Abstract
Pharmaceutical controlled-release formulations are systems developed by a set of unit operations to achieve a satisfactory combination between a drug and excipients to allow its gradual release. These devices must simultaneously meet criteria for stability, biocompatibility, safety, efficacy, scalability at industrial volumes, and technological efficiency for drug release. Controlled-release systems (CRSs) must release drugs in a way that maintains an adequate concentration in the organism, a requirement that is challenging to meet in practice. Even though novel CRSs may be designed with new materials as excipients, new drugs, or emerging manufacturing technologies, the mechanisms for drug release continue to be governed by a set of similar physicochemical phenomena such as diffusion, swelling, or erosion. These phenomena are too complex to be analyzed by numerical methods; however, they are relatively accessible by probabilistic models especially the Monte Carlo simulation. In this review, we discuss key findings related to the use of this probabilistic method for analyzing the drug-controlled release process in different pharmaceutical devices. Based on this evidence, we propose their potential application in the characterization of new drug-controlled release systems, synergy with other computational methods, and their capability to be adapted for in vivo or in vitro kinetic analysis.
Keywords:
Monte Carlo simulation; Diffusion; Drug-controlled release; Percolation threshold; Nanotechnology; Simulation; Swelling
INTRODUCTION
Pharmaceutical dosage forms are the result of a combination of excipients and drug(s) that are technologically efficient in facilitating administration in humans and animals. In the development stage, an optimal formulation is designed using trial-and-error methods; its final composition is limited by the number of tests or the expertise of the formulation specialist. These empirical formulations can be time-consuming and expensive and may have an impact on Critical Quality Attributes (CQAs) such as potency, release kinetics, process-related impurities, etc. To minimize this impact, before scaling up pharmaceutical manufacturing for a selected formulation, several assessment strategies can be used to estimate and reduce the risk for a specific formulation. For example, a Risk Assessment Analysis based on the ICH scientific guideline Q9 on Quality Risk Management could be employed to determine the failure risk concerning the critical attributes of quality in a marketed product (Coleman, 2023). Another useful approach is development based on Quality by Design (QbD), where development is initiated with predefined objectives for the relationship of the product quality to the desired clinical performance. Essentially, it emphasizes understanding of the product and process based on scientific principles, and quality risk management to decrease the failure risk during the design of the product (Davanço, Campos, de Oliveira Carvalho, 2020; Pramod et al., 2016) (Figure 1).
Overview of the QbDs development cycle. Quality Target Product Profile (QTPP), Critical Quality Attributes (CQAs), Critical Process Parameters (CPPs), Critical Material Attributes (CMAs).
To implement this strategy, the first step is to establish a Quality Target Product Profile (QTPP) which shows the requirements of the patients and to translate it into design features in the pharmaceutical formulation. For example, a formulation may require a specific drug release profile, long-term stability, or a particular pharmaceutical dosage form, etc. In this step, between the theoretical support and pre-formulation stages, in-silico methods are useful to propose pre-formulation conditions and to simultaneously reduce the design space and the number of tests required achieve an optimal formulation (Yu et al., 2014). Other tools such as Design of Experiments (Politis et al., 2017), in vitro-in vivo correlation (Huang, Goolcharran, Ghosh, 2011; Kaur et al., 2015; Peraman, Bhadraya, Padmanabha Reddy, 2015; Yekpe et al., 2018), 3D-printing technology (Tracy et al., 2023), photolithography and computer-aided design (Andreadis et al., 2022; Geraili, Xing, Mequanint, 2021), the Biopharmaceutical Classification System (Davit et al., 2016) and modeling by simulation constitute novel strategies for generating an optimal CRS. Moreover, these trends have been integrated as part of regulatory affairs dossiers used to support the reproducibility of therapeutic benefits. In this context, a new line of research is emerging that utilizes computational simulation to anticipate the design of new formulations, with the aim of maximizing compliance with specifications and avoiding negative impacts on CQAs. For example, simulation models have opened possibilities to explore new relationships in silico-in vivo, which is highly valuable for the prediction of an in-vivo drug delivery profile; to establish dissolution specifications for finished products; or to support long-term stability studies (Al-Tabakha, Alomar, 2020; Casalini, 2021; Kambayashi, 2023). To achieve all of these objectives, new challenges in computational simulation have emerged, especially in the interpretation and transference of physicochemical properties, biological properties, or descriptions of hydrodynamic phenomena to computational algorithms (Adepu, Ramakrishna, 2021). In this review, we identify some key findings generated by probabilistic simulation models used to analyze drug controlled release from different systems. Based on these reports, we propose their potential application in the characterization of new drug controlled release systems, their synergy with other computational methods, and their capability to be adapted for in vivo or in vitro kinetic analysis.
Controlled Release Systems
Controlled release systems (CRSs) are formulations designed to release active pharmaceutical ingredients of different molecular sizes, from small molecules to peptides or nucleic acids, at a constant rate over long periods of time. CRSs also allow dose frequency to be reduced, increase adherence to treatments and reduce adverse effects (Park, 2014). These formulations continue to be valid and are highly desirable for new developments; in fact, a large number of formulation proposals, including diffusion-controlled CRSs, are in advanced clinical phases. Several CRSs have been developed for specific treatments, including asthma, arthritis, Alzheimer’s disease, duodenal ulcer, cancer, cardiovascular diseases, diabetes, systemic arterial hypertension, and hypercholesterolemia, among others (Gao et al., 2023). Some of these CRSs show drug release patterns described by zero-or first-order kinetics, or a combination of the two, such as burst or sustained release (Adepu, Ramakrishna, 2021).
Diffusion as key mechanism involved in the drug-controlled release
Diffusion is the main mass transfer phenomenon associated with diffusion-controlled CRSs but its description in physical terms and mathematical expressions is complex. Diffusion phenomena can be analyzed by Fick’s law and expressed by the equation (1):
Where J is the rate of transfer per sectional unit area (cm 2 / s); ∂c is the change in concentration of the diffusing species (g/cm 3 ); D denotes the diffusion coefficient (also called diffusivity, (cm 2 /s) and ∂c is the change in distance (cm). Fick’s second law of diffusion can be derived from Fick’s first law considering mass balance. For example, for three dimensions and different diffusion coefficients that vary with position, time, and/or solute concentration, the relationship generalizes to the following equation (2):
Where c is the concentration of the diffusing species (g/ cm 3 ), D is the diffusion coefficient (cm 2 s -1 ) and x,y, z are the three spatial coordinates. The resolution of Fick’s equations should be adapted for each case, considering the geometry of the device, the size of the system, dimensionality, etc., and implying the establishment of specific conditions denoted as “initial and boundary conditions”. For example, for a cylinder, which could be described as the classical geometry for a pharmaceutical device, Fick’s second law of diffusion can be solved considering these initial and boundary conditions, leading to the following expression for the drug released at time t,
where R and H denote the radius and height of the cylinder, n and p are summation indices, and q n is the n-th root of the zero-order Bessel function of the first kind [J 0 (q n )=0]. On the one hand, the “initial conditions” concern the initial movement of particles inside a system with a particular solution when c(x,t)=0. In fact, equation (1) makes sense under initial and boundary conditions, limiting their solving to analytical solutions whose interpretation is only limited to the initial and boundary conditions. On the other hand, “boundary conditions” in non-homogeneous systems refer to constraints imposed by the system, such as critical changes of the physicochemical properties in the excipients or drugs as a transition phase or relaxation of polymers whose mathematical interpretation can be described by discontinuities that are difficult to analyze (Siepmann et al., 2012). One of the restrictions of these determinist models is that they describe the movement of drug particles along stationary boundaries, and thus do not explain completely the kinetic behavior in dynamic conditions such as swelling and erosion, among others.
One of the alternatives that make statistical physics and probabilistic theory available for the comprehensive study of the movement of drug particles during dynamic and simultaneous events is modeling using stochastic methods such as the Monte Carlo simulation and cellular automata. We conclude this section by showing that a deterministic solution for the diffusion phenomenon is not practical for analysis of the kinetics of drug release from CRSs, and therefore that alternative approaches could facilitate or aid their analysis.
What is the Monte Carlo simulation?
Monte Carlo simulation comprises a broad class of computational procedures that use randomly generated numbers to describe phenomena of interest and provide a statistical approximation when the exact values are unavailable or the solution of the problem is complex (Figure 2).
(Left side) Scheme of diffusion-controlled drug delivery systems with the drug homogeneously dispersed in a polymeric, swellable, and hydrophilic excipient. As the swelling process progresses, the gel layer gradually becomes thicker, resulting in progressively slower drug release rates. However, due to continued hydration, the polymer disentangles from the matrix surface, resulting in a gradual dissolution decreasing the thickness of the gel layer and increased dissolution rate (Right side). The figure shows an example of a random walk in a 2D network passing through a boundary. Once it crosses the boundary, the particle is counted as a released particle. The sites visited by the walker are marked in red. The walker can be water particles (blue) or hydrated drug particles (red).
