Next Article in Journal
Exosome-Based Therapy for Skin Complications in Oncology Patients Treated with EGFR Inhibitors: A Case Report Highlighting the Need for Coordinated Dermato-Oncologic Care
Next Article in Special Issue
From Sea to Therapy: Marine Biomaterials for Drug Delivery and Wound Healing
Previous Article in Journal
Carbohydrate-Based Chiral Ligands for the Enantioselective Addition of Diethylzinc to Aldehydes
Previous Article in Special Issue
Scale-Agnostic Models Based on Dimensionless Quality by Design as Pharmaceutical Development Accelerator
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Strategies for the Formulation of Poorly Water-Soluble Drug Substances by Different Physical Modification Strategies with a Focus on Peroral Applications

1
Division of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3484 CG Utrecht, The Netherlands
2
Department of Pharmacy, Institute of Pharmaceutics and Biopharmaceutics, Philipps-Universität Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
3
Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport (C_DAT), University of Greifswald, Felix-Hausdorff-Straße 3, 17489 Greifswald, Germany
4
Laboratory of Solids Process Engineering, TU Dortmund University, Emil-Figge-Str. 68, 44227 Dortmund, Germany
5
Institute for Particle Technology, Technische Universität Braunschweig, Volkmaroder Str. 4, 38104 Braunschweig, Germany
6
Center of Pharmaceutical Engineering, Technische Universität Braunschweig, Franz-Liszt-Str. 35A, 38106 Braunschweig, Germany
7
Fraunhofer Institute for Surface Engineering and Thin Films IST, Riedekamp 2, 38108 Braunschweig, Germany
8
Institute of Pharmaceutical Technology and Biopharmaceutics, University of Muenster, Corrensstraße 48, 48149 Muenster, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2025, 18(8), 1089; https://doi.org/10.3390/ph18081089
Submission received: 2 June 2025 / Revised: 14 July 2025 / Accepted: 18 July 2025 / Published: 23 July 2025
(This article belongs to the Collection Feature Review Collection in Pharmaceutical Technology)

Abstract

The number of newly developed substances with poor water solubility continually increases. Therefore, specialized formulation strategies are required to overcome the low bioavailability often associated with this property. This review provides an overview of novel physical modification strategies discussed in the literature over the past decades and focuses on oral dosage forms. A distinction is made between ‘brick-dust’ molecules, which are characterized by high melting points due to the solid-state properties of the substances, and ‘grease-ball’ molecules with high lipophilicity. In general, the discussed strategies are divided into the following three main categories: drug nanoparticles, solid dispersions, and lipid-based formulations.

Graphical Abstract

1. Introduction

High bioavailability of medicinal products is often a prerequisite for effective drug therapy. Among other critical parameters, drugs’ solubility in relevant fluids of the human body (e.g., gastrointestinal fluids in the case of an orally administered dosage form) and their ability to cross the biological barrier [1] are two key determinants of their bioavailability. Formulation scientists worldwide are continuously working on new ways to ensure that high percentages of a drug can be absorbed by an organism. However, the differing physicochemical properties of drugs can cause considerable challenges. One of the most prominent challenges is poor solubility in water. As many newly discovered drug candidates are poorly water soluble [2], overcoming this challenge becomes increasingly important, necessitating various strategies to solve this limitation.
Poorly water-soluble drugs are classified in the BCS (Biopharmaceutics Classification System) classes II or IV, which means that a single dose of the drug is not fully soluble in 250 mL of aqueous liquid [3,4]. Despite sharing this characteristic, further physicochemical properties of the substances in these classes can vary significantly. According to the General Solubility Equation (GSE) for organic nonelectrolytes, developed by Yalkowsky and Valvani [5], the following two key factors generally influence the solubility of a substance: the melting point (Tm) and the octanol-water partition coefficient (logP) [6]. In order to enable rough distinction among poorly water-soluble drugs, they are differentiated according to the component that mainly limits their solubility. High melting points indicate the limited solubility of a substance due to solid-state properties and are referred to as ‘brick-dust’ molecules. When solubility is mainly limited by solvation, indicated by high logP values, they are called ‘grease-ball’ molecules [7]. Dependent on these factors, a rough estimation of a suitable formulation strategy can be derived. Lipophilic compounds (‘grease-ball’ molecules), characterized by high logP values, are often formulated in lipid-containing formulations, while high melting points indicate hydrophobic compounds, which are usually formulated in a modified solid state [4].

2. Research Strategy

This review provides an overview of various physical modification strategies that can be used to improve the bioavailability of drugs, with a focus on oral dosage forms. Divided into three main strategies—drug nanoparticles, solid dispersions, and lipid-based formulations—each section briefly introduces the relevant technology. A literature search was conducted focusing on technologies related to the three main strategies identified in this field. Leading studies on these strategies and novel approaches published over the last decades were then selected to provide an overview of current research activities in this field and are summarized in the following review article.

3. Preparation of Drug Nanoparticles

A decreasing drug particle size significantly increases the total surface area, which enhances the dissolution rate, as described by the Noyes–Whitney and Nernst–Brunner equations [8,9]. Furthermore, it is discussed whether the surface curvature of nanoparticles could lead to an increase in the saturation solubility, as described by Kelvin [10] and Ostwald–Freundlich equations [11], which would also lead to a faster dissolution of a drug. Physical approaches applied for the preparation of nanoparticles are mainly divided into top-down methods, such as nanomilling, and bottom-up methods, like precipitation. Both mentioned technologies are the most prominent representatives of these methods and are discussed in detail below.
The term ‘nanoparticle’ or ‘nanoscale’ is, in general, used differently in the pharmaceutical context than is intended with the original definition. According to ISO/TS 80004-1:2023, a nanoparticle is a ‘nano-object with all external dimensions in the nanoscale’, meaning between 1 and 100 nm [12]. When reviewing the pharmaceutical literature, the terms nanocrystals or nanoparticles are often used somewhat differently, namely, for particles in the size range between (sometimes 1 nm but more commonly) 100 and 1000 nm (e.g., [13,14,15]). However, studies indicate that targeting drug particles with sizes below approx. 300 nm can significantly enhance the bioavailability of poorly water-soluble drugs [16].

3.1. Nanomilling of Drug Particles

In the top-down approach to preparing drug nanoparticles, wet media milling is primarily used in the pharmaceutical industry [17]. Other methods, such as high-pressure homogenization, can also be found in the literature [13], but they are not discussed in this review. The first experiments on nanomilling were carried out by Liversidge and co-workers in 1995. They prepared nanoparticles of the drugs naproxen [18] and danazol [19] with mean particle sizes of 169 nm and 270 nm, respectively, and showed a significant increase in bioavailability for both drugs in in vivo studies in rats [18] and beagle dogs [19]. In subsequent years, numerous drugs were milled using various technologies. The following provides an overview of the technologies applied, challenges encountered, and current studies focusing on processing these nanosuspensions into solid forms.

3.1.1. Milling Technologies

In principle, wet media milling describes the mechanical stressing of product particles dispersed in a liquid with grinding beads. The grinding beads can consist of various materials, such as glass or ceramics (e.g., aluminum oxide and yttrium-stabilized zirconium oxide). In some studies, organic milling beads from highly cross-linked polystyrene [20] or frozen water droplets, which melt during the milling process, were used to reduce inorganic product contamination [21]. Depending on the type of mill, the beads are set in motion in different ways. The first experiments by Liversidge et al. were carried out in a roller mill, a cylindrical vessel filled with the suspension and milling beads, which was rotated horizontally for 120 h, with the forces applied being basically the weight forces of the grinding beads [18,19]. Mills such as stirred media mills, in which the beads are set in motion by a rotating rotor, are more suitable, resulting in a much higher energy density and a significant reduction in grinding time. It was shown that particle sizes below 200 nm can be achieved in approx. 60 to 120 min, depending on the formulation [20,22], whereby various mill settings, such as the batch or recirculation mode, can be chosen. Planetary ball mills are operated in the batch mode and use centrifugal forces to move the beads in a closed milling beaker. The milling beakers rotate around their own axis while they are fixed to a disk that rotates around the central axis. This design results in high centrifugal forces, and particles can be stressed in beakers with a volume of up to 1 L [23]. Juhnke et al. introduced a newly designed beaker holder that enables the fixation of up to 24 zirconium dioxide milling beakers with a 0.05 to 1.0 mL individual chamber volume in four milling spaces in order to minimize material consumption and enable formulation screenings at an early stage of formulation development when only limited amounts of the drug are available [24]. Recent studies have shown that screening experiments can also be performed in a dual centrifuge in 2 mL polypropylene screw-cap vials equipped with grinding beads, enabling batch sizes between 10 mg and 1 g. The dual centrifuge differs from the planetary ball mill in that it orients the vials horizontally, which significantly reduces the processing time [25]. In addition, dual centrifugation shows advantages when it comes to narrow size distributions and the processing of formulations with drug concentrations of up to 40%, while experiments with a planetary ball mill showed limitations due to the increased viscosities of these formulations [26]. Upscaling experiments from a dual centrifuge to larger-scale stirred media mills were carried out successfully [27].
In the majority of studies found in the literature to date, water has been utilized as the continuous phase for particle milling. Only a few studies used other liquids such as ethanol [28] or oil [29]. However, all of these studies have in common that they were conducted at room temperature or even actively cooled during processing to prevent heating of the formulation due to mechanical stressing during processing. Igreja et al. chose a different approach and milled griseofulvin particles in a melt containing the matrix material xylitol to stabilize the milled particles directly in a matrix that turns solid at room temperature to improve long-term stability. At milling temperatures of 120 °C, the drug was milled in a custom-built annular gap mill and achieved particle sizes below 300 nm [30,31].

3.1.2. Challenges in the Preparation of Nanoparticles Using Milling

Although milling poorly water-soluble drugs has been shown to improve their bioavailability significantly, there are also challenges that must be addressed during formulation development. A common issue when working with nanoparticles is the thermodynamic instability caused by the large surface area of the particles. To prevent agglomeration of the particles, surface-active additives are added to reduce the free energy of the system and ensure colloidal formulation stability. This can be achieved either by electrostatic repulsion between the particles through the addition of charged or ionic surfactants or by steric repulsion through the addition of non-ionic polymers. A combination of both methods is also possible (electrosteric stabilization) and often used to stabilize drug particles [15,32,33]. Until now, the selection of suitable additives for particle stabilization has mainly been empirical. Recent studies have shown that the Hansen solubility parameters (HSP) could be helpful in evaluating the stabilization capacities of polymeric additives [34,35]. In addition to particle agglomeration, crystal growth phenomena have been observed during wet media milling of particles. These recrystallization and ripening effects are presumably caused by mechanically induced dissolution effects and increased dissolution of the particles after milling [28,32,36,37].
The use of inorganic grinding beads to mechanically stress drug particles can lead to wear and, thus, product contamination, which is a critical concern in pharmaceutical formulations. Even when using high-quality grinding beads, the generation of wear particles cannot be eliminated entirely; rather, this can be reduced by optimizing the grinding process. Various studies have shown that the process parameters, for example, of a stirred media mill, have a significant influence on wear generation [28,38,39]. Recently, Flach et al. found that wear particles are present in nanosuspensions and actively interact with the (nano)particles of a product. They showed that electrosteric stabilization of a suspension can lead to the formation of heteroagglomerates between the product and wear particles due to electrostatic attraction [40]. However, they also observed that product particles can act as a kind of protective shield around the milling beads, protecting them from abrasion. In this experimental setup, the particles are milled in an agglomerated state and dispersed by adding a subsequent stabilizer after the milling process [41].

3.1.3. Processing of Nanosuspension into Solid Form

Drug nanosuspensions are generally unstable systems. Although they can be properly stabilized during the milling process, they often have issues of physical stability in addition to chemical degradation and microbial contamination, which makes achieving long storage times for the formulations difficult. To overcome these stability issues, the nanosuspensions can be further processed into solid forms by various drying methods. Common to all strategies is that they aim to maintain particle stability during further processing in solid form to not affect the improved dissolution rate of the drugs. Recent studies on these different technologies are presented in the following.
During spray drying, a nanosuspension is atomized into a heated gas stream where the liquid evaporates. While thermal stress could have an impact on the redispersibility properties of the nanoparticles [42,43,44], studies have also successfully prepared nanoparticle-containing spray-dried powders with low inlet temperatures, e.g., 100 °C [45]. To enable the easy redispersion of nanoparticles from dry powders, they are often processed with matrices such as sugars, sugar alcohols, or polymers. These (water-soluble) additives are added to the nanoparticle suspension before spray drying to enable the embedding of the nanoparticles during the drying process [43,45,46,47]. Czyz et al. used various sugars and sugar alcohols as additives in the drying process of naproxen and itraconazole nanoparticles. They showed that if good particle redispersibility is to be obtained, the maximum possible drug content in the dried powders is temperature-dependent and that the interaction between the drug particles and the matrix material plays an important role. In addition, they found that with a decreasing particle size, high drug loads become more challenging, and, therefore, an optimum between high amounts of drugs and high dissolution rates needs to be found [48].
Freeze drying is another technology that forms dry nanosuspensions. It is advantageous when heat-sensitive drugs are processed or highly porous structures are to be achieved, e.g., to obtain rapid disintegration in water. Good redispersibility of nanoparticles was also demonstrated with this technology, although further processing of the dried powders could pose a challenge due to the formation of a so-called ‘cake’. In order to prepare spheres with good flowability, the technology was recently further developed with spray–freeze drying [49]. Pellets are formed from a nanosuspension by dripping the formulation into liquid nitrogen before freeze drying. Stable pellets were achieved by adding matrix materials, such as hydroxypropyl cellulose SSL (HPC SSL) [50], polyvinylpyrrolidone (PVP) [51], and sucrose laurate or lactose laurate [52], to obtain well redispersible and low-friable dried pellets [50].
Fluidized bed granulation also yields powders with good flowability. Nanosuspensions were used as granulation liquid and to form a thin soluble shell on carrier particles [53]. Several formulation parameters were determined to influence the dispersibility of nanoparticles in water. First, it was shown that the solubility of the unloaded carrier material of sugar or sugar alcohols influences the redispersibility of the nanoparticles [54,55]. When different carrier material particle sizes were investigated, the results showed that faster dissolution rates were achieved when carriers with smaller particle sizes were used [53]. In addition, the amount of polymer in the nanosuspension was identified as another important formulation parameter. Higher amounts of polymeric additives, such as hydroxypropyl methylcellulose (HPMC) [53,54,55], PVP [56], or vinylpyrrolidone-vinyl acetate copolymer (PVA/VA 64) [54,55], indicated an improved redispersibility of the nanoparticles. Wewers et al. attributed this to the greater distance between the drug nanoparticles in the shell around the carrier particles [54]. Other studies that performed in vivo experiments showed that no differences could be detected between the liquid nanosuspensions and the dried powders, regardless of the spray modes used (top or bottom spray) [57,58], although differences in the spray mode were detected when the powders were analyzed with an in vitro redispersion method [57].
Another, more innovative strategy to maintain nanoparticle size is the embedding of particles in a film-forming matrix. These are dried and form, for example, orodispersible films (ODFs). The films disintegrate and dissolve directly in the mouth so that drug nanoparticles can be swallowed with saliva [59]. Polymers used to embed nanoparticles include HPMC, polyvinyl alcohol (PVA), gelatin, maltodextrins, and starches [60,61]. Nanoparticle loadings of up to 50 wt.% were realized, with only slight increases in particle sizes after the redispersion of the films [62,63]. In addition, Steiner et al. developed a method to prepare a nanoparticle-containing solvent casting mass directly in a stirred media mill to obtain high particle loadings in ODFs and reduce additional preparation steps. Directly after particle milling, they added HPMC powder to the storage vessel of the running stirred media mill and took advantage of the high shear forces of the grinding media to dissolve and homogenize the polymer [64]. Several milled drug nanoparticles have already been embedded in ODFs, such as itraconazole [65], loratadine [66], and fenofibrate or naproxen [67].
Recently, drug-free ODFs have been used as a template for 2D printing of nanoparticular suspensions. Various ODF-like templates can be used, as follows: films with smooth surfaces or more porous films prepared from HPMC by freeze drying [68], or solvent casting of an ethanol HPMC suspension—so-called SOFTs (structured orodispersible film templates) [69]. Milled nanoparticulate drugs such as indomethacin and itraconazole [70], as well as naproxen [71], have already successfully been used in printing. Inks contain different additives such as surfactants to stabilize the particles in the liquid formulation [72] and preserve the particle sizes after ink dries on a template [71].

