1. Introduction
Acne vulgaris (AV) is a highly prevalent polymorphic dermatological condition that causes the formation of a series of diverse lesion morphologies [
1,
2,
3]. This condition mainly affects areas of the skin with a higher number of pilosebaceous units, such as the face and back [
4,
5]. Each pilosebaceous unit is comprised of a sebaceous gland (SG) and a hair follicle (HF). SGs are cutaneous appendages that secrete sebum (a lipid cocktail) into the follicular canal [
1]. In skin affected by AV, normal differentiation and proliferation of keratinocytes are disrupted due to the occlusion of pilosebaceous structures with sebum. This is often coupled with bacterial overgrowth that results in an inflammatory response.
Retinoids, or vitamin A derivatives, have been the gold standard in acne therapy since the 1960–1970s [
6,
7]. Biologically, retinoids regulate keratinocyte turnover, epidermal growth and differentiation, and inflammatory responses. These properties make them ideal for the treatment of various skin diseases [
8]. The different generations of retinoids are typically described on the basis of their affinity for retinoic acid receptors (RAR). Among them, adapalene, a third-generation synthetic retinoid, is well established for its anti-proliferative, comedolytics, and anti-inflammatory properties. These properties are mediated through high-affinity binding to RARs.
Although adapalene was approved by the US Food and Drug Administration (FDA) (in 1996) for topical treatment for mild to moderate acne vulgaris in patients 12 years or older, it has also been used for verrucae, molluscum contagiosum, Darier disease, Fox–Fordyce disease, Dowling–Degos disease, photoaging, pigmentary disorders, actinic keratoses, and alopecia areata [
9,
10]. As a naphthoic acid derivative, adapalene has fewer off-target effects, attributed to its rigid structure, which also enhances tolerability and stability compared to first- and second-generation retinoids [
7]. Its high lipophilicity (logP = 6.917) and moderate molecular weight (412.2 g/mol) enable preferential partitioning into lipophilic regions of the skin, particularly the stratum corneum (SC) and hair follicles (HF), as demonstrated by in vitro studies using confocal laser scanning microscopy [
11]. Adapalene is mainly absorbed through the trans-appendageal and intercellular routes [
10]. Adapalene is marketed in three concentrations and formulations: 0.1% gel (over the counter (OTC)), 0.1% lotion and cream (prescription only), and 0.3% gel (prescription only). In this paper, we focus on three over-the-counter 0.1% gel formulations: Differin® (Galderma), designated as the FDA’s Reference Listed Drug (RLD) in the Orange Book, and two test products, AcneFree and Effaclar.
Pharmacokinetic studies using topical adapalene have shown minimal systemic exposure, as adapalene mainly distributes within the epidermis. A study using Differin cream 0.1% on six acne patients showed no quantifiable amount of adapalene in plasma (limit of quantification: 0.35 ng/mL) after five days of daily application [
12]. Another clinical study using 0.3% Differin® gel reported low but detectable plasma concentrations, with Cmax of 0.553 ± 0.466 ng/mL and a mean AUC of 8.37 ± 8.46 ng h/mL on day 10 [
13]. The reported half-life of adapalene is 7 to 51 h. Although, human adapalene metabolism is not fully understood, it has been reported that only 25% of adapalene is metabolized (mostly glucuronides), the remainder is excreted in its original form through the biliary route at 30 ng/g of the topically applied dose [
7,
14]. Its rapid clearance is another reason why topical adapalene is typically systemically undetectable [
12].
In conventional pharmacokinetics, bioavailability is the rate and extent to which a drug reaches systemic circulation from the administered dosage form. However, systemic quantification becomes analytically challenging for some topically administered drugs due to the skin’s barrier function. The low permeability and absorption of topical drugs leads to plasma levels below the limit of quantification (LOQ) [
15]. Using typical general pharmacokinetic models to estimate pharmacokinetic parameters is challenging in this setting in regards to non-compartmental analysis (NCA) [
16,
17]. To address this limitation, physiologically based pharmacokinetic (PBPK) modeling offers a mechanistic alternative. PBPK models use mathematical representations of drug transport, partitioning, and metabolism based on anatomical and physiological parameters.