The Monte Carlo method is founded on the principles of statistical physics, a relatively unexplored field among formulation scientists; consequently, the applications of these concepts are sometimes limited. In statistical mechanics, through the partition function Z, we can determinate all the thermodynamic properties of a system with many degrees of freedom (Landau, Binder, 2021; Reichl, 2016). In particular, one application of the Monte Carlo simulation has allowed simulation of the motion of a set of particles using the random walker concept (Du et al., 2022; Weiss, 1983; Wu et al., 2022). The classical problem of the random walker, which is one of the most studied problems in the statistical mechanics of nonequilibrium systems, postulates that the walker will take an average of a certain number of steps in one direction or another, with each step being statistically independent of the others (Figure 3).
A. A Monte Carlo step: the figure show that each attempt of displacing, whether accepted or not, the time increases by a value equal to 1/Nt, where Nt is the number of drug particles remaining inside the matrix at time t and when Nt particles have been chosen, it is considered an arbitrary time unit called a Monte Carlo Step. Occasionally, when the random walker encounters barriers or obstacles on the path, new rules or conditions are implemented, for example, when the random walker is close to an obstacle and can only take one step to the right, one step to the left, or one step backwards but not to the forward. Physical barriers can be simulated considering the properties of the materials, such as the geometry of the system, the presence/absence of coatings, porosity, or inclusive environmental conditions. B. Different pharmaceutical systems can be simulated using the random walker model.
The random walker randomly chooses one of the different options to carry out a movement to a neighboring site; if the requirements are satisfied, the walker migrates to this new position, but if the rules for movement are not satisfied, the walker remains static in the same place. With each attempted displacement, whether accepted or not, the time increases by a value equal to 1/Nt, where Nt is the number of drug particles remaining inside the matrix. The time t when Nt particles have been assessed is considered an arbitrary time unit called a Monte Carlo Step (MCS) (Figure 3) (Gomes Filho, Oliveira, Barbosa, 2016).
Depending on the trajectory generated by the random walker, it is possible to determine the complete route and the final location of each particle, for instance, when a particle is either inside or outside of a pharmaceutical device (Feller, 1991; Hughes, 1995; Weiss, 1983 ). Formally, a random walker is a Markov chain with independent additive increments (Fishman, 2013). In a series of random variables X n , a random walk satisfies: where ϵ 1 , ϵ 2 ,.. are a sequence of identically distributed random variables and are generated independently of X n (Kroese, Taimre, Botev, 2013; Robert, Casella, Casella 1999). However, while the random walker model maintains a microscopic approach, the results obtained can also be seen and are measurable at the macroscopic level. For example, the movement of a molecule that diffuses in a porous solid can be explained at the macroscopic level as the amount of drug released. The particles can be used to refer to molecules, nanoparticles, granules, or aerosols, etc., and are unitary entities moving freely in a specific direction for a distance roughly equal to their mean free path. To ensure the success of the implementation of a Monte Carlo simulation algorithm, several factors must be considered, especially the use of an appropriate sampling technique, the incorporation of a pseudo-random number generator, and the generation of an ensemble of configurations with statistical value, all of which must be able to describe the phenomenon of interest (Sawilowsky, 2003). Monte Carlo simulations calculate the number and position of particles as a function of time. If the position of the particle is outside of the device and is labeled as “released” it is added to an accumulative counter. Analogously, another types of particle that moves also can be tracked during all simulations, for example, excipients. Finally, these simulation data can be correlate with the drug amount, volume, erosion percentage, gel particles generated during a phase transition, number of internal particles, hydrated particles, water amount, etc. Afterwards, it is possible to build different correlations between each simulation data and time to generate empirical or semi-empirical models that describe the kinetic patterns (Bruschi, 2015). For example, using Monte Carlo Simulation, it is possible to compare hypothetical conditions and predict their impact on critical quality attributes, such as the drug/excipient ratio needed for appropriate gel formation, which is critical for a drug-controlled release; or the calculation of the percolation threshold required to allow the total release of the drug load from a pharmaceutical device. In conclusion, to develop a computational model, it is necessary to know the drug-release mechanisms of the pharmaceutical dosage form to translate them adequately to a probabilistic simulation algorithm.
Requirements to develop a simulation model using the Monte Carlo method
To simulate a drug delivery process, it is necessary to establish the hydrodynamics implicit in the release of drug particles into the release medium or biological environment. These models should be in agreement with their classification, especially with the limiting steps that condition the drug dissolution process. (Fu, Kao, 2010; Langer, 1990).
According to the classical works by Peppas (Peppas, Franson, 1983) and Robert Langer (1990), different pharmaceutical devices can be divided into classes depending on the set of mechanisms for drug release, including diffusion-controlled, chemically controlled, solvent-activated and magnetically controlled delivery systems. Particularly, the Monte Carlo simulation method can be easily adapted to diffusion-controlled systems including novel pharmaceutical devices of nanometric size (Figure 4).
Diffusion-based controlled release systems. Some representative nanosystems available to be modeled include liposomes, dendrimers, hydrogels, nanogels, carbon nanotubes, and gold nanoparticles.
However, while this classification continues to be valid, with the emergence of new materials, new chemical classes of drugs, and new manufacturing processes, it is necessary to include additional criteria for a robust classification (Han et al., 2010; Langer, 1990; Mandal et al., 2010; Peppas, Franson, 1983). For example, in innovative pharmaceutical formulations whose release is based on erosion, pulsative systems, rupturable membranes, or hydrolysis-mediated systems, the classical diffusioncontrolled mechanism does not completely explain the kinetics of drug release (Beugeling et al., 2018; Mandal et al., 2010). In this frame, the first challenge for drug release modeling from CRSs should be to understand all, or at least the main mechanisms for drug release implicit in each class of CRS that are not yet fully characterized (Mandal et al., 2010). For example, experiments carried out in polymer-based nanosystems have shown changes in the diffusion coefficients due to modifications in the polymer biodegradation process that occurs in parallel to the drug release indicating as additional requirement the chemical reactivity or stability of the excipients (Macha et al., 2019). Interestingly, other mechanisms, such as the staggered profile, which is regulated by the biochronology of each patient, to allow regulating the discharge of drugs in a pulsatile profile could be analyzed by an in silico approach to explain the effect of the pulsatile discharge of drugs under different biological conditions (Mandal et al., 2010).
Monte Carlo simulations in fractal and Euclidean systems
In physical formalism, the drug-controlled release process could be described as the escape of particles from a system of fractal geometry, i.e. a system characterized by irregular spaces. In contrast to a Euclidean space in which all the systems are homogeneous and conserve a single value for the diffusivity of their particles, a fractal system is a disordered medium in which the diffusivity is dynamic. This problem was initially raised by Bunde et al., in 1985, who concluded that there is an empirical relationship between release rate and time. This relationship is known as the Power law or KorsmeyerPeppas Equation:
where, Mt and M∞ are the amounts of drug released at times t and infinite, respectively. k is a parameter determined under in vitro or in vivo conditions (O’Farrell et al., 2022) and n is an exponent that depends on the geometry of the boundary of the system, which can have an interpretative sense for the drug release mechanisms (Kosmidis, Argyrakis, Macheras, 2003a). Following Bunde’s work, several reports have developed this theory and improved the study of Fickian diffusion in fractal and Euclidian spaces (Kosmidis, Argyrakis, Macheras, 2003a, b ). In both cases, it was also found that the Weibull function appropriately describes the complete drug release curve when the drug release mechanism is Fickian diffusion. The Weibull model is represented by Equation 6:
where is a scale parameter, and β is the curve shape factor. In the case of release from Euclidian matrices (Kosmidis, Argyrakis, Macheras, 2003a) the value of the exponent b was found to be in the range 0.69-0.75. Experimental approaches have used this mathematical relationship to understand when a system obeys Fick’s first law (Corsaro et al., 2021; Kobryń et al., 2017; Martín-Camacho et al., 2023; Ranjan, Jha, 2021; Yu et al., 2014). Kosmidis and colleagues (2003b) were the pioneers of the introduction of Monte Carlo kinetic simulations using the concept of a random walker to calculate the diffusion coefficient in inert tortuous systems (Kosmidis, Dassios, 2019; Kosmidis, Macheras, 2007; Papadopoulou et al., 2006). In this first publication, two-dimensional networks (d = 2) were analyzed to describe the release behavior from a Euclidean and a fractal matrix and the percolating aggregate at the percolation threshold in square lattices (Kosmidis, Argyrakis, Macheras, 2003b). In the case of release from the two-dimensional percolation fractal with fractal dimension d f = 91/48 the values of b ranged from 0.35 to 0.39 (Kosmidis, Argyrakis, Macheras, 2003b). In the same way, when they studied the fractal matrix, they found that the release profile of the drug is adjusted to the Weibull equation.