3.2. Precipitation of Drug Nanoparticles

In contrast to top-down methods, such as milling or high-pressure homogenization, bottom-up methods, which start from molecular solutions of drug substances, are also studied for the preparation of drug nanosuspensions. General overviews and descriptions of the variety of precipitation methods studied are presented elsewhere [73,74,75]. Overall, liquid antisolvent precipitation and so-called flash precipitation in microsystems, which apply very short mixing times, are the most frequently used methods. In these processes, a poorly soluble drug is dissolved in a solvent with sufficient solubility for the drug. This solvent drug solution is then injected into an antisolvent, with which the solvent is miscible. Due to the high supersaturation and applied rapid mixing kinetics, a high number of nuclei will be produced to form drug nanoparticles. In this context, solvent properties influence the precipitation process and the resulting drug’s nanoparticle size distribution [76]. As defined, continuous mixing is mostly applied in low-flow-rate microfluidics, and production rates usually remain small. High-pressure antisolvent precipitation up to 230 mL/min was studied as a novel approach [77], in addition to melt precipitation methods at elevated temperature [78], which is, indeed, only suitable for drugs with very low melting points (such as ibuprofen). For the efficient production of drug nanosuspensions and to disperse and stabilize precipitated drug nanoparticles, combined methods of precipitation and high-pressure homogenization, as well as other top-down methods, have been investigated [79,80].
The drawbacks of such bottom-up precipitation methods are the necessity of finding suitable and water-miscible solvents, dealing with solvent residuals in the product, and the specific challenge of scaling-up such processes to an industrial scale [81]. Due to these drawbacks, precipitation methods are less prominent in industrial applications for the production of drug nanosuspensions.

4. Solid Dispersions

The formation of solid dispersions is another physical formulation strategy to improve drug solubility. The principal idea of solid dispersions was first described by Sekiguchi and Obi in 1961 [82]. While investigating eutectic mixtures of the poorly soluble drug sulfathiazole with different excipients, they developed the concept of finely dispersed drug particles within easily soluble compounds to improve drug dissolution. This initial description has been expanded to accommodate further developments, and, to date, three different classes of solid dispersions have been considered [83], as follows:
(1)
Eutectic mixtures;
(2)
Solid solutions;
(3)
Crystalline dispersions.
A eutectic mixture is a mixture of two compounds in a specific ratio that shows a single melting point below the melting points of the individual compounds. Solid solutions can be further differentiated into substitutional solid solutions, where a solute molecule replaces a solvent molecule; interstitial solid solutions, where solute molecules are distributed within the interstices; and amorphous solid solutions or amorphous solid dispersions (ASDs), where a drug is molecularly dissolved in an amorphous carrier. Crystalline dispersions follow the description of Sekiguchi and Obi, who described the presence of crystalline drugs in a given carrier system. Generally, it has to be said that the term ASD is commonly used for solid solutions and solid dispersions, denoting the formulation principle (drug + polymer) rather than the phase behavior (Figure 1). The mechanisms by which solid dispersions improve drug solubility depend on the type of dispersion formed and include the elimination of crystalline lattice energy by conversion to the amorphous state, a reduction in the particle size, improved wetting, and improved solubilization [4]. Another classification system for solid dispersions proposes the existence of four distinct generations [84]. The first generation includes crystalline and the second generation amorphous carriers. The third generation improves on the second generation by including carriers with surface activity or emulsifying properties. Fourth-generation solid dispersions not only exhibit improved solubility but also show extended release. As classification by generation is comparably diffuse, within the scope of this review, the traditional nomenclature listed before is considered.
In the following sections, the fundamentals and recent developments related to the different approaches to manufacturing, processing, and stabilizing solid dispersions are discussed. Separate sub-sections are dedicated to ASDs, crystalline dispersions, mesoporous systems, and aerogels.

4.1. Amorphous Solid Dispersions (ASDs)

Among the various approaches to enhancing dissolution kinetics and solubility, ASDs hold a special position, as they combine the benefits (and unfortunately disadvantages) of amorphous materials with improved wettability and solubilization utilizing a soluble matrix.
Different technologies have been developed for manufacturing ASDs, and recent developments and novel approaches are presented. The following section first covers the theoretical background of preparing ASDs and then examines the methods, including spray drying, hot-melt extrusion, and electrospinning, used to prepare ASDs. An overview of the discussed technologies is presented in Figure 2.

4.1.1. Theoretical Background to the Preparation of ASDs

As the name implies, the amorphous states of the drug and carrier play crucial roles in the performance of ASDs. The major application cases for solid dispersions are the improvements in the drug dissolution rate and drug solubility, which are among the most important critical quality attributes of dosage forms. For a crystalline drug, three steps have to be passed for the drug to dissolve in a given solvent [86]. In an endothermic reaction, the crystalline lattice has to be disrupted. To host the free drug molecules that detach from the crystalline lattice in the solvent, ‘cavities’ must be created in the solvent via an endothermic reaction called cavitation. In the last step, the drug molecules are hydrated in the cavities via an exothermic reaction. These processes occur in parallel, and the full dissolution process can be overall endo- or exothermic depending on the energy contributions of the endo- and exothermic steps. Following this description, two properties—the lattice energy of the crystalline drug and the hydrophilicity of the drug molecules—govern the dissolution of the crystalline drug in a given medium. Since drug molecules in ASDs are molecularly dispersed, no lattice energy has to be overcome, and dissolution is strongly facilitated.
To convert a drug substance to the amorphous state, sufficient energy must be put into the system to break up the complete crystalline lattice. In Figure 3, the volume and enthalpy of a drug are plotted against temperature. Starting with a crystalline drug, the volume and enthalpy increase linearly during heating until Tm is reached. At Tm, the thermal energy surpasses the lattice energy, and the intermolecular bonds are broken. This is accompanied by strong increases in the volume and enthalpy as the drug molecules now have greater mobility. With further heating, the volume and enthalpy continue to increase linearly, albeit with a higher slope. Upon slow cooling, this process chain is fully reversable. At Tm, the molecules fuse and recrystallization takes place. If the material is quench-cooled, however, the kinetic processes of fusion and recrystallization are prevented, and the molecular mobility is reduced sufficiently to stabilize the amorphous state in a so-called super-cooled liquid. During quench-cooling while still above the glass transition temperature, Tg, the volume and enthalpy decrease linearly. Upon passing Tg, the slope of the volume and enthalpy decreases, which indicates a further reduction in the molecular mobility. The solid amorphous state is, at all temperatures, energetically less favorable and metastable, meaning that recrystallization can occur spontaneously.
In ASDs, drug molecules are dispersed in amorphous matrix materials, commonly polymers, to increase the stability of the drug’s amorphous state. In Figure 4, a schematic phase diagram of a binary ASD formulation, comprising a drug and polymer, is shown. At weight fractions above the solubility line and below the Tg line, ASDs are stable solids. Above both—the solubility and Tg lines—the system is in a liquid state. Below the solubility line, the mixture is metastable, and recrystallization may occur. However, the amorphous state of the drug is kinetically stabilized via entrapment in the polymeric matrix. The time to recrystallization greatly depends on the diffusion coefficient of the drug molecules and their interaction potential. Below the Tg, the drug diffusion coefficient is much lower than if above the Tg, and the ASD is kinetically stabilized. On the other hand, a high molecular affinity of the drug molecules toward each other favors faster recrystallization. At high weight fractions of the drug, amorphous–amorphous phase separation will take place.
The drug–polymer phase behavior is a critical factor in formulation design. A carrier polymer that enables the highest possible drug solubility should be selected, ideally resulting in a single-phase system that remains stable throughout storage. Different models have been developed over time to calculate drug-in-polymer solubility. Recently, a new model that predicts drug-in-polymer solubility using an easily quantifiable thermodynamic property, the enthalpy of melting, was proposed [88]. A free and open-source software application, published alongside this model, simplifies its experimental and practical implementation [89]. Another approach to achieve drug loadings above the thermodynamic solution limit is kinetic stabilization. In such cases, a drug is not fully soluble in the polymer at storage temperature, but stability is maintained over a defined storage period due to kinetically hindered molecular mobility (reduced diffusion coefficient). A high Tg of the polymer is advantageous in this context, allowing the ASD to be stored well below its Tg. In general, higher drug loading compromises the physical stability, as the Tg of the ASD decreases with an increase in drug content. Therefore, an optimal balance must be sought between drug loading and sufficient storage stability for the intended application.

4.1.2. Spray Drying of Protein-Based ASDs

The spray drying of ASDs is a process in which a drug solution with stabilizing excipients rapidly dries from a fine spray at elevated temperatures. Water and organic solvents can be used to solubilize the drug and other formulation ingredients. Despite a high gas temperature, heat-sensitive drugs, including biologics, can be processed, as the drying rate is so high that most of the heat dissipates with the solvent vapor. Spray drying is an energy-intensive but highly versatile process, about which multiple extensive reviews have been written [90,91]. Because of its versatility, spray drying is frequently employed industrially and is of high interest to researchers. Recently, the following novel excipient class for the stabilization of ASDs in spray drying was introduced: proteins.
Initial studies that used ball milling to prepare ASDs based on whey protein isolate found strongly increased Tg values, faster dissolution rates, and high stability of the prepared ASDs [92]. In a later study, also utilizing ball milling, different isolated whey proteins were compared, and it was shown that β-lactoglobulin (BLG), the major component in whey protein isolate, outperformed the other dominant isolate components, α-lactalbumin and casein glycomacropeptides [93]. In a comparison with a common stabilizing polymer, PVP, BLG showed a higher amorphous drug-loading capacity and faster drug release [94]. Spray drying was used to compare ASDs based on BLG with formulations containing hydroxypropyl methylcellulose acetate succinate (HPMC AS) and Eudragit® L containing 50% (w/w) of the model drug rifaximin [95]. While rifaximin was fully amorphized in all ASDs, dissolution in media at pH 1.2 and 4.5, as well as FaSSIF-V2 (fasted state simulated intestinal fluid V2), was faster for the BLG-based ASDs, which also showed the highest Tg, indicating improved storage stability. In simulations, the main stabilization mechanisms in BLG-based ASDs were identified to be steric confinement and hydrogen bonding with lesser contributions by inter-drug hydrogen bond networks, ionic interactions, and increased Tg values [96]. To date, BLG has been tested with multiple model drugs, showing promising results. However, there is still a lack of research on the widespread applicability of BLG, other proteins and protein classes, and manufacturing technologies, in addition to ball milling and spray drying.

4.1.3. Hot-Melt Extrusion

Hot-melt extrusion (HME) is a versatile thermal manufacturing process and was used for the first time in a pharmaceutical context in 1971 [97]. In HME, powdered material is transported in a heated barrel along one or multiple screws. The material is melted or plasticized along the screw(s) either via the energy input from the heated barrel and shear stress caused by the rotating screw or only via the shear stress (autogenic extrusion mode) [98]. The melted drug is intimately mixed with the plasticized polymeric carrier to form an ASD. After extrusion through a die, the material cools down to room or storage temperature. To improve the dispersive and distributive mixing in the process, kneading and mixing zones are usually incorporated in the screw design. In pharmaceutical manufacturing, co-rotating twin-screw extruders are preferred over single-screw extruders because of their increased flexibility, enhanced mixing capabilities, better melt temperature control, and reduced residence time [99]. HME has been widely applied in the preparation of ASDs because it can be considered a near-optimal thermal process to mix poorly soluble drugs with viscous polymer carriers [100]. While HME is a highly capable process for improving drug solubility, the downsides include the demand for good powder flowability and feedability to ensure reproducible manufacturing and the use of high process temperatures, which may limit HME to thermostable drugs [101]. In the following, novel HME approaches to address the process shortcomings or to expand the applicability of the technique are presented and discussed:
a.
HME with carbon dioxide
As stated above, one of the major challenges of ASD manufacturing via HME is the high process temperature required to plasticize the polymer and dissolve the drug in a molecularly dispersed form [102,103]. The thermolabile drug can subsequently degrade and, in addition to reduced effectiveness, form toxic decomposition products [83,104].
A promising strategy to reduce the required temperatures during polymer extrusion is to use carbon dioxide (CO2) as a plasticizing agent [105,106]. By diffusing into the polymer, CO2 increases the free volume between the polymer chains, thereby leading to plasticization at lower temperatures [107]. CO2 is particularly suitable as a plasticizing agent, as its chemical structure facilitates a high solubility in most polymers [108]. Additionally, CO2 is an ideal candidate for pharmaceutical applications due to its inert, non-toxic nature, and it can be removed completely from the final product through decompression, leaving no harmful residues [109].
CO2-assisted extrusion of ASDs showed less degradation of the products, compared with conventional HME processes [110], and faster drug release [111,112], which can be explained by the increased surface area of the extrudates [113,114]. The porous structure also improved the downstream processing of the extrudates. For example, during grinding, higher efficiency and comparatively smaller particle sizes can be achieved with a lower energy input [115,116,117,118]. Furthermore, tablets manufactured from porous extrudates exhibited improved mechanical properties, including greater hardness, higher tensile strength, and reduced friability [116,119]. Stability tests have shown that products processed with CO2 exhibit comparable stability to those processed without it [117]. Although these application studies highlight the potential of optimizing thermal ASD processing through utilization of CO2 as a processing agent, there is a lack of fundamental knowledge concerning the effect of pressurized CO2 on the interaction and phase behavior of the polymer and drug. In a recent study, the influence of CO2 on the dissolution kinetics and phase behavior of ASDs was investigated using high-pressure differential scanning calorimetry. Some of the studied formulations showed phase separation under CO2 loading, which must be considered in the design process [120].
b.
HME and 3D printing using Fused Deposition Modeling (FDM)
HME is a fairly common industrial process in the manufacturing of commercial volumes. Because of its continuous nature and the required time to reach process equilibrium, HME is inherently not suitable for the economic preparation of small or even individualized batches. However, small or even individualized batches and medicines are necessary to fulfill the demands of personalized medicine or to supply individualized medication to pediatric, geriatric, or veterinary patients that cannot be adequately treated with existing medication. Personalized medicine encompasses the concept of utilizing ‘individuals’ phenotypes and genotypes […] for tailoring the right therapeutic strategy for the right person at the right time […]’ [121]. The advent of 3D printing technologies, specifically the fused-deposition modeling (FDM) technique, has introduced a new technology to HME downstream processing. FDM belongs to extrusion-based 3D printing technologies and utilizes continuous filaments as feedstock material, which are produced via HME. These filaments are fed into a heated print head where they are plasticized and deposited on a print bed. The print bed and print head contain actuators that enable movements in the x-, y-, and z-directions of the print head relative to the print bed. This way, 3D structures can be constructed layer-wise from the bottom up. Filaments contain at least one drug and a thermoplastic (polymeric) carrier but can contain further additives to improve the printing performance [122].
As FDM is a downstream processing technology for HME, printed medications can show the same benefits as hot-melt extruded materials. Specifically, FDM can utilize ASD formulations for printing (Figure 5). The ability to increase the drug solubility of BCS class II and IV drugs by processing ASDs into 3D printed forms has been demonstrated in multiple studies [123,124]. Anaya et al. developed a polypill containing nifedipine, simvastatine, and gliclazide, in which all drugs were fully amorphized in the HPMC AS-based matrix [125]. Parulski et al. extruded four formulations containing 25% itraconazole as the model drug [126]. In a 52-week stability study, no recrystallization was found for the printed dosage forms containing only HPMC as the polymer. As mentioned above, FDM requires a second heating step for the actual printing process, which can be leveraged. This additional heating step, which is usually performed at temperatures 20–40 °C above the extrusion temperature to plasticize the material within the short residence time in the print head, can be used to amorphize the drug. Buzukgoz et al. demonstrated this approach for griseofulvin and Hoffmann et al. for escitalopram oxalate [127,128]. This approach mitigates the risk of drug recrystallization in the filament during storage prior to printing. Generally, it can be assumed that every ASD that has the mechanical properties to be printable can be printed in individualized medication and that development approaches and analytical technologies used for ASD development in HME can be directly transferred to FDM. No studies have reported reduced stability after FDM printing.
Unfortunately, FDM also introduces additional process risks. The second heating step to temperatures above the extrusion temperature increases the possibility of drug degradation, which has been investigated in detail for the peptidomimetic drug enalapril maleate [129]. The transition from traditional HME processes, where extrudates are pelletized or milled, to the preparation of continuous filaments also increases process and product demands. Critically, inhomogeneities in the diameter of the extrudate that were previously irrelevant, as the material was homogenized via milling or pelletization and followed by mixing, now became a critical quality attribute. They directly translate to weight and dosage inconsistencies in printed dosage forms [130]. Another issue with FDM is that pharmaceutical polymers have been developed for processability in traditional processes but not in FDM. Filaments require certain mechanical properties to make them feedable and printable, and several pharmaceutical polymers pose challenges in this regard [131,132]. The authors believe that these risks and downsides are easily compensated for by the novel opportunities of FDM. If the extruded filament fulfills all critical attributes, FDM can be considered the easiest to use and most robust 3D printing technology.