In this study, we adapt a dermal PBPK model to simulate and quantify the in vitro bioavailability of three 0.1% adapalene gel formulations. This model represents the biophysical and physiological processes that govern dermal absorption more mechanistically, including drug penetration, partitioning, and diffusion across multiple skin layers, with their unique intrinsic properties. This dermal PBPK model uses a series of interconnected compartments that replicate the anatomical characteristics of the skin (
Figure 1), with mass transport between compartments represented by differential equations and mechanistic principles of mass balance. High resolution morphology is incorporated using a brick-and-mortar approach, which explicitly distinguishes between corneocytes and lipid domains in SC [
18]. To evaluate systemic distribution metrics, a holistic dermal model was integrated with a whole-body PBPK model through dermis microcirculation. To replicate experimental conditions and in vitro permeation studies, the model also incorporates formulation-specific parameters such as drug release kinetics, skin thickness, and receptor volume [
19]. As a first-principles-based approach, the dermal PBPK model can be used independently or in conjunction with full-body models to estimate dermal pharmacokinetics and support comparative evaluations.
The primary objective of this study was to evaluate and compare the dermal absorption and bioavailability of adapalene from three marketed gel formulations, including Differin® (reference), AcneFree, and Effaclar (test products), using in vitro permeation data, in silico simulations, and clinical observations of epidermal metrics. Ultimately, this work aims to advance formulation development, improve understanding of topical drug disposition, and inform regulatory strategies to establish topical bioequivalence.
4. Discussion
The FDA defines a product as bioequivalent when the rate and extent of absorption of the test drug do not show a significant difference from those of the reference drug (listed) when administered at the same molar dose of the therapeutic ingredient and under similar experimental conditions, either as a single dose or multiple doses [
36]. According to the product-specific guidance (PSG) for 0.1% adapalene gel, bioequivalence can be assessed using one of two approaches. The first option involves conducting an in vitro bioequivalence study, accompanied by additional characterization tests. The second option recommends an in vivo bioequivalence study using a clinical endpoint [
37].
According to the first option outlined in the FDA’s PSG for adapalene gel, there should be no difference in the inactive ingredients or other formulation aspects of the test product compared to the reference standard, provided both are in the same packaging format (tube or pump). This consistency is essential, as such differences may significantly affect the local or systemic availability of the active ingredient. Additionally, the test and reference products, in identical packaging configurations, must exhibit an equivalent rate of adapalene release. This requirement should be demonstrated through an acceptable IVRT bioequivalence study, comparing at least one batch of the test product to one batch of the reference standard using a suitably validated IVRT method [
37].
In the present study, several aspects of the gel formulations could not be fully analyzed, including the exact composition. Although a comparison of active and inactive ingredients was performed for the three gels (
Table 4), the precise formulations, specifically, the percentages of weight and volume of each ingredient, remain unknown, as this information is proprietary and not publicly available. In addition, the specific grades of the ingredients could not be determined. No studies were conducted to evaluate the morphology or critical quality attributes (CQA), such as the pH, viscosity, or specific gravity of the gels.
IVRT experiments were conducted according to PSG recommendations: a 24 h, single-dose, parallel design with multiple replicates per treatment group, using a synthetic membrane in a diffusion cell system. According to the PSG guidance, the analyte measured was the receptor solution. The release kinetics of adapalene differed notably among the three gel formulations. Differin exhibited rapid and quantifiable release in 30 min, while AcneFree and Effaclar showed delayed detection, with adapalene only becoming measurable after 4 h and remaining below quantifiable limits at earlier time points. Across the various time points, the release of adapalene from Differin was consistently higher than that of AcneFree and Effaclar. Only by the 24-h mark did Effaclar demonstrate a release profile comparable to Differin. These findings suggest significant differences in the release behavior between gels and underscore the importance of further investigating the equivalence of Q1, Q2, and Q3, to better understand their formulation-driven release kinetics.
Although IVPT is not recommended in the PSG for adapalene, it was deemed necessary in the context of this study to support the overarching goal: understanding the dermal pharmacokinetics of adapalene and developing a validated dermal PBPK model. Since adapalene is administered topically and not intravenously or orally, systemic circulation has limited relevance to its dermal disposition. As such, IVPT provided critical information on the absorption, distribution, and permeation of the drug within the skin.