On the other hand, in Euclidean matrices, the validity of the Higuchi equation for a one-dimensional matrix system has been demonstrated by Monte Carlo simulation. Moreover, Monte Carlo simulations have been applied to analyze the effect on a controlled release profile when a drug is arranged in random mixtures with areas of high and low diffusivity. This evokes matrices with a coating-like structure that are identified as following a Weibull distribution, suggesting that thin-layer enteric-coated systems could modify the release profile resulting in diffusion coefficients three orders of magnitude lower than the drug release at a constant rate and without coating-like properties (Kosmidis, Macheras, 2007). These Monte Carlo simulation results apparently point to a generalization between the Weibull model and the system geometry (Kosmidis, Argyrakis, Macheras, 2003a). Villalobos and colleagues have studied inert spherical matrices using the Monte Carlo methods and the implication of the percolation phenomenon exerted by the basic components of this type of matrix (drug and excipient) on drug release and showing that the fractal dimension value is useful to estimate the complexity of porous microstructures and to define when a specific medium is homogenous or heterogeneous (Villalobos et al., 2017). . Finally, these contributions conclude that fractal and Euclidean matrices can be used as models for drug controlled released and they can serve as a reference point for the creation of more complex models.
Monte Carlo simulations in new drugs, excipients, and dissolution mediums
The mechanisms of drug-controlled release are intimately associated with the properties of the drugs used, such as their hydrophobicity, electric charge, molecular size, isoelectric point, etc. Particularly, in biotechnological products such as proteins, DNA, mRNA, and siRNA these properties demand consideration (Jiang, Abedi, Shi, 2021; Park, Otte, Park, 2022; Poornima et al., 2022) including their retention properties on substrates or polymeric materials (De Izarra et al., 2021; Senapati et al., 2021). Eventually, a diffusion/erosion mechanism for the drug or polymers is required to release these particles at a constant rate. For example, in substrate-mediated delivery the DNA is immobilized to a material that functions to support cell adhesion and migration and places DNA directly in the cellular microenvironment. In this type of system, molecular interactions play a key role because charge could bind other components to the delivery vehicle. Furthermore, delivery from most polymeric devices likely occurs through a combination between binding and release mechanisms, and both the vector and the polymer can be adapted to regulate these interactions (Pannier, Shea, 2004). From this perspective, a significant number of properties of macromolecules, polymeric materials and their interactions with their biological targets can be modeled. Additionally, the release profile of a drug may be affected by various proteins, cell types, and in vivo enzymes; thereby, they also become an attractive system for probabilistic modeling (Seo, Mittal, 2021).
In vitro experiments use biorelevant dissolution media to replicate the impact of biological fluids on drug-controlled release in various pharmaceutical devices. Some of them have been characterized according to their physicochemical properties, representing a potential niche for developing simulation models and assessing the effect of the dissolution medium on the drug-controlled release kinetics. Some examples of biorelevant dissolution media are FaSSIF (fasted-state simulated intestinal fluid), FeSSIF (fed-state simulated intestinal fluid), FaSSGF (fasted-state simulated gastric fluid) and FeSSGF (fed-state simulated gastric fluid), etc.(Lemos, Prado, Rocha, 2022). It is likely that phenomena such as hydrolytic activity, pH, temperature, viscosity, and enzymatic activity are excellent physicochemical parameters accessible for an assessment by simulation approaches (Fernandez-Lopez et al., 2023; Gao et al., 2019; Liang et al., 2022; Martín-Camacho et al., 2023; Russo et al., 2022; Vernon-Carter et al., 2022). Furthermore, another line of applications for Monte Carlo simulations could be related to create in vitro conditions to establish the experimental conditions for a dissolution test. For example, simulations based on the Finite Element Method through the software COMSOL Multiphysics® have shown the impact of the perfect sink assumption and assess the effect depending on stirring speed, drug loading, and erosion and swelling properties (Ranjan, Jha, 2021).
With this approximation, it is possible to identify the initial parameters for the development of dissolution analytical methods and predict dissolution profiles and to study how these are affected by the stirring speed and volume of dissolution. However, this deterministic simulation method is distant from stochastic procedures, it could be used as a reference to validate theoretical data generated by Monte Carlo simulation.
We conclude this section by demonstrating the potential of probabilistic simulation methods to analyze the effect of biorelevant dissolution media on the drug-controlled release and in the construction of dissolution analytical methods and to evaluate new biological drugs or excipients using specific physicochemical parameters which directly affects drug-controlled release kinetics.
Monte Carlo simulations in polymeric and inert systems
CRSs designed with polymeric matrices have gained popularity for establishing simulation models because they allow a study systematic for drug release (Colombo et al., 1995). The physicochemical events that take place during the drug release initiate when a swellable polymeric CRS contacts water, forming a hydration front. As the hydration front advances, the particles begin to move (Vaitukaitis et al., 2020). Simultaneously, water interacts with the polymer promoting swelling. However, this class of polymers shows different swelling patterns; most of them are similar in layer formation, but their thickness or formation dynamic is variable. When a tablet undergoes swelling and water permeates into its core, various fronts are created. Several reports have identified three distinct zones: the diffusion front, the erosion front and the swelling front which have been characterized by highly sensitive equipment such as IR, NIR, MNR, etc. (Avalle et al., 2013; Yassin et al., 2015). The increased resolution of such analytical equipment presents new benefits in the field of pharmaceutical technology since systems with heightened sensitivity improve the quality of process description, facilitating a more precise comparison among various pharmaceutical formulations that exhibit subtle variations. Simultaneously, these systems enhance the potential for generating data that can be effectively utilized to develop computational models (Manaia et al., 2017). A recent study demonstrated that a gel formulation can produce a novel irregular region characterized by micropores and microbubbles. This region, with a thickness of 300 µm, is likely formed by internal structural rearrangements within the gel, which are induced by the dissolution of acid compounds and modifications in the pH microenvironment. This is an anomalous region which is very attractive for simulation by probabilistic methods (Juszczyk et al., 2021). In the same report, the presence of intricate spatiotemporal hydration patterns generated by swellable polymer-based matrices such as sodium alginate is highlighted. In this experiment, both the composition and the molecular properties of the constituents play a significant role in the observed behavior (Juszczyk et al., 2021). However, the diffusion mechanism is seen as the main phenomenon that orchestrates the movement of dissolved drug particles through the polymer network. There is also evidence that other zones could be formed during the swelling of tablets, suggesting new control mechanisms for drug release. This example illustrates a distinctive hydrodynamic phenomenon that emerges as a result of the implementation of new polymeric materials. To date, we know that the mechanisms that affect the release of a drug from a polymeric matrix involve diffusion, water penetration, hydration, solvation, gel formation, erosion, swelling front formation, and the formation of new zones. Furthermore, some studies have shown that also the design properties of CRSs, such as their shape and size, have an evident impact on the drug release kinetics (Kulinowski et al., 2014, 2016). Sanjeev Garg and colleagues employed predictive quantitative structure-property relationship (QSPR) models to identify an optimal diffusion-controlled drug delivery system using Paclitaxel as a model drug. Garg’s team also concluded that the type of polymer, the diffusivities, the initial loading, and the number of nanoparticles, as well as the shape and size of the system play a crucial role in regulating the effectiveness of the drug for different dosage regimens (Pramanik, Garg, 2019).
Finally, the calculation of the percolation threshold value in inert systems is another intriguing use of the Monte Carlo simulation in pharmaceutical development. This property indicates the concentration at which the polymer or drug can form an infinite cluster and percolative. Above the excipient percolation threshold value, a continuous cluster of this component is produced and is capable of regulating the hydration and release rate (Caraballo, 2009). Below this threshold value, the excipient does not regulate the release of the drug by directly affecting its release. This critical value can be predictable and analyzed by Monte Carlo Simulation (Adamczyk, Polanowski, Sikorski, 2009; Pawłowska, Sikorski, 2013; Polanowski, Sikorski, 2017) and is an indicator of the dose amount trapped at infinite time considering the initial composition of the matrix (Martínez et al., 2009; Villalobos et al., 2017). Examples concerning percolation threshold values are shown in Table I. Nonetheless, in polymeric drug controlled release devices this concentration should be assessed with caution. For example, in swellable CRSs, the drug-release is regulated by a polymer network that forms a gel layer, which alters the release kinetics. Consequently, the actual percolation threshold values can differ from the values observed in an inert and porous system. Recent experiments using fluorescent microscopy have demonstrated the feasibility of observing the gel layer formation during the initial stage of polymer hydration and revealing a potential connection between inert and swellable systems through of the concept of percolation threshold (Mason et al., 2015).
Some Percolation Threshold values determined for some excipients and drugs in different pharmaceuticals devices.