4.1.4. Electrospinning of ASDs

Descriptions of the effects of electrostatic attraction on liquids date back to 1600 [133]. However, the concept of electrospinning took some time, with the first patents on electrospinning and suitable devices filed in the late 19th and early 20th century by Cooley [134], Morton [135], and Formhals [136]. Since then, the process has become increasingly refined, and research and industrial scale-up efforts have resulted in over 45,000 publications as of the end of 2024 and created many different usage forms. Overall, the amount of articles on electrospinning has shown exponential growth since 2001 [137]. Whereas the basic setup of an electrospinner can be broken down into four components—high-voltage power supply, conductive collector, syringe pump, and spinneret—the process parameters are numerous and complex. Regarding the material to be electrospun, the process can be divided into the following three main types: melt, solution, and emulsion electrospinning. All three can be used to prepare fibers applicable as a drug delivery system, with the second being the preferred method because of its easy use.
Almost all polymers or substances (hydrophilic or hydrophobic) can be electrospun by one or all methods. The fibrous meshes are commonly classified as solid dispersions, and because of the high level of amorphousness resulting from the preparation process, they are often further categorized as ASDs [138]. Various excellent reviews discuss the physical principles [139,140], different setups [137,141], and release profiles of various ASD types [138]. Hence, this section highlights the advantages and drawbacks of melt (MES), solution (SES), and emulsion electrospinning (EES) (Figure 6). Furthermore, we evaluate and discuss the most recent developments in terms of the amorphousness of an encapsulated drug and polymer and the release profile.
a.
Solution electrospinning (SES)
One of the first and easiest ways to prepare micro- or nanofibers via electrospinning is to dissolve the chosen polymer in an organic or aqueous solvent and electrospin the solution, adjusting direct process parameters like applied voltage, solution flow rate, and the distance between the spinneret and collector. Considering the diverse solution and environmental parameters already described in detail elsewhere, achieving smooth fibers without bead defects might be challenging. However, if the correct settings are found, fiber preparation can be performed conveniently and reproducibly. Based on this, diverse attempts have been made to modify the preparation method to meet the requirements of different drugs, their medical indication, and the requirements of industrial scale-up. In general, SES necessitates the drug and polymer matrix to be dissolved in the same solvent. A high release rate can be expected in most cases because of the high surface-to-volume ratio of a fibrous mesh (especially in nanofibers).
Regarding their indication, complete depletion of a drug at the beginning of the treatment might not always be the right choice. Thus, alternative setups of the spinneret (e.g., coaxial, triaxial, and side-by-side electrospinning) were invented, broadening the possibilities. Another attempt was made by introducing previously prepared nanocarriers into the dissolved polymer solution, overcoming the restrictions imposed by the prerequisite of similar solubilities of the drug and polymer. Altogether, the variety of possible combinations can cover many indications. Regarding industrial scale-up, multi-needle spinnerets and needleless setups made upscaling and continuous manufacturing possible. Despite numerous advantages, some gaps could only be covered by other modifications (Table 1).
b.
Melt electrospinning (MES)
Compared with SES, MES is a relatively neglected topic. In MES, no organic solvent is necessary, earning it the term ‘green option’ by some researchers. The polymer is liquified by an adequate temperature input, enabling the proper flow through a spinneret. According to a web search, using www.webofscience.com, the umbrella term ‘electrospinning’ returned 45,536 publications between 2001 and 2024, whereas ‘melt electrospinning’ was only mentioned 390 times. Less than 10% of both the electrospinning and MES publications contain the term ‘drug delivery’, with approximately twice as many of the latter being review articles.
Unfortunately, only a handful of research articles directly compare SES and MES using the same polymers and drugs, complicating a direct comparison of both regarding drug delivery. Nagy et al. and Balogh et al. incorporated carvedilol, a BCS class II drug, into Eudragit® E and PVP/VA 64, respectively, via MES and SES. Polymers and carvedilol demonstrated amorphous characteristics, leading to a high release rate in both studies [142,143]. Lian et al. loaded increasing concentrations of curcumin, a BCS IV drug, into poly-ε-caprolactone (PCL) fibers. Whereas curcumin turned out to be amorphous using MES and crystalline using SES, it was the other way around for the polymer matrix. PCL’s crystallinity was higher in MES fibers than in SES fibers, resulting in a higher release rate by the latter [144]. Depending on the drug and polymer, similar or diverse degrees of crystallinity of the polymer and drug can be achieved, broadening the outcomes of the release profiles. Further investigations are necessary to enable predictions and more elaborate decision making.
c.
Emulsion electrospinning (EES)
One of the first articles on EES was published in 2006 by Xu et al., who electrospun a W/O emulsion of poly(ethylene oxide) in a poly(ethylene glycol)-poly(L-lactic acid) (PEG-PLA) diblock copolymer [145]. They could prepare core–sheath nanofibers comparable to coaxial electrospinning using a single-needle spinneret. Since then, a simpler equipment setup requiring more complex material preparation has been utilized to achieve core–sheath nanofibers, shifting the workload from process to solution parameters and bringing other advantages and disadvantages [146] (Table 1).
Depending on the medical indication, various release profiles were adapted, going from one extreme to another. By electrospinning an O/W nanoemulsion, Kamali et al. could encapsulate curcumin (BCS IV) in PVP K90 (hydrophilic polymer) fibers. Ty achieved an instantaneous release within the first 30 s, making it applicable as a fast-dissolving oral film [147]. In contrast, Shibata et al. were able to control the release of probucol (BCS II) from PVA nanofibers. They evaluated the effects of the PVA grade, used oil phase, and surfactant concentration on the release pattern. Compared with the simple SES method, which resulted in a burst release of probucol, they achieved a sustained release by fine-tuning EES parameters [148].
d.
Current trends in electrospinning
Electrospun fibers can be used in many ways, but in drug delivery, the focus in mainly on the drug release profiles. Current trends still follow the goal of adjusting the release to the intended application; however, recent advances also focus on the subsequent processes after the release (e.g., degradation, bioavailability). Yu et al. were the first to prepare electrospun PVP K60 fibers capable of the self-assembly of phosphatidylcholine (PC) liposomes when coming in contact with water. The concentration of PC had a direct effect on the liposome size [149]. Friedl et al. improved this idea and fabricated self-emulsifying drug delivery system (SEDDS)-loaded fibers (Section 5.3.2 for SEDDS). They combined Kolliphor EL, Capmul MCM, Captex 355, and Transcutol with Resomer 503H and PVP and electrospun the solution. The release of the model compound curcumin could be controlled by varying the concentration of the SEDDS in the fibers [150]. Ge et al. compared uniaxial and coaxial electrospinning and prepared SEDDS-loaded fibers with paclitaxel (BCS IV). Based on their results, they suggested that the core–shell fibers showed a better performance [151].
Another underrepresented but trending topic are Janus fibers. Janus fibers are two-compartment systems in which both compartments are in direct contact with the environment. Compared with their counterparts—core–shell fibers—as two-compartment systems, Janus fibers create other exciting options in drug delivery. As both compartments are in direct contact with their environment, other release profiles and tunable systems can be designed. Yu et al. prepared side-by-side fibers with PVP K60 (+ketoprofen) on one side and ethyl cellulose (EC) (+ketoprofen) on the other side. Higher release rates could be achieved by adding and increasing the PVP K10 concentration on the EC side [152]. In two consecutive publications, Wang et al. electrospun side-by-side zein–PVP fibers with a two-stage controlled release and, later, even prepared chimeric tri-layer side-by-side fibers [153,154].

4.2. Crystalline Dispersions

Crystalline dispersions enhance the bioavailability and therapeutic efficacy of poorly water-soluble drugs by improving their dissolution. Compared with ASDs, this effect is not attributed to the solid state of a drug but results from embedding a crystalline drug in a hydrophilic carrier matrix. This matrix increases the wettability of the drug so that the dissolution is accelerated [155]. The speed of dissolution can be further amplified by decreasing the size of the drug particles in the carrier matrix. The underlying mechanisms of increased bioavailability for smaller particles have already been described for the nanoparticle formulations above (Section 3.1) and are mainly attributed to the increased surface area, which improves the dissolution kinetics. In this section, the focus is on the superimposed influence of the hydrophilic carrier matrix and the particle size reduction. The handling of small drug particles in the nanometer range requires special measures to achieve their full therapeutic potential and prevent agglomeration. In this context, the carrier matrix captures single drug particles in place and keeps them away from each other so that the advantageous effect of small drug particles is preserved. Furthermore, crystalline dispersions are thermodynamically stable, which offers a long shelf-life. Several processing techniques have been employed to prepare pharmaceutical crystalline dispersions, including spray drying [156], extrusion [157], and melt milling [30], and are partially discussed above (Section 3.1.3).
Co-crystal formulations are a special type of crystalline dispersions. Co-crystallization techniques utilize co-formers to modify the crystalline structure of drugs [158]. The formation of crystalline complexes between a drug molecule and a co-former modulates its physicochemical properties, including solubility, dissolution rate, and stability. The selection of an appropriate co-former and an understanding of molecular interactions are crucial for designing co-crystals with the desired properties and therapeutic effects. The field of co-crystallization continues to evolve rapidly, driven by advances in formulation science, computational modeling, and materials characterization. Current trends include the exploration of novel co-formers, the development of rational design approaches, and the integration of co-crystals into personalized medicine strategies. Future research directions may involve elucidating the mechanistic insights underlying co-crystal formation, optimizing co-crystal synthesis methodologies, and advancing clinical translation to realize the full therapeutic potential of co-crystallization in diverse disease settings. Processing technologies are intended to facilitate intimate mixing and close contact between the drug and co-former molecules, which are critical for the formation of co-crystal bonds [101].

4.3. Mesoporous Systems and Aerogels

Mesoporous carrier systems are an emerging alternative approach to polymer-based systems to keep poorly soluble substances in an amorphous state, which favors improved dissolution and bioavailability. These systems arose in the early 1990s. In such mesoporous systems, which may be made of inorganic or organic materials, a high specific surface area and so-called mesopores in the range of 2 to 50 nm prevail. They provide massive surface interaction potentials and spatial confinement within the pores, which thermodynamically hinder the crystallization of embedded drugs. According to the mentioned structural properties, mesoporous carriers possess high porosity and low density, which is why they are frequently also referred to as aerogels.
In pharmaceutical approaches, mesoporous systems made of organic materials, such as alginate [159], starch [160], or cellulose [161], as well as inorganic materials, are applied, under which silica systems are those most frequently used [162,163,164]. Such amorphous silica is harmless to humans and generally recognized as safe for oral delivery. Hybrid materials combining silica and alginate further broaden the material properties and application by integrating the mechanical strength of silica with the biodegradability of alginate [165]. The predominance of silica as a mesoporous drug carrier system is based on its versatility in the geometric and chemical tunability of pore dimensions and networks, as well as its surface chemistry, due to the presence of silanoles as starting groups [164,166]. Commercial mesoporous systems, especially different types of Syloid® and Neusilin®, are available [167].
The synthesis processes of aerogels are not discussed in detail and can be found elsewhere [162,163,167,168]. In brief, sol-gel synthesis, in which aerogels are finally yielded by supercritical CO2 extraction, and surfactant-templated self-assembly, in which the structures of micelles or liquid–crystalline structures act as spacers for the pores and are chemically or thermally removed from the silica scaffold after synthesis, can mainly be differentiated. The surface functionalization of mesoporous silica is applied to tune the interaction between drug molecules and the carrier with the aim of enhancing the loading capacity and controlling the drug release rates [164,166]. The stabilization of the amorphous state of drugs in mesoporous silica is frequently associated with a reduced molecular mobility due to special confinement, a lower propensity for nucleation, a reduction in Gibbs free energy due to contact with the high surface energy of silica, and specific interactions of the H-bonding with its superficial silanol groups [167].
For embedding poorly soluble drugs into mesoporous systems, it is most important that extensive surface contact between the drug (molecules) and the carrier surface is facilitated. This includes bringing the mostly crystalline drug into an amorphous state, which can generally be achieved via energy input in solvent-free approaches or via dissolution of the crystals in suitable fluids in solvent-based approaches. Overall, a multitude of methods for drug loading into mesoporous carriers have been studied, each influencing the final drug-loading capacity, release properties, and stability of the drug [162,163,164,167,169,170]. The drug can be introduced into the carrier system at different stages during or after its synthesis. During carrier synthesis, drugs can be introduced into the precursor solution or gel or during the final synthesis step of supercritical drying with CO2 [159,171,172,173], in the case that this drying step occurs during carrier synthesis. However, drug loading during synthesis may be critical based on the chemical nature of the drug and whether it can withstand the synthesis conditions without side reactions with the precursors or degradation under the necessary conditions.
In order to prevent chemical reactions during carrier synthesis, post-synthesis loading of pre-formed mesoporous carrier materials can be realized. This may also be the industrially more realistic and economic approach, as the synthesis of the carrier (excipient production) and the loading of the drug (dosage-form manufacturing) can be divided, as is common. Depending on the susceptibility of the drug, solvent-based methods can be applied for heat-sensitive substances—although solvent-free energy input-based methods are commonly more efficient—and they may yield higher drug loadings, as well as being more scalable. Among solvent-based methods, the supercritical CO2 impregnation with the drug resembles the final drying step in aerogel synthesis. Applying this method, the achieved drug loading in the carrier is often low and, crucially, depends on the solubility of the drug in supercritical CO2 [174,175,176]. Other solvents that are liquid under ambient conditions, such as dichloromethane, may be applied in drug loading [162,163,177,178]; however, residual solvents remain a challenge for product development and quality control. The application of a drug solution in carriers can be conducted in a high excess of solvent, relying mainly on molecular adsorption onto the carrier, or without an excess of solvent (i.e., incipient wetness method) with highly concentrated drug solutions, which is meant to dispense higher doses of the applied drug directly into pores. The latter can also be combined with efficient drying methods, such as rotavapor, fluidized bed [179], or spray drying [180].
Solvent-free loading combines the challenge of the amorphization of the drug by controlled energy input with the potential of efficient processing and scalability for industrial production. The energy input can be achieved by direct or indirect introduction of heat. Most simple melt-based approaches consist of the powder blending of the drug and carrier, with subsequent elevation of the temperature in containers [177,180,181,182]. Thermal loading methods have been further developed toward applicability on larger scales using a fluidized bed setup to melt and disperse the drug particles to make contact and be absorbed into carrier particles [183]. Thermal energy can also be introduced by radiation, such as microwaves to amorphize drugs in carrier blends [184]. Some of the previously introduced solvent-based and non-mechanical physical methods have been compared regarding their loading and release kinetics, showing distinct differences depending on the loading method [185].
Further advances in continuous loading methods have been made by studying the processability via hot-melt extrusion of dry powder blends [186], which combines thermal and mechanical energy inputs. It was proven that, depending on the processability of the carrier, hot-melt extrusion can be applied. However, the classical approach to mechanically introducing energy to support the amorphization of drugs in blends with mesoporous carriers is co-milling [187,188]. Herein, drug and carrier powder are blended and, in most cases, stressed by means of mills, applying a grinding media. Most of the energy is dissipated to the particles by means of plastic deformation and heat, which can, in turn, locally facilitate the amorphization and imbibition into the carrier particles. However, comparing the solvent deposition and co-milling showed that, depending on the carrier used, the loading method has an effect on the loading capacity, as well as the dissolution rate, with lower values for the co-milling approach [189]. These effects were traced back to the reduction in pore volume of the carrier due to the mechanical stresses from co-milling. Other studies showed that the physical state of the loaded drug in the pores depended on the loading method, as well as the chemical nature of the drug [179,190].
The loading capacity is generally governed by the specific surface area, mesopore volume, pore size (distribution), pore structure (e.g., ordered or unordered and interconnectivity), and chemical surface properties [164,167,191]. The resultant release rates from mesoporous systems may be up to ten times quicker than for pure drug substances [175,192]. With chemical surface modifications, the release may also be sustained depending on the interactions with the drug [164,176,193]. Additionally, higher amorphous drug loading may also result in a slower release of the drug [167]. The storage stability may be influenced by the abovementioned parameters, as well as the ratio between the critical nucleus size of the drug to be crystallized and the mesopore diameter [194].
Subsequent to the drug loading in the mesoporous carriers, there is the need for further formulation and processing into the final dosage form such as tablets. During tableting, mechanical stresses may again compromise the pore volume of the mesoporous carriers. Nonetheless, the literature reports that improved dissolution kinetics by amorphous loading into mesoporous systems can also be retained after tableting [179,195]. However, mesoporous systems may negatively alter the mechanical properties of tablets [196], producing softer tablets due to the loss in compatibility. Further application strategies for orally delivered mesoporous systems include the formulation of gastric-floating dosage forms [197], loading capacity enhancement of individualized orodispersible films [198], and the enhancement of the loading capacity of amorphous poorly soluble drugs in the 3D printing of pastes [199].
Finally, it should be mentioned that mesoporous silica has also been studied in nanodrug delivery devices in parenteral applications to overcome biological barriers [200] and in cancer treatment [201], as well as against bacteria and their resistances against antibiotics [202].