The IVPT results demonstrated a consistently higher distribution of adapalene in the epidermis in all three gel formulations. With a logP of 6.9, adapalene is highly lipophilic. SC, the outermost layer of the epidermis, is rich in lipids and keratinocytes, making it a favorable environment for lipophilic molecules. However, differences in formulation composition, microstructure, and critical quality attributes (CQA), such as viscosity and pH, can significantly influence drug permeation profiles.
As noted earlier, Differin exhibited a faster and higher release of adapalene in the IVRT studies. This trend was mirrored in the IVPT results, where significantly higher concentrations of adapalene were detected in the dermis, along with trace levels in the receptor compartment. In contrast, the dermis concentrations of adapalene for the AcneFree and Effaclar gels were considerably lower. These findings suggest that the observed differences in dermal delivery may be attributed to formulation-specific properties, which warrant further investigation, particularly with respect to the equivalence of Q1, Q2, and Q3, as well as the characterization of CQA.
Although the primary site of action for adapalene in the treatment of acne vulgaris is within the epidermis and hair follicles, where it promotes cellular turnover and removes keratin plugs, emerging in vitro evidence suggests broader biological effects. Studies using cell and tissue culture systems have reported that adapalene may stimulate collagen and elastin production, delay or reverse dermal thinning, facilitate wound healing, and exert immunomodulatory effects [
7,
33,
38]. Given that Differin delivered higher levels of adapalene to the dermis, these additional pharmacological effects may be particularly relevant in the treatment of chronic or inflamed acne, potentially offering additional therapeutic benefits beyond conventional expectations.
The FDA PSG non-binding guideline suggests either conducting IVRT or an in vivo study with a clinical endpoint to establish bioequivalence for adapalene gel 0.1% [
37]. The PSG recommends the following criteria: a randomized, placebo-controlled trial design and selection of a study population with acne vulgaris. While we followed the PSG exclusion criteria and avoidance of confounding variables (such as application of other topical products to the skin during the observation period), the study was not designed to fit the guidelines for bioequivalence. Our study was designed to identify key physiological responses to adapalene using a novel non-invasive imaging technique. We sought to explain these physiological effects of adapalene on healthy skin by monitoring changes over time.
Our clinical study showed that the Differin formulation produced the most significant decrease in SC thickness from baseline at the 48 h time point, which correlates with our PK model, which showed higher rates of drug progression to the dermis. Interestingly, there was no significant improvement in epidermal/dermal thickness, which is consistent with the literature reporting adapalene effects with OCT imaging [
39]. While all formulations showed decreased infundibular to isthmus ratio at the 4–6 h timepoint, the Acne Free formulation had the most statistically significant decrease (−54.4%) compared to Differin (−27.3%) and Effaclar (−25%). This effect may be explained by Acne Free’s increased potency, as noted by a lower EC50 compared to the other formulations. However, at the 48 h timepoint, all formulations showed a comparable, and significant decrease in percentage (%) infundibular/isthmus ratio compared to baseline (Differin, −50%; Effaclar, −50%; Acne Free, −54.8%).
Studies with in vitro human keratinocytes microscopy have shown that from the
stratum basale differentiation, keratinocyte size increases up to 10 times once reaching the SC [
40]. Adapalene is reported to inhibit proliferation and/or normalize differentiation of keratinocytes. However, our study failed to show any significant difference in keratinocyte count or size at the level of the stratum spinosum for any of the subjects or formulations. This finding can be explained by the short-term course of the treatment and imaging. Long-term studies are needed, as keratinocyte differentiation takes up to four weeks [
41]. The PSG guidelines recommend using endpoints such as changes in inflammatory lesion count at week 12. Despite a shorter study, we were able to detect subclinical effects within the skin.
Furthermore, in this manuscript, to further explore the potential differences in the penetration of the dermal and pharmacokinetics of adapalene, we adapted our previously published dermal PBPK model to develop predictive in silico IVPT models for the three formulations. The original PBPK framework has been validated in multiple drugs and formulations, demonstrating the applicability of the model. However, one key challenge encountered during the model development was the lack of detailed formulation-specific information, which occasionally led to non-unique sets of model parameters. To address this limitation, we incorporated IVRT data alongside IVPT data to support and constrain calibration during model development. IVRT data were specifically used to calibrate the in silico release model, serving as a surrogate to quantify formulation-dependent parameters that could not be reliably estimated through empirical relationships alone, due to incomplete knowledge of the composition of the formulation.