Monte Carlo simulations in hydrogels and nanogels
Hydrogels are novel pharmaceutical devices that have exceptional swellable and water-uptake properties because they are composed of a polymeric network with excellent mechanical strength (Hu et al., 2022; Romischke et al., 2022). Other properties, such as biodegradability, sensitivity to stimulus, and the ability to absorb fluids, have generated a new field for the study of these new formulations. Some properties involved in the release of drugs, such as their permeability, diffusion, and erosion characteristics make hydrogels excellent systems for controlled drug delivery and for their modeling by probabilistic simulation (Liao, Hou, Huang, 2022). For example, the diffusion of drugs in hydrogels is generally modeled by one of the following three theoretical approaches: (a) hydrodynamic theory, which considers interactions between the drug and the surrounding hydrogel matrix; (b) free volume theory, which assumes that the solute is transported via dynamic empty spaces between molecules without effects caused by interaction; and (c) obstruction theory, which models the polymer net as a barrier for the diffusion of the drug within the liquid (Turnbull, Cohen 1970; Cukier, 1984; Mackie, Meares, 1955). Recently, an analysis of diffusion in hydrogels by positron annihilation lifetime spectroscopy (PALS), a technique capable of measuring the molecular pores in biomaterials under wet conditions, showed the relevance of interactions between the hydrogel and the drug (Axpe et al., 2019). In addition to the study of diffusive phenomena, hydrogels have also been used as excellent models to analyze the hydrodynamics of gel expansion or water-uptake properties (Benkő et al., 2022; Jagusiak et al., 2020; Kashkool, Soltani, Souri, 2020; Kocaaga, Guner, Kurkcuoglu, 2022; Pérez-Mas et al., 2018). For example, Perez-Mas and colleagues (2018) analyzed molecular properties in neutral hydrogels by means of coarse-grained grand canonical Monte Carlo Simulations. In this work, it was possible to establish theoretical conditions in which the materials could maximize the adsorption of solute. Recent studies showed that the size of cosolutes, together with the magnitude of hydrophobic attractions and of steric repulsion play a key role in improving the absorption of cosolutes in neutral hydrogels (Pérez-Mas et al., 2018). However, hydrogels continue to be studied, and several theoretical models can be used to understand their mechanical movements at the nanometric level (Ahualli et al., 2017; Quesada-Pérez, Martín-Molina, 2013). Recently, the same research group reported the effect of different molecular parameters of hydrogels on solute diffusion using coarse-grained simulations, concluding that chain flexibility inside the polymeric network is relevant for the movement of particles (Quesada-Pérez et al., 2022). Additionally, Quesada-Perez and Martin-Molina published a historical overview that showed the progress made in coarse-grain simulations of solute diffusion in hydrogels and nanogels. These results have been crucial for understanding how drug-controlled release is carried out in this type of pharmaceutical device (Quesada-Pérez, Martín-Molina, 2021). Finally, Monte Carlo simulations are establishing a synergy with other tools such as network analysis and artificial intelligence with the aim of enhancing the predictability of outcomes generated by pharmaceutical developments. Unsupervised methodologies possess the ability to discern patterns; nevertheless, their limitation is in the necessity of having enough data to correctly describe each event. It is feasible to employ this probabilistic method to generate data that can be exploited by these approaches (Bannigan et al., 2023; Han et al., 2019; Manolis, Lagaros, 2002; Santana et al., 2020; Sarrut, Krah, 2021).
Another interesting application for Monte Carlo simulations has been the analysis of ideal conditions for transdermal delivery in epidermal membranes of hair follicles, suggesting the successful application of these models in more complex systems (Barry, 2002). For example, currently, DNA immunization can be implemented through topical administration, suggesting follicular transport, so that the hair follicle promises to be a target for gene therapy. This is interesting because several formulations for controlled release could be ideal for alopecia therapy; however, this type of model has not been completely explored by simulation (Fan et al., 1999; Hoffman, 2000; Ryu et al., 2020). Another interesting model that explores the interactions between drugs and human tissue by stochastic procedures is reported by Islam and colleagues. Their report conducted an analysis using a time-adaptive Brownian Dynamics algorithm to investigate the impact of particle size on the dispersion and penetration of drugs within intracellular tissues (Islam, Barua, Barua, 2017).
Monte Carlo simulations in liposomes
Liposomes are spherical vesicles constituted by phospholipids and steroids, usually in the size range that can be used as a model for are analogous to cell plasma membranes (Bozzuto, Molinari, 2015; Sercombe et al., 2015). Here, compatibility with drugs and physical barriers generated by the lipid bilayer are key features when considering liposomes as drug delivery systems. For example, liposomes are effective systems in containing and protecting labile molecules such as RNAs or proteins, but there are still several challenges to improve their stability, loading mechanism, and reproducibility, which limit their use to specific liposome-drug pairs. Moreover, these nanosystems employ a set of physical mechanisms for drug-controlled release, which include Fickian diffusion and erosion of the bilayer (Figure 5).
A. Schematic overview of extracellular matrix and its major components. Although the extracellular matrix varies depending on the tissue, the matrix is mainly composed of a variety of proteins such as collagen, elastin, fibronectin, among others and polysaccharides assembled into an organized meshwork. B. Schematization of the extracellular matrix can be considered as a porous system through which particles with different diffusive properties can transit.
Therefore, these pharmaceutical devices present potential subjects for computational modeling, yet remain unexplored to date (Liu, Bravo, Liu, 2021; Wu et al., 2019). Analyzing a liposome as a model system, we observe that drugs always maintain a probability to diffuse by active transport or diffusion through pores.
If the encapsulated drug or the bilayer is altered in any way, for instance by biodegradation, the load of the liposomes can be accelerated, leading to changes in the drug-controlled release kinetics. Due to their analogy with the cell membrane, liposomes facilitate the incorporation of some drugs, enhancing their bioavailability (Wahab, Mögel, Schiller, 2011). Design of drug-controlled release by liposomes involves new materials, including PEGylated materials based on polyethylene glycol (PEG), surface-anchored ligands like antibodies, carbohydrates, peptides and systems that amalgamate with nanomaterials. A recent report predicted the release of drugs from liposomes into the bloodstream, modeling the drug release and the collective motion of the liposome and its surrounding blood cells with elegance (Kaoui, 2018). In the context of a single liposome and red blood cells within a limited area of the blood vessel, the targeted site can be defined as a specific region within which the flow closely approximates a simple shear flow. This results in the vessel walls becoming rigid and therefore resistant to deformation under hydrodynamic pressures (Kaoui, 2018).
Some mechanisms manipulate electrical or ultrasound field-induced membrane permeation to lead to drug release. Using this mechanism, Nily Dan (2015) analyzed a model for liposomal release induced by low-frequency ultrasound by probabilistic simulation. With this assumption, the release is largely dominated by diffusion through the membrane. Using Monte Carlo simulations, Dan concluded that pore properties in the liposomes, such as distribution, pore size, and relative pore surface, are key to an adequate drug-controlled release profile. Here, a complex combination of different diffusive processes of the drug can be represented using liposomes as model system, but these have not yet been explored by simulation.
Monte Carlo simulations in exosomes
Exosomes are extracellular vesicles that are promising pharmaceutical dosage systems for drug-controlled release. These systems, which are conserved across all kingdoms of life and feature in biological processes such as cell-to-cell communication, maintain biophysical properties that can be modeled and considered as novel biopharmaceutical vehicles (Jimenez-Jimenez et al., 2019a). Moreover, exosomes can be used for different biological targets, including different tissues or species such as parasites, bacteria, and plants, so that, in each new environment, different simulation parameters are needed (Koomullil et al., 2021). Recently, Lenzini and colleagues (2020) showed that at the cellular level, exosomes must traverse the extracellular matrix which is composed of nanopores that are highly selective to particle size. A formulation designed by pharmaceutical criteria remains challenging based on several characteristics, such as efficiency of loading methods, reproducibility, pharmacological targets, and long-term stability (Herrmann, Wood, Fuhrmann, 2021). A strategy based on hydrogels can regulate the transport of EVs through the extracellular matrix. This combined hydrogel - exosome system facilitates motion leading to free diffusion and enhancing water permeation (Lenzini et al., 2020). Interestingly, this type of phenomenon arguably has potential for computational modeling by Monte Carlo simulation.
Simulations by cellular automata
Cellular automata (CA) models are discrete computational models for analyzing dynamic systems in which the elements are in close interaction. Simulation using cellular automata has been used in different fields to resolve issues related to diffusive phenomena (Cao et al., 2023; Fukś, Mudiyanselage, 2022; Kulagin, Shapovalov, 2023; Liang et al., 2023; Ma, Lin, 2022; Medina et al., 2022). This model uses identically programmed units denominated “cells” which are in direct contact with their neighbors and subject to a finite set of prescribed rules for local transitions. One of the main similarities between MCS and CA is their integration within regular spatial lattices. The movement of each cell follows a rule and is assessed in each step. When the system is updated synchronously, only if the state of the cell at the time is satisfied is the transition carried out (El Yacoubi, El Jai, 2002) (Figure 6).