5. Lipid-Based Formulations

Lipid-based formulations are often used to formulate ‘grease-ball’ compounds with high logP values. They can be associated with physical modification methods, as they utilize non-covalent encapsulation mechanisms. However, the complex interactions between drugs and lipids suggest that these formulations represent a hybrid approach that bridges the gap between purely physical and chemical modification strategies.
Various lipids can be used in these formulations, such as phospholipids, cholesterol, and long- or medium-chain mono-, di-, or triglycerides, as the sole substance or in combination with surfactants. Depending on the utilized additives, some formulations may also have self-emulsifying properties [203,204]. A classification system was introduced in 2000 to enable better evaluation of the critical performance characteristics of lipids as excipients. The LFCS (lipid formulation classification system) divides lipid formulations into Types I to IV. While Type-I formulations consist only of oils, Type-II and -III systems are self-emulsifying drug delivery systems with water-insoluble and water-soluble surfactants, respectively. Formulations with hydrophilic surfactants and co-solvents are classified as LFCS Type IV [205,206].
The first positive effects of lipids on the bioavailability of poorly water-soluble drugs were reported in 1961, when it was shown that the serum concentration of griseofulvin almost doubled after a high-fat breakfast [207]. Later studies showed that lipids could positively influence the intestinal permeability of drugs, whereby a formulation-dependent increase in passive paracellular or transcellular permeation was detected. Paracellular transport occurs when the drug passes through the intercellular gaps between the epithelial cells, which are regulated by the tight junctions. During transcellular permeation, a drug is transported into the cell by membrane vehicles [204]. Furthermore, lymphatic transport by nanoparticular lipid formulations that bypass the first-pass effect in the liver has been reported [208].
Recent studies have reported that various factors can influence the permeability of lipid-based formulations. In addition to the size and shape of lipid particles, the surface charge, surface modification, and type of lipid have a significant impact. It has been shown that oral bioavailability is improved with digestible lipids, such as triglycerides, cholesterol, and phospholipids. The body can assimilate the digested products after hydrolysis. Indigestible lipids (e.g., mineral, essential, and flavor oils) are not hydrolyzed, which limits their positive influence on the bioavailability of drugs. However, these lipids can improve the stability of a nanoparticular formulation. Therefore, combining digestible and indigestible lipids may be advantageous [208]. Differences were also found in relation to the chain length and degree of unsaturation. It was shown that medium-chain triglycerides improve absorption better than long-chain triglycerides, which degrade more slowly [206].
In addition, the enhanced solubilization of lipophilic drugs could increase exposure and reduce food effects, which are often observed after oral administration of poorly water-soluble drugs [209]. Another advantage of lipid-based formulations is that lipids are physiological and, therefore, have a high tolerance in the human body. No toxic co-solvents or pH adjustments are required to solubilize lipophilic drugs in such carrier systems [204]. Because the drug is usually dissolved in lipid-based formulations, they are mainly suitable for low-dose components [209].
In general, lipid-based drug delivery systems are divided into liposomes, micelles, lipid dispersions, such as emulsions and suspensions, and self-(micro/nano)emulsifying systems [204,210] (Figure 7). Nanostructured lipid carriers, which consist of a mixture of liquid and solid lipids, play a rather subordinate role and are not discussed further. In this last section, the latest research progress regarding these different types of formulations is discussed. Although liposomes and micelles are rarely used in the formulation of solid oral dosage forms, they have been included in this review as they represent important lipid-based approaches for poorly water-soluble drugs.

5.1. Liposomes

The success story of liposomes goes back to the mid-1960s, when Bangham et al. first described ‘liquid crystals of lecithin’ that exhibited similar diffusion properties for ions as biological membranes [211]. Being then first used as artificial models for biological membranes, these newly discovered vesicles were soon identified as promising carriers for enzymes and drugs [212,213,214,215]. Their unique structure enables the encapsulation of both hydrophobic and hydrophilic cargoes. After another decade of extensive research in this field, the development of liposomal formulations finally resulted in the first in vivo studies in humans with amphotericin B and doxorubicin in the late 1980s [216,217] and in the clinical application of AmBisome® and Doxil® since the 1990s [218,219]. Their ability to carry a wide range of compounds, including drugs, biologicals, and imaging agents, makes them valuable in both drug delivery and therapeutic applications.

5.1.1. Composition of Liposomes

One of the key components of liposomes are phospholipids. Phospholipids are based on a glycerol backbone with fatty acids esterified at the C1 and C2 positions, as well as a phosphate group at the C3 position. This structure gives phospholipids amphiphilic properties, making them valuable natural solubilizers, wetting agents, and excipients for the formation of (mixed) micelles and liposomes [220]. Phospholipids consist of a hydrophobic tail formed by fatty acids and a hydrophilic head composed of the phosphate group, which is typically further esterified with an additional alcohol, such as choline, ethanolamine, or inositol. This structure allows phospholipids to form vesicles that closely resemble the structure of cell membranes when dispersed in water. In the case of liposomes, phospholipids assemble to form a single or several concentric bilayer structures, with the hydrophobic tails forming a lipid bilayer and the hydrophilic heads facing both the aqueous core and the surrounding aqueous medium. Based on this structure, liposomes are capable of including both hydrophilic cargo molecules in the aqueous core and hydrophobic cargo molecules in the lipophilic bilayer. As liposomal bilayers are fluidic systems, incorporated drugs may leak or be released over time, which can result in unwanted stability issues but also offers the opportunity to modify the drug release [221]. Additional therapeutic benefits, such as a prolonged half-life in the blood stream, improved stability in the gastrointestinal tract, or a targeted delivery of the active drug to, for example, specific tumor tissues, may be achieved by modifying the liposome surface [221,222]. Thus, apart from improving the solubility of poorly water-soluble compounds, drug encapsulation in liposomes is also used to improve drug stability, alter drug pharmacokinetics, or to enable drug delivery to target cells or tissues, making liposomes a very versatile drug delivery technology [223].

5.1.2. Preparation of Liposomal Formulations Containing Poorly Water-Soluble Drugs

Over the years, several different techniques for obtaining liposomal formulations have been reported. As this review aims to provide formulation approaches for improved bioavailability of poorly water-soluble compounds, this section only addresses liposome preparation methods that are primarily capable of incorporating lipophilic drugs into vesicular systems. The most used approach for this purpose is the film hydration technique. In this process, lipids and the lipophilic drug are dissolved in an organic solvent. The solvent is then removed, e.g., by rotary evaporation. Afterwards, the resulting film is hydrated by adding aqueous (buffer) media, which mostly results in the formation of multilamellar vesicles that are then subjected to a homogenization step for reductions in both the particle size and particle size distribution. Another approach, referred to as reverse-phase evaporation, is to directly mix the lipophilic component solution in organic solvents with water to form an emulsion and then to evaporate the organic solvent. With microfluidic methods, liposomes can also be prepared without organic solvent use. In this technique, the lipid phase is directly mixed with the aqueous phase, allowing for precise process control regarding the particle size, vesicle charge, and surface modification [221]. An alternative methodology for conducting the homogenization process in vial is dual centrifugation. This innovative technique, first described by Massing et al., employs a centrifugation mechanism that includes an additional rotational movement of the sample along its vertical axis. This approach generates substantial shear forces, thereby enhancing the efficiency of the homogenization procedure for liposomal formulations [25,224].
A significant limitation of these methods is the relatively low entrapment efficacy, meaning that the drug cargo is not completely incorporated into vesicles [225]. For hydrophilic and ionizable drugs, various techniques have been proposed to actively load the cargo into liposomes. As the drug is loaded into liposomes by a specific trigger after the vesicles are formed, these approaches are also referred to as active loading. Most of these methods leverage the non-ionized state of the drug to facilitate its passage through the lipid bilayer into the aqueous core, where it is directly ionized and trapped. Some principles for actively entrapping hydrophilic drugs inside the aqueous liposome core include the utilization of pH gradients, (calcium)-citrate-based methods, and ammonium sulfate gradients, among others [225]. Considering the loading of liposomes with lipophilic compounds, there are few cases where poorly water-soluble drugs have been remotely loaded into liposomes. A prominent example of a poorly water-soluble drug in a liposomal formulation is paclitaxel, which has been a subject of interest for formulation scientists for decades. While conventional preparation strategies such as thin-film dispersion or extrusion are accompanied with a low entrapment efficacy of approx. 50%, recent advances allowed for significantly increasing the amount of paclitaxel entrapped in liposomal formulations. Recently, two methods for the active loading of liposomes with paclitaxel based on a calcium acetate gradient were reported. Since the mother compound, paclitaxel, is a non-ionizable drug, in the first instance, weak acid derivatives with succinic or phenylboronic acid were synthesized, which were then loaded into aqueous cores of liposomes. Using this approach allowed for the achievement of a notably high encapsulation efficiency of more than 95% [226,227]. In another study, Yu et al. utilized a paclitaxel–doxorubicin prodrug, featuring an anthracene structure capable of interacting with copper ions. By implementing a copper ion gradient, they successfully loaded the paclitaxel–doxorubicin prodrug into liposomes, facilitating a combination therapy involving these two poorly water-soluble anti-cancer agents [228].

5.2. Mixed Micelles

As early as 1909, Moore et al. observed that the addition of lecithin to a solution of bile salts (BS) resulted in the formation of a transparent solution rather than an emulsion or suspension [229]. Furthermore, this solution demonstrated enhanced lipid solubilization properties compared with individual BS or lecithin solutions. In addition, further studies proved that the addition of equimolar amounts of phospholipids to BS solutions resulted in the mitigation of the hemolytic and cytotoxic effects of BS solutions [230,231,232]. The resulting species that derived from mixing BS and phospholipids components, mixed micelles (MMs) exhibit a spherical structure with a hydrophilic surface and hydrophobic core that enable the entrapment of lipophilic compounds (Figure 7). Physiologically, MMs play a key role as natural solubilizers in fat digestion and absorption, but with their amphiphilic structure they can also act as carriers for lipophilic xenobiotics. Based on these structural requisites, MMs also became the focus of formulation scientists as a potential solubilization technology for use in pharmaceutical development. In the early 1970s, initial attempts were made to use MMs in pharmaceutical formulation development for lipophilic and poorly water-soluble drugs. This resulted in the approval of a diazepam MM formulation (Valium® MM) by Hoffmann-La Roche in 1976 [233]. Since then, MM formulations have been investigated for several poorly soluble drugs across various therapeutic areas, including analgesics, sedatives, glucocorticoids, steroid hormones, immunosuppressants, and cytostatic drugs [234,235,236,237]. However, only a few MM formulations, such as Cernevit® (a vitamin blend) or Konakion® MM (a vitamin K1 formulation), have gained marketing authorization and, to date, only Konakion® MM is still in use as a liquid MM formulation that can either be given via the parenteral or the oral route.
The combination of both an excellent toxicity profile of necessary excipients and the versatile use of a single formulation for various routes of administration make MMs a very promising formulation approach for the development of pediatric formulations [238]. Many enabling technologies that are frequently used in adult formulation development make use of excipients, such as synthetic polymers or surfactants, for which evident toxicity data in children are still lacking. While MM formulations containing phospholipids and BS as costly excipients are less attractive than other more cost-effective approaches for adult formulations, MMs may close an important gap in pediatric formulation development. Excipient safety is one key aspect that needs to be specifically addressed when developing novel and state-of-the-art pediatric formulations [239]. The excipients used in MM formulations, phospholipids and BS, are endogenous substances and, thus, come with little to no safety concerns.

5.2.1. Liquid MM Formulations

The solubilizing properties of MMs are very drug-specific and strongly depend on formulation parameters such as the applied phospholipid/BS ratio or the MM concentration. Thus, when developing a novel MM formulation, an initial formulation screening (i.e., determining a formulation composition that allows for maximum drug solubilization) is mandatory. The most widely used method for preparing MM formulations in this screening stage is thin-film dispersion. In this process, the individual components (i.e., phospholipids, BSs, and the drug) are dissolved in organic solvents and mixed at predetermined ratios. In a following step, the solvent is then removed by means of rotary evaporation, and the resulting phospholipid/BS/drug layer is redispersed in a suitable solvent, such as water or a buffer solution, in an inert atmosphere to finally obtain a drug-loaded liquid MM formulation. To obtain MM formulations in a solvent-free process, which should be favored for large-scale production, they can also be prepared in a different manner. At low concentrations, the physiological benefit of MMs as mild solubilizing agents impedes drug loading in the formulations, since MMs are unable to overcome the lattice energy of crystalline drugs to enable drug dissolution. To increase the solubilizing properties of the MM components as part of the production process, there are mainly two ways to prepare MM formulations without organic solvents, as follows: dissolving the drug in highly concentrated BS solutions followed by the addition of phospholipids or the use of highly concentrated MM solutions that are terminally diluted to the target MM concentration after previous drug loading [238].

5.2.2. Solid MM Formulations

All former and presently approved MM formulations are primarily liquids for parenteral use, but due to stability concerns, processes to transform liquid MM formulations to solid, with lyophilization being the most prominent, have been studied. With Cernevit®, the marketing authorization level was reached [240]. With the aim of obtaining oral formulations, the incorporation of MMs in more convenient solid oral dosage forms, such as pellets, mucoadhesive buccal tablets, or fast-dissolving oral films, containing MM precursors and forming MMs after redispersion upon contact with saliva or gastrointestinal fluids was also addressed on an investigational basis but not intended to undergo the approval process [222,241,242,243].