The calibration, using IVRT experimental data, was performed thorough iterative manual fitting of the model predictions to the experimental IVRT release profiles. The calibration of the partition and permeability coefficients, governing the transport between the discrete and continuous phases, was found to be insufficient to capture the delay observed in the release behavior of some of the formulations. To account for this, an artificial delay term was explicitly introduced. Additionally, it is worth noting that the use of IVRT data to inform and constrain the release model parameters limited the model overfitting during comparison of the in silico IVPT model with corresponding experimental data. Future research in this area could expand upon the current study by incorporating additional factors that may impact drug release, such as co-solvent effects, excipient effects, and the impact of other formulation parameters. For a more robust calibration of the model parameters, detailed information on the exact composition of the formulations would significantly improve the initial parameter estimates derived from empirical relations [
28]. Following this, the IVPT experimental data were used to fine-tune skin-related model parameters. As hypothesized above, the use of IVRT data resulted in minimal calibration of the skin-related model parameters. Specifically, the calibrated parameters were limited to the partition coefficient between the continuous phase of the formulation and the SC lipids, and the partition coefficients involving dermis compartment. This was also supported by the observations from the sensitivity studies conducted on the model (see
Appendix D). Thus, by sequentially calibrating the different model parameters i.e., using IVRT data as the first step, followed by model calibration using IVPT data, the approach enabled systematic separation of formulation-dependent and skin-related parameter calibration, allowing for a minimal and more targeted calibration in the IVPT model. Such separation might reduce the parameter space during calibration, improves reproducibility, and ensures that differences in model output are primarily driven by formulation-specific attributes. By maintaining a uniform set of parameters and adopting a sequential separation approach, the model allowed a direct and meaningful comparison between the different formulations (without a need for individual calibration), highlighting their relative performance and efficacy under identical conditions. It is important to note that this approach assumed limited variability across the skin specimens used in the IVPT experiments, such that the skin-related physiological parameters could be reasonably treated as constant across formulations. While some inter-sample variability is expected, this assumption allows for clearer attribution of differences in permeation profiles to formulation-specific factors rather than biological variation. This not only saves time and resources but also underscores the model’s capability to generalize its predictions, making it a valuable tool for preliminary screening of various formulations, before conducting more extensive in vitro or in vivo tests [
19]. The uniform parameter set can also facilitate easier replication of the study by other researchers, contributing to the reproducibility and transparency of the research. Overall, this methodological consistency strengthens the validity of the findings and supports the use of the model as a reliable predictor of formulation behavior in IVPT studies.
We also acknowledge that there are some limitations with the PBPK modeling effort that require further consideration. Firstly, the model did not account for the rheology of the formulations, mainly the viscosity, which can play a major role in drug diffusivity into the SC. Although a global sensitivity analysis using the Morris screening method was performed, additional efforts focused on uncertainty quantification could improve the robustness of the model predictions and parameter confidence. Furthermore, the model assumed the same skin physiology and did not account for individual differences or the different skin properties present at different anatomical sites. These omissions could limit the generalizability of the model in broader clinical or regulatory settings. The model also needs to be optimized and externally validated with an independently observed dataset. This would be useful to validate the robustness and extend the use of the model. Despite these limitations, the model required only minimal calibration of skin-related parameters, indicating that the underlying skin model is mechanistically sound and can potentially be adapted for other drugs with relative ease. The modeling strategy employed is broadly consistent with the white paper on the qualification of the PBPK platform [
42], particularly in its structured stepwise calibration using IVRT and IVPT data to inform the formulation and skin-specific parameters. Although some predictive performance metrics showed modest fits, these results reflect the inherent biological variability and complexity in topical drug delivery and pharmacokinetics, a challenge well recognized in the field. Importantly, the model nonetheless captured essential trends and formulation-dependent differences, highlighting its value as a foundational tool that can be refined further to enhance quantitative understanding and predictive capability for dermal drug delivery.