Cell states and transitions in a cellular automaton model is composed as a set of cells forming 3D rectangular lattice. Molecular motion is modeled for each block independently.
Several controlled delivery systems have been modeled by cellular automata and we will describe some of them to exemplify the potential of cellular automata in the field. Another similarity between Monte Carlo simulation and cellular automata is that the diffusion mechanism based on random walking avoids the need to solve Fick’s law for these systems. For instance, Laaksonen and colleagues presented one of the first models for swelling drug-controlled release using a cellular automata approach. In this model, for different defined cell types with water, drug, erodible polymer and wet polymer in each, established transition rules operate when the tablet interacts physically with water molecules. Using the solid drug/saturated drug ratio as a parameter in the model, it is possible to simulate two compounds with different solubilities (Laaksonen,
Hirvonen, Laaksonen, 2009). Recently, García-Fandiño and colleagues showed the impact of a synergistic in silico-in vitro approach, demonstrating the predictive power of computational simulations for vitamin E−sphingomyelin nanosystems (Bouzo et al., 2020). Fathi and colleagues developed a stochastic model using cellular automata to simulate encapsulant release from lipid nanocarriers containing hespertin, which considered diffusion as the predominant mechanism, using MATLAB®. In this simulation, the impact of the load of drug, the type of neighborhood defined, and the solubility, encapsulant distribution, and type of release medium were assessed in two and three dimensions. Here, it was possible to identify concordance between real experimental data for hespertin release and the simulation results (Fathi et al., 2013). Finally, some material biosorbents have been used for the pharmaceutical design of microspheres, and they have been characterized by modeling with a three-dimensional cellular automaton showing a good correlation with experimental data (Bertrand, Leclair, Hildgen, 2007).
CONCLUSION
We have conducted a brief review and discussion on the use of probabilistic methods, such as Monte Carlo and Cellular Automata simulations to describe their impact in the analysis of the kinetic patterns generated during drug-controlled release. We identified some key points:
-
-Systems that can be simulated must maintain a basic set of physicochemical phenomena that orchestrate the drug-controlled release, especially the diffusion of the drug and dissolution medium.
-
- Simulation of drugs mobilized in biological systems such as the extracellular matrix, skin tissue, blood tissue, synovial fluid, etc., can be studied as fractal systems.
-
- Drug properties such as their solubility, molecular weight, coefficients of partition, topology, or molecular interactions can be useful parameters for design of a Monte Carlo simulation algorithm with greater capability to predict the kinetic phenomena associated with the drug-controlled release. Moreover, enzymatic activity and heterogeneous environments can also be simulated.
-
- Nanopharmaceutical devices can be seen as new models for probabilistic simulations and analysis of drug release kinetics under different design conditions.
-
- Dynamic events that occur during drug-controlled release in different pharmaceutical devices, such as swelling, erosion, water diffusion, changes of viscosity, enzymatic action, and the effect of new components in the formulation, may also be modeled or coupled in Monte Carlo simulations. To improve the efficiency and prediction of Monte Carlo simulations, it is necessary to integrate new in silico approaches such as machine learning, artificial intelligence, deterministic methods, or novel computational strategies.
-
- Simulation findings exhibit significant agreement with experimental data, indicating that this approach has he capability to either predict or validate kinetic data for various pharmaceutical devices.
Finally, despite the limited number of reports on these models, we are optimistic about their potential in the early stages of formulation of new pharmaceutical dosages.
ACKNOWLEDGMENTS
We thank to the PhD Homero Gomez Velasco for his generous support and helpful discussions. We also thank to Silvia Lizbeth Reyes Malagón for her valuable support in the design of some figures for this report. This paper presents part of the PhD thesis of Saúl Jiménez-Jiménez, who is a doctoral student from Programa de Doctorado en Ciencias Químicas, Universidad Nacional Autónoma de México (UNAM). Saul Jiménez-Jiménez thanks for the fellowship 314168 from CONAHCYT previously received. José-Gerardo Mejía-Hernández thanks for the fellowship 1313023 received from CONAHCYT.
REFERENCES
- Adamczyk P, Polanowski P, Sikorski A. Percolation in polymer-solvent systems: A Monte Carlo study. J Chem Phys. 2009 Dec 21;131(23).
- Adepu S, Ramakrishna S. Controlled drug delivery systems: current status and future directions. Molecules. 2021 Sep 29;26(19):5905.
- Aguilar-De-Leyva A, Gonçalves-Araujo T, Daza V, Caraballo I. A new deferiprone controlled release system obtained by ultrasound-assisted compression. Pharmaceutical Development and Technology. 2014 Sep 1;19(6):728-34.
- Ahualli S, Martin-Molina A, Maroto-Centeno JA, Quesada-Perez M. Interaction between ideal neutral nanogels: A monte carlo simulation study. Macromolecules. 2017 Mar 14;50(5):2229-38.
- Al-Tabakha MM, Alomar MJ. In vitro dissolution and in silico modeling shortcuts in bioequivalence testing. Pharmaceutics. 2020 Jan 4;12(1):45.
- Andreadis II, Gioumouxouzis CI, Eleftheriadis GK, Fatouros DG. The advent of a new era in digital healthcare: a role for 3D printing technologies in drug manufacturing?. Pharmaceutics . 2022 Mar 10;14(3):609.
- Avalle P, Pygall SR, Pritchard J, Jastrzemska A. Interrogating erosion-based drug liberation phenomena from hydrophilic matrices using near infrared (NIR) spectroscopy. Eur J Pharm Sci. 2013 Jan 23;48(1-2):72-9.
- Axpe E, Chan D, Offeddu GS, Chang Y, Merida D, Hernandez HL, et al. A multiscale model for solute diffusion in hydrogels. Macromolecules . 2019 Sep 3;52(18):6889-97.
- Bannigan, P., Bao, Z., Hickman, R.J. et al. Machine learning models to accelerate the design of polymeric long-acting injectables. Nat Commun. 2023;14:35.
- Barry BW. Drug delivery routes in skin: a novel approach. Adv Drug Delivery Rev. 2002 Nov 1;54:S31-40.
- Benkő E, Ilič IG, Kristó K, Regdon Jr G, Csóka I, Pintye-Hódi K, et al. Predicting drug release rate of implantable matrices and better understanding of the underlying mechanisms through experimental design and artificial neural network-based modelling. Pharmaceutics . 2022 Jan 19;14(2):228.
- Bertrand N, Leclair G, Hildgen P. Modeling drug release from bioerodible microspheres using a cellular automaton. Int J Pharm. 2007 Oct 1;343(1-2):196-207.
- Beugeling M, Grasmeijer N, Born PA, van der Meulen M, van der Kooij RS, Schwengle K, et al. The mechanism behind the biphasic pulsatile drug release from physically mixed poly (dl-lactic (-co-glycolic) acid)-based compacts. Int J Pharm . 2018 Nov 15;551(1-2):195-202.
- Bouzo BL, Calvelo M, Martin-Pastor M, Garcia-Fandino R, de la Fuente M. In vitro-in silico modeling approach to rationally designed simple and versatile drug delivery systems. J Phys Chem B. 2020 Jun 11;124(28):5788-800.
- Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomed. 2015 Feb 2:975-99.
- Bruschi ML. Strategies to modify the drug release from pharmaceutical systems. Woodhead Publisher. 2015 Jun 16.
- Bunde A, Havlin S, Nossal R, Stanley HE, Weiss GH. On controlled diffusion‐limited drug release from a leaky matrix. The Journal of chemical physics. 1985 Dec 1;83(11):5909-13
- Campiñez MD, Benito E, Romero-Azogil L, Aguilar-de-Leyva A, de Gracia Garcia-Martin M, Galbis JA, Caraballo I. Development and characterization of new functionalized polyurethanes for sustained and site-specific drug release in the gastrointestinal tract. European journal of pharmaceutical sciences. 2017 Mar 30;100:285-95.
- Cao J, Liu T, Han Z, Tu B. Sulfate ions diffusion in concrete under coupled effect of compression load and dry-wet circulation. Math Biosci and Engineering. 2023 Mar 1;20(6):9965-91.
- Caraballo I, Melgoza LM, Alvarez-Fuentes J, Soriano MC, Rabasco AM. Design of controlled release inert matrices of naltrexone hydrochloride based on percolation concepts. International journal of pharmaceutics. 1999 Apr 20;181(1):23-30.
- Caraballo I. Critical points in the formulation of pharmaceutical swellable controlled release dosage forms-Influence of particle size. Particuology. 2009 Dec 1;7(6):421-5.
- Casalini T. Not only in silico drug discovery: Molecular modeling towards in silico drug delivery formulations. J Controlled Release. 2021 Apr 10;332:390-417.