5.3. Lipid Nanoemulsions

5.3.1. Nanoemulsions

In the current context, lipid nanoemulsions are commonly known as dispersions of drug-loaded lipids in an aqueous phase. Caused by their structure, they enable a higher solubilization capacity than micellar systems [244], and the drugs can be localized in the liquid core, droplet interface, and aqueous phase [245,246]. Lipid nanoemulsions have been used in a pharmaceutical context for over five decades and were introduced for parenteral application in patients who are unable to take food orally [247]. For parenteral use, the droplet sizes are strictly controlled to avoid embolisms and, thus, are often in the range between 200 and 400 nm [248]. These formulations are not restricted to only one application route but could be used for different delivery modes, such as topical, ocular, intranasal, or peroral [244,249].
The preparation of lipid emulsions requires successfully overcoming the interfacial tension between the two immiscible liquids. Therefore, an energy input is required, which is often based on high-energy methods. Sonication devices are mostly used in a research environment. The generated vibrations lead to pressure fluctuations in the fluid and cause bubble cavitation, resulting in high shearing energy. Rotor-stator, as well as high-pressure, devices are common in large-scale manufacturing [250]. Another possibility to introduce high shear forces into a formulation is the use of grinding media. This can be conducted, on the one hand, by applying a stirred media mill [251] and, on the other hand, by dual centrifugation in a small-scale approach of approx. 1 mL [25,252]. Although all of these technologies enable the manufacturing of emulsions as soon as the interfacial tension is overcome, these material systems are highly instable, which manifests in various destabilizing mechanisms such as flocculation, Ostwald ripening, and coalescence [250]. Stabilization of a formulation is possible by adding stabilizing agents such as surfactants to the aqueous phase to decrease the interfacial tension and prevent destabilization. Nanoemulsions, in particular, are known to be stable against sedimentation and creaming due so their small droplet sizes, and Ostwald ripening appears to be one of the main causes of a breakdown in the stability of O/W emulsions [253].
Drug loading in lipid emulsions can be performed actively prior to the emulsification process or passively by an incubation step after the emulsion’s preparation. Passive drug loading is advantageous for small setups such as screening approaches, while carrier overloading, possibly resulting in drug recrystallization during storage, was not observed [254]. During active loading, the drug is directly dissolved in the liquid lipid prior to emulsification. As a result of both manufacturing approaches, the drug is dissolved in the dispersed lipid phase of the emulsion in order to enhance the bioavailability of the poorly water-soluble substances. Additionally, it was shown that the encapsulation of the drug molecules in the lipid droplets could prevent oxidation and hydrolysis processes [244,255].
Focusing on the peroral delivery of poorly water-soluble drugs, as this application route is most preferred by patients [256], the positive effects of lipid-based nanoemulsions have been recorded. An increase in antimalarial activity was shown for primaquine when loaded in lipid nanoemulsions, resulting in 25% lower doses [257]. Furthermore, an improved anti-inflammatory effect was identified for a curcumin-loaded lipid nanoemulsion [258], and improved oral bioavailability was reported for drugs such as paclitaxel (dispersed in pine nut oil) [259], breviscapine (dispersed in ethyl oleate) [260], and pterostilbene (dispersed in isopropyl myristate) [255]. So far, most of the lipid-containing systems under investigation are liquid formulations. But studies showed that the lipid nanoemulsions can also be further processed to semi-solid or solid dosage forms. Yu and Huang formulated organogel-based nanoemulsions loaded with curcumin [261], and in other studies, the emulsions were further processed to dry powders by spray drying. These showed that improved oral bioavailability was achieved when the drugs itraconazole [262] or 5-PDTT (5-phenyl-1,2-dithiole-3-thione) [263] were formulated in lipid emulsions and embedded in matrixes during spray drying. In addition, studies showed that the nanoparticular properties of lipid emulsions could be preserved after spray drying, independently of the used oil, when embedded in a matrix containing lactose and the surfactant SDS (sodium dodecyl sulfate) [264]. Furthermore, the latest studies show that lipid nanoemulsions can also be embedded in an orodispersible film matrix. Although the nanoparticular properties of the lipid droplets could not be completely preserved when using HPMC as a film-forming polymer, dissolution studies showed an increased dissolution rate for the embedded fenofibrate [67].

5.3.2. Self-(Micro/Nano)emulsifying Formulations

Unlike nanoemulsions, self-(micro/nano)emulsifying drug delivery systems (nomenclature used is SEDDS, SMEDDS, or SNEDDS) form O/W emulsions when they come into contact with an aqueous phase. The first self-emulsifying drug delivery systems were formulated in the 1970s and 1980s [265,266] using, for example, medium-chain triglycerides and non-ionic surfactants [266] encapsuled in gelatin capsules to enable oral administration [265].
Classic SEDDS contain liquid oils in which the drug is dissolved. Modified or hydrolyzed vegetable oils have been used to improve the success of systems by enabling higher drug loadings. With a share of 30 to 60%, surfactants play an important role in a formulation. Mostly non-ionic surfactants with a high hydrophilic–lipophilic balance (HLB) such as Polysorbate 80 are used [267]. Due to their high share in formulations, emulsifiers from natural sources are expected to be safer, and recent studies have successfully used, for example, phospholipids such as lecithin, which has been classified as GRAS (Generally Recognized As Safe) by the FDA [268]. To improve the solubility of the drug or the surfactant in the lipid, co-solvents such as ethanol or propylene glycol can be added to formulations [267]. During a formulation’s preparation, the dissolution of the drug in the lipid formulation can easily be achieved with low-viscous lipids by, for example, simple stirring processes. However, it is more challenging when lipids with high viscosities or semi-solid formulations are to be formulated. In some cases, this could be solved by higher process temperatures, but this requires the usage of no thermosensitive drugs or excipients. A preparation technique that proved to be particularly suitable for highly viscous systems was introduced by Gruene and Bunjes in 2024. They used dual centrifugation to prepare formulations with five different poorly water-soluble drugs, where the applied centrifugal forces intensify the mixing process [269].
The majority of the SEDDS formulations are applied perorally in patients [270], and the lipid-based, drug-containing formulation is encapsuled in hard or soft capsules [271]. Recent studies show that SEDDS can also be encapsuled in more innovative dosage forms. For example, self-microemulsifying mouth-dissolving films were introduced by Xiao et al. in 2011 [272] and later used in the formulation of the poorly water-soluble drug indomethacin [273]. In a 2019 study, Talekar et al. prepared self-nanoemulsifying orodispersible films (SNEODF) using the BCS class III drug captopril [274]. They formed a stable W/O/W nanoemulsion from the oil phase and an external phase containing the film-forming polymer HPMC, a plasticizer, and water and casted a film from this mixture. Solid self-emulsifying drug delivery systems (S-SEDDS) were introduced by Abdalla and Maeder in 2007 [275], which were further developed in the 2020s by 3D printing oral tablets. Vithani et al. published a proof of concept for the preparation of S-SEDDS in four different shapes loaded with fenofibrate and cinnarizine by printing the lipophilic formulation at 65 °C using a printer-mounted syringe [276]. In addition to printing the molten lipid formulation [277], it was shown that emulsion gels can also be successfully formulated from self-emulsifying formulations and 3D printed [278]. However, in addition to peroral application, other routes of application for SEDDS have also been investigated, such as intranasal application, to allow for better targeting of the brain, which is not discussed in this review but can be referred to in [270,279].

5.4. Solid-Lipid-Based Formulations

Compared with lipid nanoemulsions, lipid nanosuspensions are prepared from a lipid with a crystallin structure at room temperature. The first publications regarding solid lipid nanoparticles go back to the year 1992, where Lucks et al. introduced lipid particles as an alternative parenteral drug delivery system [280]. Lipid nanoparticles are mainly prepared by techniques such as solvent injection, ultrasound technique, microemulsion technique, or high-pressure homogenization [281,282]. Most technologies require a melting of the lipids above their melting point prior to processing to enable emulsification with the aqueous phase. Crystalline particles are formed from the lipid droplets when cooled afterwards. In terms of stability, it has to be considered during formulation development that some lipids such as triglycerides tend to initially crystallize into an almost spherical, meta-stable α-polymorphic form [283]. During storage and time-dependently, the crystals rearrange and transform into a more ordered, platelet-like, and thermodynamically stable β form. Thus, this causes the formation of new surfaces, which could result in instabilities in the absence of a sufficient formulation, on the one hand [284], and impact the drug-loading capacity of particles, on the other hand. Previous studies indicated that a higher drug-loading capacity is achieved when the crystallinity of particles is lower and, thus, drug molecules could be embedded in the defects of the crystal lattice [285]. During particle recrystallization into the stable β-polymorphic form, the number of defects decrease, which can cause drug expulsion [286] and result in crystallization of the pure drug outside of the lipid particle. Thus, the general aim during formulation development should be to either stabilize the solid lipid nanoparticles directly in the α-polymorphic form to prevent polymorphic transition during storage or to formulate particles in the stable β form right at the beginning. Although particles in the β-polymorphic form cannot embed drug molecules in the crystal lattice, high drug loads can be realized with platelet-shaped (tri)glycerides particles (in the β-polymorphic form). The high surface-to-volume ratios enable the accommodation of drug molecules at particle surfaces, which results in high drug loads [283,287]. Investigations with amphotericin B and curcumin showed that, for these drugs, higher loads can be realized with solid lipid nanoparticles as carrier systems compared to lipid nanoemulsions, where embedding in a liquid matrix is possible [246].
Solid lipid nanoparticles have been investigated as carrier systems in various fields for application. Generally, similar to lipid nanoemulsions, they showed that they are biocompatible and suitable for improving the oral bioavailability of embedded substances. Furthermore, they proved to be advantageous in cancer therapy by overcoming several physiological barriers that usually obstruct the delivery of drugs to tumors [288]. Lately, solid lipid nanoparticles have also proved to be a delivery platform of interest for antibiotics, e.g., by overcoming some of the resistance mechanisms that bacterial strains have developed or by providing protection against enzymatic drug degradation [289].
Although prepared lipid nanosuspensions are liquid at the beginning, several studies have focused on further processing drug-loaded nanoparticles to semi-solid or solid dosage forms in order to improve their physical and chemical stability. Recently, freeze- and spray drying have often been used in combination with different matrix materials, such as lactose, glucose, mannitol, or trehalose, to prepare a stable solid formulation [264,290,291]. Studies embedding different lipids in an orodispersible film matrix indicated that this dosage form serves well as a delivery platform for lipid nanoparticles [67,292]. Furthermore, different mucoadhesive forms containing lipid nano- or microparticles were formulated, as follows: buccal films embedding didanosine [293], coumarin 6 [294], or fluconazole [295] into a lipid nanocarrier, as well as lyophilized sponges loaded with curcumin-containing lipid particles [296] or dextran hydrogels for the delivery of ibuprofen [297].

6. Major Findings and Future Perspectives

6.1. Major Findings

The variety of technologies in this review shows that different strategies are available depending on the drug substance, as well as the desired formulation properties. However, it should be noted that each formulation technology has both benefits and drawbacks and not all are suitable to formulate BCS class II and IV drugs. Table 2 provides an overview of the formulation technologies to facilitate selection.
The mentioned technologies are usually first used on a lab scale in a scientific environment. Their industrial relevance often goes hand in hand with their scalability to an industrial scale, but regulatory aspects must also be considered.
Drug nanoparticles have been produced by nanomilling in stirred media mills on an industrial scale for marketed products for more than two decades. The process parameterization for efficient and wear-avoiding process guidance is well established, while suitable formulations still mostly depend on experience and empirics. Accordingly, there are methods for formulation screening on the smallest of lab scales and for scaling up to production scale (such as in [27]). On the industrial scale, aseptic nanomilling lines are established. In contrast to these top-down processes, bottom-up processes, mainly continuous anti-solvent precipitation, are less popular for development and production beyond academic research.
ASDs are already produced on an industrial scale, and corresponding products are commercially available. Successful process development is ensured by using standard equipment (e.g., spray dryers, extruders, and electrospinners), which also allows for the application of established scaling-up strategies. In particular, manufacturing via hot-melt extrusion aligns with the trend toward establishing continuous production processes. Furthermore, this method eliminates the need for solvents. Considering FDM 3D printing as a manufacturing technology, scalability and regulatory issues are highly interconnected. FDM is primarily considered a point-of-care manufacturing technology, requiring centralized filament manufacturing and distribution to end users for final preparation. However, current legislation does not permit the manufacturing of drug-loaded half-products and their distribution to facilities without a manufacturing license, hindering the scalability of this technology. The regulatory framework has to be adapted to reflect such distributed manufacturing models. Additionally, methods where ASD formation may occur in situ during the final printing step at the point of care are associated with uncertain quality requirements. Conventional compounding technologies typically do not alter the solid state of APIs, resulting in a gap in established testing procedures. Therefore, the industrial relevance of filament extrusion coupled with FDM 3D printing is currently low and the future uncertain. In electrospinning, there are two main strategies equipment manufacturers use to upscale production. Multi-needle and needleless electrospinning combined with a continuously moving collector in a roll-to-roll system allow for high production capacities. Multi-needle systems consist of an array of needles, while needleless systems can vary significantly in their overall design. When it comes to the challenges of scaling-up, SES is by far the easiest, followed by EES. Different equipment designs have been proposed for MES upscaling; however, because of the thermal and rheological demands of the polymer melts, MES is rarely implemented on an industrial scale.
Liposomal formulations have been available on the market for nearly three decades; however, they continue to present significant challenges when transitioning processes from laboratory to production scales. During scale-up, it is imperative to implement measures that ensure batch-to-batch homogeneity, particularly in maintaining uniform particle size, encapsulation efficiency, and stability [298]. Although thin-film hydration is most commonly used, from a sustainability perspective, microfluidic technologies offer promising solutions for scalable production processes without the use of organic solvents. In contrast to the success of liposomes, MM formulations remain a relatively rare solubilization technology for commercial formulations, with only a few products available on the market. This is surprising, as the production technology is relatively simple, scalable, and sustainable. In general, regarding regulatory aspects, (phospho-)lipids are considered safe excipients, as well as for pediatric use, which could further promote the use of lipid-based formulations for drug solubilization in future product development.
Lipid nanoemulsions have also been prepared on an industrial scale for several decades in the form of parenteral nutrition by high-pressure homogenization, indicating that there is good certainty of action and that there will be no major regulatory challenges. Lipid nanosuspensions can also be prepared by high-pressure homogenization, but this process should then take place at temperatures above the melting temperature of the lipid. The high temperatures could lead to degradation of the drug, and lipid crystallization, formation of supercooled melts, and gelation phenomena also have be taken into account during preparation. For these reasons, promising approaches focusing on continuous, scalable emulsification processes are still being researched.

6.2. Future Prospective

In the future, research on drug nanoparticles will further focus on the modeling and prediction of formulation parameters, especially stabilization based on the physicochemical properties of the drug molecule to be processed. Additionally, development may focus on wear avoidance or wear separation from product streams, mitigating this major drawback of nanomilling approaches. For nanoparticle precipitation processes, further developments toward continuous process chains for the concentration of nanosuspensions and safe solvent recovery and recycling would be needed to make such processes industrially feasible.
In terms of ASD formulations, in the future, new theoretical modeling approaches are needed to predict drug-in-polymer solubility, ASD stability, and dissolution to reduce experiment efforts. Also, the development of new techniques that enable a better understanding of characteristic ASD behavior is needed. One novel technology is stimulated Raman-scattering microscopy coupled with sum frequency generation, revealing complex phenomena involving water ingress, swelling, drug dissolution, amorphous–amorphous and liquid–liquid phase separation and recrystallization in dissolving ASDs [299]. Models that describe the observed complex behavior need to be developed to predict the physical events during dissolution, and not only the dissolution rate.
Similar to ASD formulations, future research in the field of lipid carrier systems should also focus on predicting the solubility of drugs in different lipids in order to realize high drug loads. In addition, the focus should be on the next generation of solid lipid nanoparticles from polyglycerol esters of fatty acids to overcome stability challenges caused by recrystallization and polymorphism.
In recent years, artificial intelligence (AI) has become an increasingly powerful tool in various areas of research and is also being used to address issues in the field of formulation development. Different AI and machine learning models and tools have already been developed for pharmaceutical product development [300], and new ones are continually being introduced in the literature. Highly specialized platforms such as ‘FormulationAI’ [301] and ‘FormulationDT’ [302] have been developed to support data-driven formulation development, taking into account the solubility aspects of poorly water-soluble drugs. With ‘PharmSD’, a machine leaning platform was developed that focuses exclusively on solid dispersion formulations by predicting the physical stability, dissolution type, and dissolution rate of different drug–polymer combinations [303]. What all of these AI-based models have in common is that they enable the rapid evaluation of possible formulation strategies without time-consuming wet experiments [303]. They also aim to predict pharmacokinetic parameters, drug dosages, and routes of administration [300]. However, a current challenge is the lack of reliable negative examples for training models, as often only positive examples are available [304]. In addition, the reproducibility of the models could be difficult due to different operating environments [301]. It is generally assumed that the prediction accuracy of AI-based models and their importance in formulation development will continue to increase in the coming years.