Observations and the PD model suggested that AcneFree may produce a faster and more pronounced reduction in the infundibular area than Differin and Effaclar, especially at earlier time points (e.g., 4 h). Although this trend was evident in the model predictions, the EBEs did not indicate statistically significant differences among the formulations. The small sample size and sparse data probably reduced the power of the covariate modeling, which in turn contributed to high parameter variability and an unreliable model structure. Therefore, we relied on a simplified approach using EBEs to explore formulation-level effects. These findings highlight the need for future studies with larger sample sizes and richer data, to robustly explore formulation-specific differences in PD response. Upon evaluating the pharmacodynamic model for changes in SC thickness, it was observed that the Emax and EC50 values for Differin gel were higher than those for AcneFree and Effaclar. Both Emax and EC50 are considered intrinsic properties of the active drug, adapalene, and are typically expected to remain consistent across formulations. However, the observed differences may suggest that formulation-specific excipients or inactive ingredients in Differin gel exert additive or synergistic effects that enhance adapalene’s pharmacological response.
Empirical Bayes estimates (EBE), although derived from the model and not fully independent, supported this observation by indicating significantly higher Emax and EC50 for the Differin gel. Furthermore, the need to achieve a higher maximum effect (Emax) can inherently increase the concentration required to reach 50% of that effect, resulting in a higher EC50. It is possible that Differin gel is formulated to release adapalene more rapidly or in greater amounts to facilitate this response. In support of this, IVRT and IVPT studies have shown greater release and permeation of adapalene from Differin gel, further strengthening this hypothesis. However, we acknowledge that to explore this hypothesis, covariate modeling is needed to show the significance of the formulation. Further studies with placebo control, rich sampling, and a larger population size are needed to assess the significance of covariates. The methodology presented in this manuscript, employing PBPK modeling in combination with pharmacodynamic modeling, has the potential to support early-stage product development, particularly for generic formulations. Although the approach requires further refinement and validation, it offers a promising alternative to reduce the burden on traditional clinical endpoint studies by cutting time and costs.
In this study, we explored a relatively underutilized approach: the application of computational modeling in the development of topical and transdermal products. Owing to the limited availability of data specific to the topical route of administration, and the high inter- and intra-individual variability in skin properties, such models remain largely exploratory and have yet to be fully integrated into regulatory or routine development frameworks. The potential regulatory applicability of such models can be exemplified by an FDA study in which a dermal PBPK model was employed to support the demonstration of bioequivalence and subsequent approval of a generic diclofenac sodium topical gel [
43].
Accurate determination of local drug concentrations within the skin often requires invasive and uncomfortable in vivo techniques such as skin biopsies, tape stripping, or dermal microperfusion. These procedures can be particularly challenging to implement in clinical trials, potentially limiting participant enrollment. As a result, in vitro studies are frequently used to estimate dermal pharmacokinetics. However, these in vitro findings do not always correlate directly with in vivo outcomes, thereby limiting the ability to establish a robust concentration–effect relationship.
For example, in vitro permeation testing (IVPT) does not account for systemic perfusion from the dermis. Drug uptake into the systemic circulation may influence observed flux profiles, particularly in the deeper layers of the skin. Although higher delivery of adapalene was observed with Differin gel, the impact of systemic clearance was not assessed in our study. While Differin may deliver greater amounts of adapalene to the dermis, the fraction that is rapidly taken up by the systemic circulation remains unclear and warrants further investigation. Understanding this aspect could provide additional insight into the clinical effects observed with Differin.
Adapalene is primarily excreted via the biliary route, with approximately 75% of the drug remaining unmetabolized [
14], and it exhibits a short half-life, ranging from 7 to 51 h (average: 17.2 h) [
44]. It is therefore plausible that, despite enhanced delivery to the skin, a significant portion of the drug reaching the dermis is promptly cleared systemically. This hypothesis remains to be confirmed and represents an important direction for future research. Addressing these limitations in subsequent studies would enhance the robustness, accuracy, and translational value of computational models, thereby strengthening their utility in the development of dermal formulations.
Dermal PBPK modeling presents a valuable solution to this challenge. In this manuscript, we adapted a previously validated dermal PBPK model [
18] to estimate adapalene concentrations in both the SC and the infundibular area. While further refinement of the model is warranted, the current version provided predictions that aligned well with experimental trends for the three formulations. This modeling framework can form the foundation for real-world applications and may be further optimized for the development of generic adapalene gels. Ultimately, this computational approach has the potential to streamline product development, reduce dependency on clinical endpoint studies, and lower overall development costs, making it a valuable tool in the evolution of regulatory science for topical generics.