- Turnbull D, Cohen MH. On the free‐volume model of the liquid‐glass transition. The Journal of chemical physics . 1970 Mar 15;52(6):3038-41.
- Coleman K. The ICH Q9 Revision and a Renewed Focus on Quality Risk Management Fundamentals. Pharm Technol. 2023 Dec 2;47(12):28-30.
- Colombo P, Bettini R, Massimo G, Catellani PL, Santi P, Peppas NA. Drug diffusion front movement is important in drug release control from swellable matrix tablets. J Pharm Sci. 1995 Aug;84(8):991-7.
- Corsaro C, Neri G, Mezzasalma AM, Fazio E. Weibull modeling of controlled drug release from Ag-PMA Nanosystems. Polymers. 2021 Aug 27;13(17):2897.
- Cukier RI. Diffusion of Brownian spheres in semidilute polymer solutions. Macromolecules . 1984 Mar;17(2):252-5.
- Dan N. Drug release through liposome pores. Colloids Surf B. 2015 Feb 1;126:80-6.
- Davanço MG, Campos DR, de Oliveira Carvalho P. In vitro-In vivo correlation in the development of oral drug formulation: A screenshot of the last two decades. Int J Pharm . 2020 Apr 30;580:119210.
- Davit BM, Kanfer I, Tsang YC, Cardot JM. BCS biowaivers: similarities and differences among EMA, FDA, and WHO requirements. The AAPS J. 2016 May;18:612-8.
- De Izarra A, Jang YH, Lansac Y. DNA-assisted assembly of cationic gold nanoparticles: Monte Carlo simulation. Soft Matter. 2021;17(41):9315-25.
- Draksler P, Mikac U, Laggner P, Paudel A, Janković B. Polyethylene oxide matrix tablet swelling evolution: The impact of molecular weight and tablet composition. Acta Pharmaceutica. 2021 Jun 30;71(2):215-43.
- Du J, Strenzke G, Bück A, Tsotsas E. Monte Carlo modeling of spray agglomeration in a cylindrical fluidized bed: From batch-wise to continuous processes. Powder Technol. 2022 Jan 1;396:113-26.
- Fan H, Lin Q, Morrissey GR, Khavari PA. Immunization via hair follicles by topical application of naked DNA to normal skin. Nat Biotechnol. 1999 Sep;17(9):870-2.
- Fathi M, Mohebbi M, Varshosaz J, Shahidi F. Cellular automata modeling of hesperetin release phenomenon from lipid nanocarriers. Food Bioprocess Technol. 2013 Nov;6:3134-42.
- Feller W. An introduction to probability theory and its applications, Volume 2. John Wiley & Sons; 1991 Jan 8.
- Fernandez-Lopez L, Roda S, Robles-Martín A, Muñoz-Tafalla R, Almendral D, Ferrer M, et al. Enhancing the Hydrolytic Activity of a Lipase towards Larger Triglycerides through Lid Domain Engineering. Int J Mol Sci. 2023 Sep 6;24(18):13768.
- Fishman G. Monte Carlo: concepts, algorithms, and applications. Springer Science & Business Media; 2013 Mar 9.
- Fu Y, Kao WJ. Drug release kinetics and transport mechanisms of non-degradable and degradable polymeric delivery systems. Expert Opin Drug Delivery. 2010 Apr 1;7(4):429-44.
- Fuertes I, Miranda A, Millán M, Caraballo I. Estimation of the percolation thresholds in acyclovir hydrophilic matrix tablets. European journal of pharmaceutics and biopharmaceutics. 2006 Nov 1;64(3):336-42.
- Fuertes I, Caraballo I, Miranda A, Millán M. Study of critical points of drugs with different solubilities in hydrophilic matrices. International journal of pharmaceutics . 2010 Jan 4;383(1-2):138-46.
- Fukś H, Mudiyanselage SA. Deterministic cellular automata resembling diffusion. International Journal of Modern Physics C. 2022 Nov 21;33(11):2250148.
- Galdón E, Millán-Jiménez M, Mora-Castaño G, de Ilarduya AM, Caraballo I. A Biodegradable Copolyester, Poly (butylene succinate-co-ε-caprolactone), as a High Efficiency Matrix Former for Controlled Release of Drugs. Pharmaceutics . 2021 Jul 10;13(7):1057.
- Gao J, Karp JM, Langer R, Joshi N. The future of drug delivery. Chem Mater. 2023 Jan 24;35(2):359-63.
- Gao L, Guo Q, Lin H, Pan D, Huang X, Lin J, et al. Modeling of lactose enzymatic hydrolysis using Monte Carlo method. Electron J Biotechnol. 2019 Jul 1;40:78-83.
- Geraili A, Xing M, Mequanint K. Design and fabrication of drug‐delivery systems toward adjustable release profiles for personalized treatment. View. 2021 Oct;2(5):20200126.
- Gonçalves-Araújo T, Rajabi-Siahboomi AR, Caraballo I. Polymer percolation threshold in HPMC extended release formulation of carbamazepine and verapamil HCl. AAPS PharmSciTech. 2010 Jun;11:558-62.
- Gomes Filho MS, Oliveira FA, Barbosa MA. A statistical mechanical model for drug release: Investigations on size and porosity dependence. Phys A: Statistical Mechanics and its Applications. 2016 Oct 15;460:29-37.
- Grund J, Koerber M, Walther M, Bodmeier R. The effect of polymer properties on direct compression and drug release from water-insoluble controlled release matrix tablets. International journal of pharmaceutics . 2014 Jul 20;469(1):94-101.
- Han R, Xiong H, Ye Z, Yang Y, Huang T, Jing Q, et al. Predicting physical stability of solid dispersions by machine learning techniques. J Controlled Release . 2019 Oct 1;311:16-25.
- Han Y, Shchukin D, Fernandes P, Mutihac RC, Möhwald H. Mechanism and kinetics of controlled drug release by temperature stimuli responsive protein nanocontainers. Soft Matter . 2010;6(19):4942-7.
- Herrmann IK, Wood MJ, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021 Jul;16(7):748-59.
- Hoffman RM. The hair follicle as a gene therapy target. Nat Biotechnol . 2000 Jan;18(1):20-1.
- Hu Y, Kim Y, Jeong JP, Park S, Shin Y, Hong IK, et al. Novel temperature/pH-responsive hydrogels based on succinoglycan/poly (N-isopropylacrylamide) with improved mechanical and swelling properties. Eur Polym J. 2022 Jul 5;174:111308.
- Huang J, Goolcharran C, Ghosh K. A quality by design approach to investigate tablet dissolution shift upon accelerated stability by multivariate methods. European Journal of Pharmaceutics and Biopharmaceutics. 2011 May 1;78(1):141-50.
- Hughes BD. Random walks and random environments. Oxford University Press; 1996 Jun 13.
- Hwang KM, Cho CH, Tung NT, Kim JY, Rhee YS, Park ES. Release kinetics of highly porous floating tablets containing cilostazol.European Journal of Pharmaceutics and Biopharmaceutics. 2017 Jun 1;115:39-51.
- Islam MA, Barua S, Barua D. A multiscale modeling study of particle size effects on the tissue penetration efficacy of drug-delivery nanoparticles. BMC Syst Biol. 2017 Dec;11:1-3.
-
Jagusiak A, Chlopas K, Zemanek G, Wolski P, Panczyk T. Controlled release of doxorubicin from the drug delivery formulation composed of single-walled carbon nanotubes and congo red: A molecular dynamics study and dynamic light scattering analysis. Pharmaceutics. 2020 Jul 3;12(7):622. Pharmaceutics , 12. https://api.semanticscholar.org/CorpusID:220412589
» https://api.semanticscholar.org/CorpusID:220412589 - Jiang X, Abedi K, Shi J. Polymeric nanoparticles for RNA delivery. Reference Module in Mater Sci Mater Eng. 2021.
- Jimenez-Jimenez S, Hashimoto K, Santana O, Aguirre J, Kuchitsu K, Cárdenas L. Emerging roles of tetraspanins in plant inter-cellular and inter-kingdom communication. Plant signaling & behavior. 2019 Apr 3;14(4):e1581559.
- Juszczyk E, Kulinowski P, Baran E, Birczyński A, Klaja J, Majda D, et al. Hydration Patterns in Sodium Alginate Polymeric Matrix Tablets-The Result of Drug Substance Incorporation. Materials. 2021 Jan;14(21):6531.
- Khizer Z, Nirwan JS, Conway BR, Ghori MU. Okra (Hibiscus esculentus) gum based hydrophilic matrices for controlled drug delivery applications: Estimation of percolation threshold. International journal of biological macromolecules. 2020 Jul 15;155:835-45.
- Kambayashi A. In Silico Modeling Approaches Coupled with In Vitro Characterization in Predicting In Vivo Performance of Drug Delivery System Formulations. Mol Pharm. 2023 Jul 31;20(9):4344-53.