7. Conclusions

Poorly water-soluble drugs continue to pose a challenge in the development of suitable formulations that ensure high bioavailabilities of these compounds after oral administration. This review provides an overview of the physical modification strategies discussed in the literature to overcome these limitations, focusing on formulations for peroral dosage forms and highlighting the advancements in this field over the past decade. The following three major formulation categories were identified and discussed in detail: drug nanoparticles, solid dispersions, and lipid-based formulations.
Drug nanoparticles can increase the dissolution rate of poorly soluble substances and are mainly prepared by nanomilling or precipitation. Since top-down technology has been the most common in the literature in recent years, different trends in milling equipment and the latest solution-based strategies to overcome challenges during the preparation of nanoparticles were considered. In addition, various studies were highlighted that deal with the further processing of nanoparticles into powders, such as spray- or freeze drying and fluidized bed granulation, as well as more innovative dosage forms like oral films.
With regard to solid dispersions, this section focused majorly on the formulation of ASDs. In addition to discussing spray drying for the preparation of ASDs, different novel HME approaches were presented. Besides current trends in the use of carbon dioxide for the preparation of HMEs, the extrusion-based 3D printing technology FDM was highlighted, which enables the preparation of personalized dosage forms. The latest research findings in the field of electrospinning were then discussed, focusing on the three most relevant technologies in recent years, namely, solution (SES), melt (MES), and emulsion electrospinning (EES). In addition to ASDs, the focus in this section was also on crystalline solid dispersions and amorphous deposition in mesoporous systems, which enable the embedding of drugs in inorganic or organic highly porous structures and, thus, improve bioavailability.
Another established concept for improving the solubility and bioavailability of lipophilic drugs is their formulation in lipid-based formulations. For this purpose, various carrier systems of various sizes have been developed, with (mixed) micelles and liposomes being representatives of the smallest size category. In these formulation concepts, poorly water-soluble drug molecules are incorporated into lipophilic domains, facilitating drug solubilization. Lipid nanoemulsions are larger carrier systems that are either directly dispersed in aqueous media or formulated as self-emulsifying systems that form emulsions upon contact with aqueous media. Solid lipid nanoparticles are based on a lipid that is solid at room temperature and are also used as a carrier system for lipophilic drugs. Recent studies showed successful further processing of lipid-based formulations into solid dosage forms.
Although it was shown that ‘brick-dust’-like molecules, whose solubility is limited mainly due to their solid-state properties that lead to a high melting point, are often formulated in drug nanoparticles or solid dispersions and drugs with high lipophilicity, referred to as ‘grease-ball’ molecules, are primarily formulated in lipid systems, this article also showed that there are many exceptions. Strategies such as drug milling are primarily suitable for many various types of poorly soluble drugs, and the structure of liposomes allows for loading of hydrophilic and lipophilic compounds, indicating that an appropriate formulation strategy is highly dependent on more drug characteristics than only its solubility in water. Therefore, an appropriate formulation strategy has to be chosen that depends on the drug to be applied and the specific requirements for each formulation, such as, for example, the intended drug dose to be administered to the patient. The recent developments highlighted in this review show that, despite numerous publications, there is still room for innovation and improvement in current formulation strategies, and the possibilities are only limited by the researcher’s imagination.

