source Snippets |
|---|---|---|---|
| Topical (Filsuvez®) | Side Effects | Generally well-tolerated. Local wound site reactions (pain, pruritus), often indistinguishable from underlying disease. | 3 |
| Topical (Filsuvez®) | Risks | Potential for hypersensitivity/allergic reaction. Low incidence of wound infections. Theoretical risk of skin malignancies. | 1 |
| Systemic/Oral | Side Effects | Diuretic effects, potential nausea, allergic reactions (rash, itching). | 5 |
| Systemic/Oral | Contraindications | Impaired cardiac or renal function. Hypersensitivity to aspirin. | 40 |
| Systemic/Oral | Drug Interactions | Diuretics (additive effect, risk of dehydration/hypotension). Aspirin (cumulative salicylate effect). | 37 |
| Systemic/Oral | Precautions | Use with caution in hypertension. Avoid in pregnancy/lactation. Cross-allergy with celery, carrot, nuts, etc. | 5 |
Beyond its pharmaceutical application, Birch bark extract is widely utilized in the cosmetics and skincare industry due to the multifaceted beneficial properties of its constituents. It is incorporated into a variety of formulations, including creams, serums, lotions, and toners, where it serves multiple functions.[32]
The approval of Birch bark extract for EB marks a significant achievement, but it may only be the first chapter in its therapeutic story. The rich phytochemistry of the extract provides a valuable source of bioactive compounds that hold promise for development in other therapeutic areas. The extract can be viewed as a "platform" with dual potential: first, as a multi-component topical agent for epithelial repair with applications beyond EB, and second, as a natural chemical library for the discovery and isolation of lead compounds for new systemic drugs.
Birch bark extract (DB16536) represents a triumph of modern pharmaceutical science in validating and refining a traditional botanical remedy. Its journey from the forests of Eurasia to a regulated, approved therapy for the rare and severe genetic disorder epidermolysis bullosa is a testament to rigorous phytochemical characterization, mechanistic elucidation, and well-designed clinical investigation.
The therapeutic efficacy of the extract is rooted in a sophisticated, biphasic mechanism of action that intelligently modulates the wound healing process—initiating a transient pro-inflammatory phase to cleanse the wound, followed by a robust pro-proliferative phase to promote re-epithelialization. This action is driven by the synergistic interplay of multiple triterpenoid constituents, primarily betulin, lupeol, and betulinic acid. The formidable challenge of these compounds' poor water solubility was pragmatically overcome through the development of a scientifically rational oleogel formulation, which ensures stable and effective local delivery to the wound bed. The resulting product, Filsuvez®, has a favorable safety profile for topical use and has been proven in a pivotal Phase III trial to significantly accelerate wound healing, addressing a long-standing unmet medical need for the EB community.
Looking forward, the value of Birch bark extract extends beyond its current indication. It stands as a platform for further therapeutic innovation. As a topical agent, its fundamental mechanism of promoting epithelial repair suggests potential applications in other challenging wound healing scenarios, such as burns and skin graft sites. As a chemical reservoir, it provides an abundant source of highly bioactive molecules like betulinic acid, which are promising lead compounds for the development of next-generation therapies in fields as diverse as oncology and virology. Thus, the ancient birch tree, long valued in traditional medicine, is now firmly established as a source of modern, evidence-based therapeutics with a promising future.
Published at: September 19, 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.