- Kaoui B. Computer simulations of drug release from a liposome into the bloodstream. Eur Phys J E. 2018 Feb;41:1-6.
- Kashkooli FM, Soltani M, Souri M. Controlled anti-cancer drug release through advanced nano-drug delivery systems: Static and dynamic targeting strategies. J Controlled Release . 2020 Nov 10;327:316-49
- Kaur P, Jiang X, Duan J, Stier E. Applications of in vitro-in vivo correlations in generic drug development: case studies. The AAPS Journal. 2015 Jul;17:1035-9.
- Kimura G, Puchkov M, Leuenberger H. An attempt to calculate in silico disintegration time of tablets containing mefenamic acid, a low water-soluble drug. Journal of pharmaceutical sciences. 2013 Jul 1;102(7):2166-78.
- Kobryń J, Sowa S, Gasztych M, Dryś A, Musiał W. Influence of hydrophilic polymers on the factor in weibull equation applied to the release kinetics of a biologically active complex of Aesculus hippocastanum. Int J Polym Sci. 2017;2017.
- Kocaaga B, Guner FS, Kurkcuoglu O. Molecular dynamics simulations can predict the optimum drug loading amount in pectin hydrogels for controlled release. Mater Today Commun. 2022 Jun 1;31:103268.
- Koomullil R, Tehrani B, Goliwas K, Wang Y, Ponnazhagan S, Berry J, Deshane J. Computational simulation of exosome transport in tumor microenvironment. Frontiers in Medicine. 2021 Apr 13;8:643793.
- Kosmidis K, Argyrakis P, Macheras P. A reappraisal of drug release laws using Monte Carlo simulations: the prevalence of the Weibull function. Pharm Res. 2003a Jul;20:988-95.
- Kosmidis K, Argyrakis P, Macheras P. Fractal kinetics in drug release from finite fractal matrices. J Chem Phys. 2003b Sep 22;119(12):6373-7.
- Kosmidis K, Dassios G. Monte Carlo simulations in drug release. J Pharmacokinet Pharmacodyn. 2019 Apr 1;46:165-72.
- Kosmidis K, Macheras P. Monte Carlo simulations for the study of drug release from matrices with high and low diffusivity areas. Int J Pharm . 2007 Oct 1;343(1-2):166-72.
- Kosmidis K, Rinaki E, Argyrakis P, Macheras P. Analysis of Case II drug transport with radial and axial release from cylinders. Int J Pharm . 2003 Mar 26;254(2):183-8.
- Kroese DP, Taimre T, Botev ZI. Handbook of monte carlo methods. John Wiley & Sons; 2013 Jun 6.
- Kulagin AE, Shapovalov AV. Analytical description of the diffusion in a cellular automaton with the Margolus neighbourhood in terms of the two-dimensional Markov chain. Mathematics. 2023 Jan 22;11(3):584.
- Kulinowski P, Hudy W, Mendyk A, Juszczyk E, Węglarz WP, Jachowicz R, et al. The relationship between the evolution of an internal structure and drug dissolution from controlled-release matrix tablets. Aaps Pharmscitech. 2016 Jun;17:735-42
- Kulinowski P, Młynarczyk A, Jasiński K, Talik P, Gruwel ML, Tomanek B, et al. Magnetic resonance microscopy for assessment of morphological changes in hydrating hydroxypropylmethylcellulose matrix tablets in situ-is it possible to detect phenomena related to drug dissolution within the hydrated matrices? Pharm Res . 2014 Sep;31:2383-92.
- Laaksonen H, Hirvonen J, Laaksonen T. Cellular automata model for swelling-controlled drug release. Int J Pharm . 2009 Oct 1;380(1-2):25-32.
- Landau D, Binder K. A guide to Monte Carlo simulations in statistical physics. Cambridge University Press. 2021 Jul 29.
- Langer R. New methods of drug delivery. Science. 1990 Sep 28;249(4976):1527-33.
- Lemos HD, Prado LD, Rocha HV. Use of biorelevant dissolution media in dissolution tests as a predictive method of oral bioavailability. Braz J Pharm Sci . 2022 Jul 13;58:e19759.
- Lenzini S, Bargi R, Chung G, Shin JW. Matrix mechanics and water permeation regulate extracellular vesicle transport. Nat Nanotechnol . 2020 Mar;15(3):217-23.
- Liang J, Pitsillou E, Ververis K, Guallar V, Hung A, Karagiannis TC. Small molecule interactions with the SARS-CoV-2 main protease: In silico all-atom microsecond MD simulations, PELE Monte Carlo simulations, and determination of in vitro activity inhibition. J Mol Graphics Modell. 2022 Jan 1;110:108050.
- Liang X, Bos C, Hermans M, Richardson I. An improved cellular automata solidification model considering kinetic undercooling. Metall Mater Trans B. 2023 Jun;54(3):1088-98.
- Liao J, Hou B, Huang H. Preparation, properties and drug controlled release of chitin-based hydrogels: An updated review. Carbohydr Polym. 2022 May 1;283:119177.
- Liu Y, Bravo KM, Liu J. Targeted liposomal drug delivery: a nanoscience and biophysical perspective. Nanoscale Horiz. 2021;6(2):78-94.
- Ma J, Lin P. Simulation approach for random diffusion of chloride in concrete under sustained load with cellular automata. Materials. 2022 Jun 21;15(13):4384.
- Macha IJ, Ben-Nissan B, Vilchevskaya EN, Morozova AS, Abali BE, Müller WH, et al. Drug delivery from polymer-based nanopharmaceuticals-an experimental study complemented by simulations of selected diffusion processes. Frontiers in bioengineering and biotechnology. 2019 Mar 8;7:37.
- Mackie JS, Meares P. The diffusion of electrolytes in a cation-exchange resin membrane I. Theoretical. Proceedings of the Royal Society of London. Series A. Math Phys Sci. 1955 Nov 22;232(1191):498-509.
- Maghsoodi M, Barghi L. Polymer percolation threshold in multi-component HPMC matrices tablets. Advanced Pharmaceutical Bulletin. 2011 Jun;1(1):27.
- Manaia EB, Abuçafy MP, Chiari-Andréo BG, Silva BL, Oshiro Junior JA, Chiavacci LA. Physicochemical characterization of drug nanocarriers. Int J Nanomed . 2017 Jul 13:4991-5011.
- Mandal AS, Biswas N, Karim KM, Guha A, Chatterjee S, Behera M, et al. Drug delivery system based on chronobiology-A review. J Controlled Release . 2010 Nov 1;147(3):314-25.
- Malonis P, Lagaros ND. Reliability-based structural optimization using neural networks and Monte Carlo simulation. Computer Methods in Applied Mechanics and Engineering. 2002 Jun 7;191(32):3491-507.
- Martín-Camacho UJ, Rodríguez-Barajas N, Sánchez-Burgos JA, Pérez-Larios A. Weibull β value for the discernment of drug release mechanism of PLGA particles. Int J Pharm . 2023 Jun 10;640:123017.
- Martínez L, Betz G, Villalobos R, Melgoza L, Young PM. Correlation between compactibility values and excipient cluster size using an in silico approach. Drug Development and Industrial Pharmacy. 2013 Feb 1;39(2):374-81.
- Martínez L, Villalobos R, Sánchez M, Cruz J, Ganem A, Melgoza LM. Monte Carlo simulations for the study of drug release from cylindrical matrix systems with an inert nucleus. Int J Pharm . 2009 Mar 18;369(1-2):38-46.
- Mason LM, Campiñez MD, Pygall SR, Burley JC, Gupta P, Storey DE, et al. The influence of polymer content on early gel-layer formation in HPMC matrices: The use of CLSM visualisation to identify the percolation threshold. Eur J Pharm Biopharm. 2015 Aug 1;94:485-92.
- Medina P, Carrasco SC, Jofré MS, Rogan J, Valdivia JA. Characterizing diffusion processes in city traffic. Chaos, Solitons & Fractals. 2022 Dec 1;165:112846.
- O’Farrell C, Simmons MJ, Batchelor HK, Stamatopoulos K. The effect of biorelevant hydrodynamic conditions on drug dissolution from extended-release tablets in the dynamic colon model. Pharmaceutics . 2022 Oct 14;14(10):2193.
- Pannier AK, Shea LD. Controlled release systems for DNA delivery. Mol Ther. 2004 Jul 1;10(1):19-26.
- Papadopoulou V, Kosmidis K, Vlachou M, Macheras P. On the use of the Weibull function for the discernment of drug release mechanisms. Int J Pharm . 2006 Feb 17;309(1-2):44-50.
- Park H, Otte A, Park K. Evolution of drug delivery systems: From 1950 to 2020 and beyond. J Controlled Release . 2022 Feb 1;342:53-65.