Author Contributions

Conceptualization, J.Q., E.P., F.K., J.W., J.H.F. and D.S.; writing—original draft preparation, J.Q., E.P., F.K., J.W., J.H.F. and D.S.; writing—review and editing, J.Q., E.P., F.K., J.W., J.H.F. and D.S.; visualization, J.Q. and E.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The authors would like to acknowledge the PETRS Network (Pharmaceutical Engineering and Technology Research Scientists), a scientific network of young researchers in the fields of pharmaceutical technology and biopharmaceutics (as well as other related disciplines), to which the authors belong.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stielow, M.; Witczynska, A.; Kubryn, N.; Fijalkowski, L.; Nowaczyk, J.; Nowaczyk, A. The bioavailability of drugs-The current state of knowledge. Molecules 2023, 28, 8038. [Google Scholar] [CrossRef] [PubMed]
  2. Lipp, R. The innovator pipeline: Bioavailability challenges and advanced oral drug delivery opportunities. Am. Pharm. Rev. 2013, 16, 10–16. [Google Scholar]
  3. Amidon, G.L.; Lennernäs, H.; Shah, V.P.; Crison, J.R. A theoretical basis for a biopharmaceutic drug classification: The correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm. Res. 1995, 12, 413–420. [Google Scholar] [CrossRef] [PubMed]
  4. Williams, H.D.; Trevaskis, N.L.; Charman, S.A.; Shanker, R.M.; Charman, W.N.; Pouton, C.W.; Porter, C.J. Strategies to address low drug solubility in discovery and development. Pharmacol. Rev. 2013, 65, 315–499. [Google Scholar] [CrossRef] [PubMed]
  5. Yalkowsky, S.H.; Valvani, S.C. Solubility and partitioning I: Solubility of nonelectrolytes in water. J. Pharm. Sci. 1980, 69, 912–922. [Google Scholar] [CrossRef] [PubMed]
  6. Jain, N.; Yalkowsky, S.H. Estimation of the aqueous solubility I: Application to organic nonelectrolytes. J. Pharm. Sci. 2001, 90, 234–252. [Google Scholar] [CrossRef] [PubMed]
  7. Bergstrom, C.A.S.; Charman, W.N.; Porter, C.J.H. Computational prediction of formulation strategies for beyond-rule-of-5 compounds. Adv. Drug. Deliv. Rev. 2016, 101, 6–21. [Google Scholar] [CrossRef] [PubMed]
  8. Noyes, A.A.; Whitney, W.R. The rate of solution of solid substances in their own solution. J. Am. Chem. Soc. 1897, 19, 930–934. [Google Scholar] [CrossRef]
  9. Wurster, D.E.; Taylor, P.W. Dissolution Rates. J. Pharm. Sci. 1965, 54, 169–175. [Google Scholar] [CrossRef] [PubMed]
  10. Junghanns, J.-U.A.H.; Müller, R.H. Nanocrystal technology, drug delivery and clinical applications. Int. J. Nanomed. 2008, 3, 295–309. [Google Scholar] [CrossRef] [PubMed]
  11. Van Eerdenbrugh, B.; Vermant, J.; Martens, J.A.; Froyen, L.; Humbeeck, J.V.; Van den Mooter, G.; Augustijns, P. Solubility increases associated with crystalline drug nanoparticles: Methodologies and significance. Mol. Pharm. 2010, 7, 1858–1870. [Google Scholar] [CrossRef] [PubMed]
  12. International Standard ISO 80004-1:2023; Nanotechnologies—Vocabulary—Part 1: Core Vocabulary. ISO: Geneva, Switzerland, 2023.
  13. Keck, C.M.; Muller, R.H. Drug nanocrystals of poorly soluble drugs produced by high pressure homogenisation. Eur. J. Pharm. Biopharm. 2006, 62, 3–16. [Google Scholar] [CrossRef] [PubMed]
  14. Gao, L.; Zhang, D.; Chen, M. Drug nanocrystals for the formulation of poorly soluble drugs and its application as a potential drug delivery system. J. Nanopart. Res. 2008, 10, 845–862. [Google Scholar] [CrossRef]
  15. Rabinow, B.E. Nanosuspensions in drug delivery. Nat. Rev. Drug Discov. 2004, 3, 785–796. [Google Scholar] [CrossRef] [PubMed]
  16. Gupta, R.B.; Uday, B.K. Nanoparticle Technology for Drug Delivery; CRC Press: Boca Raton, FL, USA, 2006. [Google Scholar]
  17. Moschwitzer, J.P. Drug nanocrystals in the commercial pharmaceutical development process. Int. J. Pharm. 2013, 453, 142–156. [Google Scholar] [CrossRef] [PubMed]
  18. Liversidge, G.G.; Conzentino, P. Drug particle size reduction for decreasing gastric irritancy and enhancing absorption of naproxen in rats. Int. J. Pharm. 1995, 125, 309–313. [Google Scholar] [CrossRef]
  19. Liversidge, G.G.; Cundy, K.C. Particle size reduction for improvement of oral bioavailability of hydrophobic drugs: I. Absolute oral bioavailability of nanocrystalline danazol in beagle dogs. Int. J. Pharm. 1995, 125, 91–97. [Google Scholar] [CrossRef]
  20. Merisko-Liversidge, E.; Liversidge, G.G. Nanosizing for oral and parenteral drug delivery: A perspective on formulating poorly-water soluble compounds using wet media milling technology. Adv. Drug. Deliv. Rev. 2011, 63, 427–440. [Google Scholar] [CrossRef] [PubMed]
  21. Juhnke, M.; Weichert, R. Zerkleinerung weicher Materialien ohne Verunreinigung der Produkte durch die Mahlkörper. Chem. Ing. Tech. 2005, 77, 90–94. [Google Scholar] [CrossRef]
  22. Patravale, V.B.; Date, A.A.; Kulkarni, R.M. Nanosuspensions: A promising drug delivery strategy. J. Pharm. Pharmacol. 2004, 56, 827–840. [Google Scholar] [CrossRef] [PubMed]
  23. Burmeister, C.F.; Kwade, A. Process engineering with planetary ball mills. Chem. Soc. Rev. 2013, 42, 7660–7667. [Google Scholar] [CrossRef] [PubMed]
  24. Juhnke, M.; Berghausen, J.; Timpe, C. Accelerated Formulation Development for nanomilled active pharmaceutical ingredients using a screening approach. Chem. Eng. Technol. 2010, 33, 1412–1418. [Google Scholar] [CrossRef]
  25. Koehler, J.K.; Schmager, S.; Bender, V.; Steiner, D.; Massing, U. Preparation of nanosized pharmaceutical formulations by dual centrifugation. Pharmaceuticals 2023, 16, 1519. [Google Scholar] [CrossRef] [PubMed]
  26. Hagedorn, M.; Bögershausen, A.; Rischer, M.; Schubert, R.; Massing, U. Dual centrifugation—A new technique for nanomilling of poorly soluble drugs and formulation screening by an DoE-approach. Int. J. Pharm. 2017, 530, 79–88. [Google Scholar] [CrossRef] [PubMed]
  27. Hagedorn, M.; Liebich, L.; Bogershausen, A.; Massing, U.; Hoffmann, S.; Mende, S.; Rischer, M. Rapid development of API nano-formulations from screening to production combining dual centrifugation and wet agitator bead milling. Int. J. Pharm. 2019, 565, 187–198. [Google Scholar] [CrossRef] [PubMed]
  28. Steiner, D.; Finke, J.H.; Breitung-Faes, S.; Kwade, A. Breakage, temperature dependency and contamination of Lactose during ball milling in ethanol. Adv. Powder Technol. 2016, 27, 1700–1709. [Google Scholar] [CrossRef]
  29. Mahlberg, L.; Steiner, D. Patient-individual dosing of poorly water-soluble drugs—Printing drug-containing, oily inks on structured film templates. In Proceedings of the 14th World Meeting on Pharmaceutics, Biopharmaceutics and Pharmaceutical Technology, Vienna, Austria, 18–21 March 2024. Poster Presentation. [Google Scholar]
  30. da Igreja, P.; Erve, A.; Thommes, M. Melt milling as manufacturing method for solid crystalline suspensions. Eur. J. Pharm. Biopharm. 2021, 158, 245–253. [Google Scholar] [CrossRef]
  31. da Igreja, P.; Klump, D.; Bartsch, J.; Thommes, M. Reduction of submicron particle agglomeration via melt foaming in solid crystalline suspension. J. Dispers. Sci. Technol. 2024, 45, 307–316. [Google Scholar] [CrossRef]
  32. Bitterlich, A.; Laabs, C.; Busmann, E.; Grandeury, A.; Juhnke, M.; Bunjes, H.; Kwade, A. Challenges in nanogrinding of active pharmaceutical ingredients. Chem. Eng. Technol. 2014, 37, 840–846. [Google Scholar] [CrossRef]
  33. Wu, L.; Zhang, J.; Watanabe, W. Physical and chemical stability of drug nanoparticles. Adv. Drug Deliv. Rev. 2011, 63, 456–469. [Google Scholar] [CrossRef] [PubMed]
  34. Hansen, C.M. The Three Dimensional Solubility Parameter and Solvent Diffusion Coefficient; Danish Technical Press: Vanløse, Denmark, 1967. [Google Scholar]
  35. Peppersack, C.; Flach, F.; Prziwara, P.; Damm, C.; Breitung-Faes, S.; Peukert, W.; Kwade, A. Conceptual stabilizer selection for nanomilling based on dispersibility parameters. Adv. Powder Technol. 2023, 34, 104197. [Google Scholar] [CrossRef]
  36. Bitterlich, A.; Laabs, C.; Krautstrunk, I.; Dengler, M.; Juhnke, M.; Grandeury, A.; Bunjes, H.; Kwade, A. Process parameter dependent growth phenomena of naproxen nanosuspension manufactured by wet media milling. Eur. J. Pharm. Biopharm. 2015, 92, 171–179. [Google Scholar] [CrossRef] [PubMed]
  37. Konnerth, C.; Damm, C.; Schmidt, J.; Peukert, W. Mechanical activation of trans-stilbene during wet grinding. Adv. Powder Technol. 2014, 25, 1808–1816. [Google Scholar] [CrossRef]
  38. Li, M.; Yaragudi, N.; Afolabi, A.; Dave, R.; Bilgili, E. Sub-100nm drug particle suspensions prepared via wet milling with low bead contamination through novel process intensification. Chem. Eng. Sci. 2015, 130, 207–220. [Google Scholar] [CrossRef]
  39. Flach, F.; Konnerth, C.; Peppersack, C.; Schmidt, J.; Damm, C.; Breitung-Faes, S.; Peukert, W.; Kwade, A. Impact of formulation and operating parameters on particle size and grinding media wear in wet media milling of organic compounds—A case study for pyrene. Adv. Powder Technol. 2016, 27, 2507–2519. [Google Scholar] [CrossRef]
  40. Flach, F.; Breitung-Faes, S.; Kwade, A. Grinding media wear induced agglomeration of electrosteric stabilized particles. Colloids Surf. A Physicochem. Eng. Asp. 2017, 522, 140–151. [Google Scholar] [CrossRef]
  41. Flach, F.; Breitung-Faes, S.; Kwade, A. Tailoring product formulation properties to reduce grinding media wear. Chem. Eng. Sci. 2019, 207, 69–78. [Google Scholar] [CrossRef]
  42. Zhang, X.; Hu, H. Preparation and analysis of a polyacrylate grinding aid for grinding calcium carbonate (GCC) in an ultrafine wet grinding process. Powder Technol. 2014, 254, 470–479. [Google Scholar] [CrossRef]
  43. Schonfeld, B.; Sundermann, J.; Keller, B.L.; Westedt, U.; Heinzerling, O. Transformation of ABT-199 nanocrystal suspensions into a redispersible drug product-impact of vacuum drum drying, spray drying and tableting on re-nanodispersibility. Pharmaceutics 2024, 16, 782. [Google Scholar] [CrossRef] [PubMed]
  44. Ouranidis, A.; Gkampelis, N.; Vardaka, E.; Karagianni, A.; Tsiptsios, D.; Nikolakakis, I.; Kachrimanis, K. Overcoming the Solubility Barrier of Ibuprofen by the Rational Process Design of a Nanocrystal Formulation. Pharmaceutics 2020, 12, 969. [Google Scholar] [CrossRef] [PubMed]
  45. Wei, Q.; Keck, C.M.; Muller, R.H. Solidification of hesperidin nanosuspension by spray drying optimized by design of experiment (DoE). Drug Dev. Ind. Pharm. 2018, 44, 1–12. [Google Scholar] [CrossRef] [PubMed]
  46. Kesisoglou, F.; Panmai, S.; Wu, Y. Nanosizing--oral formulation development and biopharmaceutical evaluation. Adv. Drug Deliv. Rev. 2007, 59, 631–644. [Google Scholar] [CrossRef] [PubMed]
  47. Christensen, K.L.; Pedersen, G.P.; Kristensen, H.G. Preparation of redispersible dry emulsions by spray drying. Int. J. Pharm. 2001, 212, 187–194. [Google Scholar] [CrossRef] [PubMed]
  48. Czyz, S.; Wewers, M.; Finke, J.H.; Kwade, A.; van Eerdenbrugh, B.; Juhnke, M.; Bunjes, H. Spray drying of API nanosuspensions: Importance of drying temperature, type and content of matrix former and particle size for successful formulation and process development. Eur. J. Pharm. Biopharm. 2020, 152, 63–71. [Google Scholar] [CrossRef] [PubMed]
  49. Wanning, S.; Suverkrup, R.; Lamprecht, A. Pharmaceutical spray freeze drying. Int. J. Pharm. 2015, 488, 136–153. [Google Scholar] [CrossRef] [PubMed]
  50. Touzet, A.; Pfefferle, F.; Lamprecht, A.; Pellequer, Y. Formulation of ketoconazole nanocrystal-based cryopellets. AAPS PharmSciTech 2020, 21, 50. [Google Scholar] [CrossRef] [PubMed]
  51. Niwa, T.; Danjo, K. Design of self-dispersible dry nanosuspension through wet milling and spray freeze-drying for poorly water-soluble drugs. Eur. J. Pharm. Sci. 2013, 50, 272–281. [Google Scholar] [CrossRef] [PubMed]
  52. Prajapati, H.; Serajuddin, A.T.M. Development of fully redispersible dried nanocrystals by using sucrose laurate as stabilizer for increasing surface area and dissolution rate of poorly water-soluble drugs. J. Pharm. Sci. 2022, 111, 780–793. [Google Scholar] [CrossRef] [PubMed]
  53. Azad, M.; Moreno, J.; Bilgili, E.; Dave, R. Fast dissolution of poorly water soluble drugs from fluidized bed coated nanocomposites: Impact of carrier size. Int. J. Pharm. 2016, 513, 319–331. [Google Scholar] [CrossRef] [PubMed]
  54. Wewers, M.; Czyz, S.; Finke, J.H.; John, E.; Van Eerdenbrugh, B.; Juhnke, M.; Bunjes, H.; Kwade, A. Influence of formulation parameters on redispersibility of naproxen nanoparticles from granules produced in a fluidized bed process. Pharmaceutics 2020, 12, 363. [Google Scholar] [CrossRef] [PubMed]
  55. Wewers, M.; Finke, J.H.; Czyz, S.; Van Eerdenbrugh, B.; John, E.; Buch, G.; Juhnke, M.; Bunjes, H.; Kwade, A. Evaluation of the formulation parameter-dependent redispersibility of API nanoparticles from fluid bed granules. Pharmaceutics 2022, 14, 1688. [Google Scholar] [CrossRef]
  56. Sahnen, F.; Kamps, J.P.; Langer, K. Conversion of indomethacin nanosuspensions into solid dosage forms via fluid bed granulation and compaction. Eur. J. Pharm. Biopharm. 2020, 154, 89–97. [Google Scholar] [CrossRef] [PubMed]
  57. Figueroa, C.E.; Bose, S. Spray granulation: Importance of process parameters on in vitro and in vivo behavior of dried nanosuspensions. Eur. J. Pharm. Biopharm. 2013, 85, 1046–1055. [Google Scholar] [CrossRef] [PubMed]
  58. Bose, S.; Schenck, D.; Ghosh, I.; Hollywood, A.; Maulit, E.; Ruegger, C. Application of spray granulation for conversion of a nanosuspension into a dry powder form. Eur. J. Pharm. Sci. 2012, 47, 35–43. [Google Scholar] [CrossRef] [PubMed]
  59. Krampe, R.; Visser, J.C.; Frijlink, H.W.; Breitkreutz, J.; Woerdenbag, H.J.; Preis, M. Oromucosal film preparations: Points to consider for patient centricity and manufacturing processes. Expert Opin. Drug Deliv. 2016, 13, 493–506. [Google Scholar] [CrossRef] [PubMed]
  60. Jacob, S.; Boddu, S.H.S.; Bhandare, R.; Ahmad, S.S.; Nair, A.B. Orodispersible films: Current innovations and emerging trends. Pharmaceutics 2023, 15, 2753. [Google Scholar] [CrossRef] [PubMed]
  61. Hoffmann, E.M.; Breitenbach, A.; Breitkreutz, J. Advances in orodispersible films for drug delivery. Expert Opin. Drug Deliv. 2011, 8, 299–316. [Google Scholar] [CrossRef] [PubMed]
  62. Steiner, D.; Finke, J.H.; Kwade, A. Redispersion of nanoparticle-loaded orodispersible films: Preservation of particle fineness. Chem. Ing. Tech. 2017, 89, 1034–1040. [Google Scholar] [CrossRef]
  63. Steiner, D.; Finke, J.H.; Kwade, A. Instant ODFs—Development of an intermediate, nanoparticle-based product platform for individualized medication. Eur. J. Pharm. Biopharm. 2018, 126, 149–158. [Google Scholar] [CrossRef] [PubMed]
  64. Steiner, D.; Finke, J.H.; Kwade, A. Efficient production of nanoparticle-loaded orodispersible films by process integration in a stirred media mill. Int. J. Pharm. 2016, 511, 804–813. [Google Scholar] [CrossRef] [PubMed]
  65. Karagianni, A.; Peltonen, L. Production of itraconazole nanocrystal-based polymeric film formulations for immediate drug release. Pharmaceutics 2020, 12, 960. [Google Scholar] [CrossRef] [PubMed]
  66. Van Nguyen, K.; Nguyen, H.T.; Nghiem, L.H.T.; Van Can, M.; Tran, T.H. Nanosized-loratadine embedded orodispersible films for enhanced bioavailability: Scalable preparations and characterizations. AAPS PharmSciTech 2022, 23, 78. [Google Scholar] [CrossRef] [PubMed]
  67. Steiner, D.; Tidau, M.; Finke, J.H. Embedding of poorly water-soluble drugs in orodispersible films-comparison of five formulation strategies. Pharmaceutics 2022, 15, 17. [Google Scholar] [CrossRef] [PubMed]
  68. Elele, E.; Shen, Y.; Susarla, R.; Khusid, B.; Keyvan, G.; Michniak-Kohn, B. Electrodeless electrohydrodynamic drop-on-demand encapsulation of drugs into porous polymer films for fabrication of personalized dosage units. J. Pharm. Sci. 2012, 101, 2523–2533. [Google Scholar] [CrossRef] [PubMed]
  69. Steiner, D.; Finke, J.H.; Kwade, A. SOFTs—Structured orodispersible film templates. Eur. J. Pharm. Biopharm. 2019, 137, 209–217. [Google Scholar] [CrossRef] [PubMed]
  70. Palo, M.; Kolakovic, R.; Laaksonen, T.; Maattanen, A.; Genina, N.; Salonen, J.; Peltonen, J.; Sandler, N. Fabrication of drug-loaded edible carrier substrates from nanosuspensions by flexographic printing. Int. J. Pharm. 2015, 494, 603–610. [Google Scholar] [CrossRef] [PubMed]
  71. Leung, D.H. Development of Nanosuspension Formulations Compatible with Inkjet Printing for the Convenient and Precise Dispensing of Poorly Soluble Drugs. Pharmaceutics 2022, 14, 449. [Google Scholar] [CrossRef] [PubMed]
  72. Carou-Senra, P.; Rodriguez-Pombo, L.; Awad, A.; Basit, A.W.; Alvarez-Lorenzo, C.; Goyanes, A. Inkjet Printing of Pharmaceuticals. Adv. Mater. 2024, 36, e2309164. [Google Scholar] [CrossRef] [PubMed]
  73. Ding, Y.; Zhao, T.; Fang, J.; Song, J.; Dong, H.; Liu, J.; Li, S.; Zhao, M. Recent developments in the use of nanocrystals to improve bioavailability of APIs. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2024, 16, e1958. [Google Scholar] [CrossRef] [PubMed]
  74. Shahidulla, S.M.; Miskan, R.; Sultana, S. Nanosuspensions in pharmaceutical sciences: A comprehensive review. Int. J. Health Sci. Res. 2023, 13, 332–342. [Google Scholar] [CrossRef]
  75. Ran, Q.; Wang, M.; Kuang, W.; Ouyang, J.; Han, D.; Gao, Z.; Gong, J. Advances of combinative nanocrystal preparation technology for improving the insoluble drug solubility and bioavailability. Crystals 2022, 12, 1200. [Google Scholar] [CrossRef]
  76. Schikarski, T.; Trzenschiok, H.; Avila, M.; Peukert, W. Impact of solvent properties on the precipitation of active pharmaceutical ingredients. Powder Technol. 2023, 415, 118032. [Google Scholar] [CrossRef]
  77. Melzig, S.; Finke, J.H.; Schilde, C.; Vierheller, A.; Dietzel, A.; Kwade, A. Fluid mechanics and process design of high-pressure antisolvent precipitation of fenofibrate nanoparticles using a customized microsystem. Chem. Eng. J. 2019, 371, 554–564. [Google Scholar] [CrossRef]