- Park K. Controlled drug delivery systems: past forward and future back. J Controlled Release . 2014 Sep 28;190:3-8.
- Pawłowska M, Sikorski A. Monte carlo study of the percolation in two-dimensional polymer systems. J Mol Model. 2013 Oct;19:4251-8.
- Peppas NA, Franson NM. The swelling interface number as a criterion for prediction of diffusional solute release mechanisms in swellable polymers. J Polym Sci, Polym Phys Ed. 1983 Jun;21(6):983-97.
- Peraman R, Bhadraya K, Padmanabha Reddy Y. Analytical quality by design: a tool for regulatory flexibility and robust analytics. Int J Anal Chem. 2015 Feb 2;2015.
- Pérez-Mas L, Martín-Molina A, Quesada-Pérez M, Moncho-Jordá A. Maximizing the absorption of small cosolutes inside neutral hydrogels: steric exclusion versus hydrophobic adhesion. Phys Chem Chem Phys. 2018;20(4):2814-25.
- Polanowski P, Sikorski A. Diffusion of small particles in polymer films. J Chem Phys . 2017 Jul 7;147(1).
- Politis SN, Colombo P, Colombo G, Rekkas DM. Design of experiments (DoE) in pharmaceutical development. Drug Dev Ind Pharm. 2017 Jun 3;43(6):889-901.
- Poornima G, Harini K, Pallavi P, Gowtham P, Girigoswami K, Girigoswami A. RNA-A choice of potential drug delivery system. Int J Polym Mater Polym Biomater. 2023 Jul 3;72(10):778-92.
- Pramanik A, Garg S. Design of diffusion‐controlled drug delivery devices for controlled release of Paclitaxel. Chem Biol Drug Des. 2019 Aug;94(2):1478-87.
- Pramod K, Tahir MA, Charoo NA, Ansari SH, Ali J. Pharmaceutical product development: A quality by design approach. Int J Pharm Invest. 2016 Jul;6(3): 129.
- Queiroz AL, Wood B, Faisal W, Farag F, Garvie-Cook H, Glennon B, Vucen S, Crean AM. Application of percolation threshold to disintegration and dissolution of ibuprofen tablets with different microcrystalline cellulose grades. International Journal of Pharmaceutics . 2020 Nov 15;589:119838.
- Quesada-Pérez M, Maroto-Centeno JA, Ramos-Tejada MD, Martín-Molina A. Coarse-grained simulations of solute diffusion in crosslinked flexible hydrogels. Macromolecules . 2022 Feb
- Quesada-Pérez M, Martín-Molina A. Monte Carlo simulation of thermo-responsive charged nanogels in salt-free solutions. Soft Matter . 2013;9(29):7086-94.
- Quesada-Pérez M, Martín-Molina A. Solute diffusion in gels: thirty years of simulations. Adv Colloid Interface Sci. 2021 Jan 1;287:102320.
- Ranjan A, Jha PK. Studying drug release through polymeric controlled release formulations in United States pharmacopoeia 2 apparatus using multiphysics simulation and experiments. Mol Pharm . 2021 May 31;18(7):2600-11.
- Reichl LE. A modern course in statistical physics. John Wiley & Sons; 2016 May 31.
- Robert CP, Casella G, Casella G. Monte Carlo statistical methods. New York: Springer; 1999 Aug.
- Romischke J, Scherkus A, Saemann M, Krueger S, Bader R, Kragl U, et al. Swelling and mechanical characterization of polyelectrolyte hydrogels as potential synthetic cartilage substitute materials. Gels. 2022 May 12;8 (5) : 296.
- Russo V, Grénman H, Salmi T, Tesser R. A novel approach to inulin depolymerization: A Monte Carlo based model. Chem Eng Sci. 2022 Jul 20;256:117712.
- Ryu JY, Won EJ, Lee HA, Kim JH, Hui E, Kim HP, et al. Ultrasound-activated particles as CRISPR/Cas9 delivery system for androgenic alopecia therapy. Biomaterials. 2020 Feb 1;232:119736.
- Santana R, Zuluaga R, Gañán P, Arrasate S, Onieva E, González-Díaz H. Predicting coated-nanoparticle drug release systems with perturbation-theory machine learning (PTML) models. Nanoscale. 2020;12(25):13471-83.
- Sarrut D, Etxebeste A, Munoz E, Krah N, Letang JM. Artificial intelligence for Monte Carlo simulation in medical physics. Front Phys. 2021 Oct 28;9:738112.
- Sawilowsky SS. You think you’ve got trivials?. Journal of Modern Applied Statistical Methods. 2003;2: 218-225.
- Senapati S, Upadhyaya A, Dhruw S, Giri D, Maiti P. Controlled DNA delivery using poly (lactide) nanoparticles and understanding the binding interactions. J Phys Chem B . 2021 Aug 26;125(35):10009-17.
- Seo JH, Mittal R. Computational modeling of drug dissolution in the human stomach. Front Phys . 2022 Jan 10;12:755997.
- Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharm. 2015 Dec 1;6:163819.
- Siepmann J, Siegel RA, Rathbone MJ. Fundamentals and applications of controlled release drug delivery. New York: Springer; 2012 Jan 1.
- Tolia G, Li SK. Study of drug release and tablet characteristics of silicone adhesive matrix tablets. European journal of pharmaceutics and biopharmaceutics . 2012 Nov 1;82(3):518-25.
- Tracy T, Wu L, Liu X, Cheng S, Li X. 3D printing: Innovative solutions for patients and pharmaceutical industry. Int J Pharm . 2023 Jan 25;631:122480.
- Vaitukaitis P, Maggiolo D, Remmelgas J, Abrahmsén-Alami S, Bernin D, Siiskonen M, et al. Water transport and absorption in pharmaceutical tablets-a numerical study. Meccanica. 2020 Feb;55:421-33.
- Vernon-Carter EJ, Meraz M, Bello-Perez LA, Alvarez-Ramirez J. Analysis of starch digestograms using Monte Carlo simulations. Carbohydr Polym . 2022 Sep 1;291:119589.
- Villalobos R, V Garcia E, Quintanar D, M Young P. Drug release from inert spherical matrix systems using Monte Carlo simulations. Curr Drug Delivery. 2017 Feb 1;14(1):65-72.
- Wahab M, Mögel HJ, Schiller P. Monte Carlo Simulations of Lipid Bilayers and Liposomes Using Coarse-Grained Models. Adv Planar Lipid Bilayers Liposomes. 2011 Jan 1 (Vol. 14, pp. 157-200). Academic Press.
- Weiss GH. Random walks and their applications: Widely used as mathematical models, random walks play an important role in several areas of physics, chemistry, and biology. Am Sci. 1983 Jan 1;71(1):65-71.
- Wenzel T, Stillhart C, Kleinebudde P, Szepes A. Influence of drug load on dissolution behavior of tablets containing a poorly water-soluble drug: estimation of the percolation threshold. Drug Development and Industrial Pharmacy . 2017 Aug 3;43(8):1265-75.
- Wu IY, Bala S, Škalko-Basnet N, Di Cagno MP. Interpreting non-linear drug diffusion data: Utilizing Korsmeyer-Peppas model to study drug release from liposomes. Eur J Pharm Sci . 2019 Oct 1;138:105026.
- Wu S, Chen Y, Qi C, Liu C, Li G, Zhu H. A 3D Monte Carlo simulation of convective diffusional deposition of ultrafine particles on fiber surfaces. Atmosphere. 2022 Aug 18;13(8):1319.
- El Yacoubi S, El Jai A. Cellular automata modelling and spreadability. Mathematical and Computer Modelling. 2002 Dec 1;36(9-10):1059-74.
- Yassin S, Su K, Lin H, Gladden LF, Zeitler JA. Diffusion and swelling measurements in pharmaceutical powder compacts using terahertz pulsed imaging. J Pharm Sci . 2015 May 1;104(5):1658-67.
- Yekpe K, Abatzoglou N, Bataille B, Gosselin R, Sharkawi T, Simard JS, et al. Developing a quality by design approach to model tablet dissolution testing: an industrial case study. Pharm Dev Technol. 2018 Jul 3;23(6):646-54.
- Yu LX, Amidon G, Khan MA, Hoag SW, Polli J, Raju GK, et al. Understanding pharmaceutical quality by design. The AAPS Journal. 2014 Jul;16:771-83.
- Zhang K, Qian S, Liu Z, Liu H, Lin Z, Heng W, Gao Y, Zhang J, Wei Y. Specific surface area of mannitol rather than particle size dominant the dissolution rate of poorly water-soluble drug tablets: A study of binary mixture. International Journal of Pharmaceutics . 2024 Jul 20;660:124280.
Publication Dates
-
Publication in this collection
20 Jan 2025 -
Date of issue
2025
History
-
Received
08 May 2024 -
Accepted
24 Sept 2024