  78. Melzig, S.; Finke, J.H.; Schilde, C.; Kwade, A. Formation of long-term stable amorphous ibuprofen nanoparticles via antisolvent melt precipitation (AMP). Eur. J. Pharm. Biopharm. 2018, 131, 224–231. [Google Scholar] [CrossRef] [PubMed]
  79. Shegokar, R.; Müller, R.H. Nanocrystals: Industrially feasible multifunctional formulation technology for poorly soluble actives. Int. J. Pharm. 2010, 399, 129–139. [Google Scholar] [CrossRef] [PubMed]
  80. Sinha, B.; Muller, R.H.; Moschwitzer, J.P. Bottom-up approaches for preparing drug nanocrystals: Formulations and factors affecting particle size. Int. J. Pharm. 2013, 453, 126–141. [Google Scholar] [CrossRef] [PubMed]
  81. Raghava Srivalli, K.M.; Mishra, B. Drug nanocrystals: A way toward scale-up. Saudi Pharm. J. 2016, 24, 386–404. [Google Scholar] [CrossRef] [PubMed]
  82. Sekiguchi, K.; Obi, N. Studies on absorption of eutectic mixture. I. A comparison of the behavior of eutectic mixture of sulfathiazole and that of ordinary sulfathiazole in man. Chem. Pharm. Bull. 1961, 9, 866–872. [Google Scholar] [CrossRef]
  83. Baghel, S.; Cathcart, H.; O’Reilly, N.J. Polymeric amorphous solid dispersions: A review of amorphization, crystallization, stabilization, solid-state characterization, and aqueous solubilization of biopharmaceutical Classification System Class II Drugs. J. Pharm. Sci. 2016, 105, 2527–2544. [Google Scholar] [CrossRef] [PubMed]
  84. Tekade, A.R.; Yadav, J.N. A Review on solid dispersion and carriers used therein for solubility enhancement of poorly water soluble drugs. Adv. Pharm. Bull. 2020, 10, 359–369. [Google Scholar] [CrossRef] [PubMed]
  85. Tejaa, S.B.; Patil, S.P.; Shete, G.; Patel, S.; Bansal, A.K. Drug-excipient behavior in polymeric amorphous solid dispersions. J. Excip. Food Chem. 2013, 4, 70–94. [Google Scholar]
  86. Van den Mooter, G. The use of amorphous solid dispersions: A formulation strategy to overcome poor solubility and dissolution rate. Drug Discov. Today Technol. 2012, 9, e79–e85. [Google Scholar] [CrossRef] [PubMed]
  87. Luebbert, C.; Huxoll, F.; Sadowski, G. Amorphous-amorphous phase separation in API/polymer formulations. Molecules 2017, 22, 296. [Google Scholar] [CrossRef] [PubMed]
  88. Singh, A.; Van den Mooter, G. Spray drying formulation of amorphous solid dispersions. Adv. Drug Deliv. Rev. 2016, 100, 27–50. [Google Scholar] [CrossRef] [PubMed]
  89. Meiland, P.; Larsen, B.S.; Knopp, M.M.; Tho, I.; Rades, T. A new method to determine drug-polymer solubility through enthalpy of melting and mixing. Int. J. Pharm. 2022, 629, 122391. [Google Scholar] [CrossRef] [PubMed]
  90. Larsen, B.S.; Meiland, P.; Tzdaka, E.; Tho, I.; Rades, T. A unifying approach to drug-in-polymer solubility prediction: Streamlining experimental workflow and analysis. Eur. J. Pharm. Biopharm. 2024, 203, 114478. [Google Scholar] [CrossRef] [PubMed]
  91. Ziaee, A.; Albadarin, A.B.; Padrela, L.; Femmer, T.; O’Reilly, E.; Walker, G. Spray drying of pharmaceuticals and biopharmaceuticals: Critical parameters and experimental process optimization approaches. Eur. J. Pharm. Sci. 2019, 127, 300–318. [Google Scholar] [CrossRef] [PubMed]
  92. Mishra, J.; Bohr, A.; Rades, T.; Grohganz, H.; Löbmann, K. Whey proteins as stabilizers in amorphous solid dispersions. Eur. J. Pharm. Sci. 2019, 128, 144–151. [Google Scholar] [CrossRef] [PubMed]
  93. Leng, D.; Bulduk, B.; Widmer, T.; Wiborg, O.; Sanchez-Felix, M.; Löbmann, K. Protein based amorphous solid dispersion: A case study investigating different whey proteins at high drug loading. Pharm. Res. 2023, 40, 1865–1872. [Google Scholar] [CrossRef] [PubMed]
  94. Leng, D.; Bulduk, B.; Anlahr, J.; Müllers, W.; Löbmann, K. Enhanced dissolution rate of nimodipine through β-lactoglobulin based formulation. Int. J. Pharm. 2023, 635, 122693. [Google Scholar] [CrossRef] [PubMed]
  95. Zhuo, X.; Margrethe Brekstad Kjellin, M.; Schaal, Z.; Zhang, T.; Löbmann, K.; Leng, D. A comparative study between a protein based amorphous formulation and other dissolution rate enhancing approaches: A case study with rifaximin. Pharmaceutics 2022, 15, 126. [Google Scholar] [CrossRef] [PubMed]
  96. Zhuo, X.; Foderà, V.; Larsson, P.; Schaal, Z.; Bergström, C.A.S.; Löbmann, K.; Kabedev, A. Analysis of stabilization mechanisms in β-lactoglobulin-based amorphous solid dispersions by experimental and computational approaches. Eur. J. Pharm. Sci. 2024, 192, 106639. [Google Scholar] [CrossRef] [PubMed]
  97. el-Egakey, M.A.; Soliva, M.; Speiser, P. Hot extruded dosage forms. I. Technology and dissolution kinetics of polymeric matrices. Pharm. Acta Helv. 1971, 46, 31–52. [Google Scholar] [PubMed]
  98. Gottschalk, T.; Özbay, C.; Feuerbach, T.; Thommes, M. Predicting throughput and melt temperature in pharmaceutical hot melt extrusion. Pharmaceutics 2022, 14, 1757. [Google Scholar] [CrossRef] [PubMed]
  99. Ghebre-Sellassie, I.; Martin, C.E.; Zhang, F.; DiNunzio, J. Pharmaceutical Extrusion Technology; CRC Press: Boca Raton, FL, USA, 2018. [Google Scholar]
  100. Patil, H.; Tiwari, R.V.; Repka, M.A. Hot-melt extrusion: From theory to application in pharmaceutical formulation. AAPS PharmSciTech 2016, 17, 20–42. [Google Scholar] [CrossRef] [PubMed]
  101. Repka, M.A.; Bandari, S.; Kallakunta, V.R.; Vo, A.Q.; McFall, H.; Pimparade, M.B.; Bhagurkar, A.M. Melt extrusion with poorly soluble drugs—An integrated review. Int. J. Pharm. 2018, 535, 68–85. [Google Scholar] [CrossRef] [PubMed]
  102. Winck, J.; Gottschalk, T.; Thommes, M. Predicting residence time and melt temperature in pharmaceutical hot melt extrusion. Pharmaceutics 2023, 15, 1417. [Google Scholar] [CrossRef] [PubMed]
  103. Winck, J.; Daalmann, M.; Berghaus, A.; Thommes, M. In-line monitoring of solid dispersion preparation in small scale extrusion based on UV–vis spectroscopy. Pharm. Dev. Technol. 2022, 27, 1009–1015. [Google Scholar] [CrossRef] [PubMed]
  104. LaFountaine, J.S.; McGinity, J.W.; Williams, R.O. Challenges and strategies in thermal processing of amorphous solid dispersions: A review. AAPS PharmSciTech 2016, 17, 43–55. [Google Scholar] [CrossRef] [PubMed]
  105. Kasturirangan, A.; Koh, C.A.; Teja, A.S. Glass-transition temperatures in CO2 + polymer systems: Modeling and experiment. Ind. Eng. Chem. Res. 2011, 50, 158–162. [Google Scholar] [CrossRef]
  106. Kikic, I.; Vecchione, F.; Alessi, P.; Cortesi, A.; Eva, F.; Elvassore, N. Polymer plasticization using supercritical carbon dioxide:  experiment and modeling. Ind. Eng. Chem. Res. 2003, 42, 3022–3029. [Google Scholar] [CrossRef]
  107. Handge, U.A.; Altstädt, V. Viscoelastic properties of solutions of polystyrene melts and carbon dioxide: Analysis of a transient shear rheology approach. J. Rheol. 2012, 56, 743–766. [Google Scholar] [CrossRef]
  108. Kiran, E.; Sarver, J.A.; Hassler, J.C. Solubility and diffusivity of CO2 and N2 in polymers and polymer swelling, glass transition, melting, and crystallization at high pressure: A critical review and perspectives on experimental methods, data, and modeling. J. Supercrit. Fluids 2022, 185, 105378. [Google Scholar] [CrossRef]
  109. Zhang, X.; Heinonen, S.; Levänen, E. Applications of supercritical carbon dioxide in materials processing and synthesis. RSC Adv. 2014, 4, 61137–61152. [Google Scholar] [CrossRef]
  110. Verreck, G.; Decorte, A.; Heymans, K.; Adriaensen, J.; Liu, D.; Tomasko, D.; Arien, A.; Peeters, J.; Van den Mooter, G.; Brewster, M.E. Hot stage extrusion of p-amino salicylic acid with EC using CO2 as a temporary plasticizer. Int. J. Pharm. 2006, 327, 45–50. [Google Scholar] [CrossRef] [PubMed]
  111. Andrews, G.P.; Abu-Diak, O.; Kusmanto, F.; Hornsby, P.; Hui, Z.; Jones, D.S. Physicochemical characterization and drug-release properties of celecoxib hot-melt extruded glass solutions. J. Pharm. Pharmacol. 2010, 62, 1580–1590. [Google Scholar] [CrossRef] [PubMed]
  112. Nagy, Z.K.; Sauceau, M.; Nyúl, K.; Rodier, E.; Vajna, B.; Marosi, G.; Fages, J. Use of supercritical CO2-aided and conventional melt extrusion for enhancing the dissolution rate of an active pharmaceutical ingredient. Polym. Adv. Technol. 2012, 23, 909–918. [Google Scholar] [CrossRef]
  113. Lyons, J.G.; Hallinan, M.; Kennedy, J.E.; Devine, D.M.; Geever, L.M.; Blackie, P.; Higginbotham, C.L. Preparation of monolithic matrices for oral drug delivery using a supercritical fluid assisted hot melt extrusion process. Int. J. Pharm. 2007, 329, 62–71. [Google Scholar] [CrossRef] [PubMed]
  114. Verreck, G.; Decorte, A.; Li, H.; Tomasko, D.; Arien, A.; Peeters, J.; Rombaut, P.; Van den Mooter, G.; Brewster, M.E. The effect of pressurized carbon dioxide as a plasticizer and foaming agent on the hot melt extrusion process and extrudate properties of pharmaceutical polymers. J. Supercrit. Fluids 2006, 38, 383–391. [Google Scholar] [CrossRef]
  115. Verreck, G.; Decorte, A.; Heymans, K.; Adriaensen, J.; Liu, D.; Tomasko, D.L.; Arien, A.; Peeters, J.; Rombaut, P.; Van den Mooter, G.; et al. The effect of supercritical CO2 as a reversible plasticizer and foaming agent on the hot stage extrusion of itraconazole with EC 20cps. J. Supercrit. Fluids 2007, 40, 153–162. [Google Scholar] [CrossRef]
  116. Rahimi, S.K.; O’Donnell, K.; Haight, B.; Machado, A.; Martin, C.; Meng, F.; Listro, T.; Zhang, F. Supercritical-CO2 foam extrusion of hydroxypropyl methyl cellulose acetate succinate/itraconazole amorphous solid dispersions: Processing-structure-property relations. J. Pharm. Sci. 2021, 110, 1444–1456. [Google Scholar] [CrossRef] [PubMed]
  117. Almutairi, M.; Almutairy, B.; Sarabu, S.; Almotairy, A.; Ashour, E.; Bandari, S.; Batra, A.; Tewari, D.; Durig, T.; Repka, M.A. Processability of AquaSolve™ LG polymer by hot-melt extrusion: Effects of pressurized CO2 on physicomechanical properties and API stability. J. Drug Deliv. Sci. Technol. 2019, 52, 165–176. [Google Scholar] [CrossRef]
  118. Verreck, G.; Decorte, A.; Heymans, K.; Adriaensen, J.; Cleeren, D.; Jacobs, A.; Liu, D.; Tomasko, D.; Arien, A.; Peeters, J.; et al. The effect of pressurized carbon dioxide as a temporary plasticizer and foaming agent on the hot stage extrusion process and extrudate properties of solid dispersions of itraconazole with PVP-VA 64. Eur. J. Pharm. Sci. 2005, 26, 349–358. [Google Scholar] [CrossRef] [PubMed]
  119. Ashour, E.A.; Kulkarni, V.; Almutairy, B.; Park, J.B.; Shah, S.P.; Majumdar, S.; Lian, Z.; Pinto, E.; Bi, V.; Durig, T.; et al. Influence of pressurized carbon dioxide on ketoprofen-incorporated hot-melt extruded low molecular weight hydroxypropylcellulose. Drug Dev. Ind. Pharm. 2016, 42, 123–130. [Google Scholar] [CrossRef] [PubMed]
  120. Klueppelberg, J.; Handge, U.A.; Thommes, M.; Winck, J. Influence of carbon dioxide on the phase behavior of pharmaceutical drug-polymer dispersions. Macromol. Chem. Phys. 2025, 226, 2400359. [Google Scholar] [CrossRef]
  121. Council of the European Union. Council conclusions on personalised medicine for patients. Off. J. Eur. Union 2015, 431, 2–5. [Google Scholar]
  122. Nashed, N.; Lam, M.; Nokhodchi, A. A comprehensive overview of extended release oral dosage forms manufactured through hot melt extrusion and its combination with 3D printing. Int. J. Pharm. 2021, 596, 120237. [Google Scholar] [CrossRef] [PubMed]
  123. Roche, A.; Sanchez-Ballester, N.M.; Bataille, B.; Delannoy, V.; Soulairol, I. Fused deposition modelling 3D printing and solubility improvement of BCS II and IV active ingredients—A narrative review. J. Control. Release 2024, 365, 507–520. [Google Scholar] [CrossRef] [PubMed]
  124. Khalid, G.M.; Billa, N. Solid dispersion formulations by FDM 3D printing—A review. Pharmaceutics 2022, 14, 690. [Google Scholar] [CrossRef] [PubMed]
  125. Anaya, B.J.; Cerda, J.R.; D’Atri, R.M.; Yuste, I.; Luciano, F.C.; Kara, A.; Ruiz, H.K.; Ballesteros, M.P.; Serrano, D.R. Engineering of 3D printed personalized polypills for the treatment of the metabolic syndrome. Int. J. Pharm. 2023, 642, 123194. [Google Scholar] [CrossRef] [PubMed]
  126. Parulski, C.; Gresse, E.; Jennotte, O.; Felten, A.; Ziemons, E.; Lechanteur, A.; Evrard, B. Fused deposition modeling 3D printing of solid oral dosage forms containing amorphous solid dispersions: How to elucidate drug dissolution mechanisms through surface spectral analysis techniques? Int. J. Pharm. 2022, 626, 122157. [Google Scholar] [CrossRef] [PubMed]
  127. Buyukgoz, G.G.; Kossor, C.G.; Davé, R.N. Enhanced supersaturation via Fusion-Assisted Amorphization during FDM 3D printing of crystalline poorly soluble drug loaded filaments. Pharmaceutics 2021, 13, 1857. [Google Scholar] [CrossRef] [PubMed]
  128. Hoffmann, L.; Breitkreutz, J.; Quodbach, J. Investigation of the degradation and in-situ amorphization of the enantiomeric drug escitalopram oxalate during Fused Deposition Modeling (FDM) 3D printing. Eur. J. Pharm. Sci. 2023, 185, 106423. [Google Scholar] [CrossRef] [PubMed]
  129. Hoffmann, L.; Breitkreutz, J.; Quodbach, J. Fused Deposition Modeling (FDM) 3D printing of the thermo-sensitive peptidomimetic drug enalapril maleate. Pharmaceutics 2022, 14, 2411. [Google Scholar] [CrossRef] [PubMed]
  130. Ponsar, H.; Wiedey, R.; Quodbach, J. Hot-melt extrusion process fluctuations and their impact on critical quality attributes of filaments and 3D-printed dosage forms. Pharmaceutics 2020, 12, 511. [Google Scholar] [CrossRef] [PubMed]
  131. Nasereddin, J.M.; Wellner, N.; Alhijjaj, M.; Belton, P.; Qi, S. Development of a simple mechanical screening method for predicting the feedability of a pharmaceutical FDM 3D printing filament. Pharm. Res. 2018, 35, 151. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, J.; Feng, X.; Patil, H.; Tiwari, R.V.; Repka, M.A. Coupling 3D printing with hot-melt extrusion to produce controlled-release tablets. Int. J. Pharm. 2017, 519, 186–197. [Google Scholar] [CrossRef] [PubMed]
  133. Tucker, N.; Stanger, J.J.; Staiger, M.P.; Razzaq, H.; Hofman, K. The history of the science and technology of electrospinning from 1600 to 1995. J. Eng. Fibers Fabr. 2012, 7, 702. [Google Scholar] [CrossRef]
  134. Cooley, J.F. Apparatus for Electrically Dispersing Fluids. U.S. Patent US1962599A, 2 April 1902. [Google Scholar]
  135. Morton, W.J. Method of Dispersing Fluids. U.S. Patent US590500A, 29 July 1902. [Google Scholar]
  136. Formhals, A. Process and Apparatus for Preparing Artificial Threads. U.S. Patent US1975504A, 2 October 1934. [Google Scholar]
  137. Vass, P.; Szabó, E.; Domokos, A.; Hirsch, E.; Galata, D.; Farkas, B.; Démuth, B.; Andersen, S.K.; Vigh, T.; Verreck, G.; et al. Scale-up of electrospinning technology: Applications in the pharmaceutical industry. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1611. [Google Scholar] [CrossRef] [PubMed]
  138. Yu, D.-G.; Li, J.-J.; Williams, G.R.; Zhao, M. Electrospun amorphous solid dispersions of poorly water-soluble drugs: A review. J. Control. Release 2018, 292, 91–110. [Google Scholar] [CrossRef] [PubMed]
  139. Brown, T.D.; Dalton, P.D.; Hutmacher, D.W. Melt electrospinning today: An opportune time for an emerging polymer process. Prog. Polym. Sci. 2016, 56, 116–166. [Google Scholar] [CrossRef]
  140. Li, D.; Xia, Y. Electrospinning of nanofibers: Reinventing the wheel? Adv. Mater. 2004, 16, 1151–1170. [Google Scholar] [CrossRef]
  141. Xue, J.; Wu, T.; Dai, Y.; Xia, Y. Electrospinning and electrospun nanofibers: Methods, materials, and applications. Chem. Rev. 2019, 119, 5298–5415. [Google Scholar] [CrossRef] [PubMed]
  142. Balogh, A.; Farkas, B.; Faragó, K.; Farkas, A.; Wagner, I.; van Assche, I.; Verreck, G.; Nagy, Z.K.; Marosi, G. Melt-blown and electrospun drug-loaded polymer fiber mats for dissolution enhancement: A comparative study. J. Pharm. Sci. 2015, 104, 1767–1776. [Google Scholar] [CrossRef] [PubMed]
  143. Nagy, Z.K.; Balogh, A.; Drávavölgyi, G.; Ferguson, J.; Pataki, H.; Vajna, B.; Marosi, G. Solvent-free melt electrospinning for preparation of fast dissolving drug delivery system and comparison with solvent-based electrospun and melt extruded systems. J. Pharm. Sci. 2013, 102, 508–517. [Google Scholar] [CrossRef] [PubMed]
  144. Lian, H.; Meng, Z. Melt electrospinning vs. solution electrospinning: A comparative study of drug-loaded poly (ε-caprolactone) fibres. Mater. Sci. Eng. C 2017, 74, 117–123. [Google Scholar] [CrossRef] [PubMed]
  145. Xu, X.; Zhuang, X.; Chen, X.; Wang, X.; Yang, L.; Jing, X. Preparation of core-sheath composite nanofibers by emulsion electrospinning. Macromol. Rapid Commun. 2006, 27, 1637–1642. [Google Scholar] [CrossRef]
  146. Yarin, A.L. Coaxial electrospinning and emulsion electrospinning of core–shell fibers. Polym. Adv. Technol. 2011, 22, 310–317. [Google Scholar] [CrossRef]
  147. Kamali, H.; Farzadnia, P.; Movaffagh, J.; Abbaspour, M. Optimization of curcumin nanofibers as fast dissolving oral films prepared by emulsion electrospinning via central composite design. J. Drug Deliv. Sci. Technol. 2022, 75, 103714. [Google Scholar] [CrossRef]
  148. Shibata, T.; Yoshimura, N.; Kobayashi, A.; Ito, T.; Hara, K.; Tahara, K. Emulsion-electrospun polyvinyl alcohol nanofibers as a solid dispersion system to improve solubility and control the release of probucol, a poorly water-soluble drug. J. Drug Deliv. Sci. Technol. 2022, 67, 102953. [Google Scholar] [CrossRef]
  149. Yu, D.-G.; Branford-White, C.; Williams, G.R.; Bligh, S.W.A.; White, K.; Zhu, L.-M.; Chatterton, N.P. Self-assembled liposomes from amphiphilic electrospun nanofibers. Soft Matter 2011, 7, 8239. [Google Scholar] [CrossRef]
  150. Friedl, J.D.; Walther, M.; Vestweber, P.K.; Wächter, J.; Knoll, P.; Jörgensen, A.M.; Bernkop-Schnürch, A.; Windbergs, M. SEDDS-loaded mucoadhesive fiber patches for advanced oromucosal delivery of poorly soluble drugs. J. Control. Release 2022, 348, 692–705. [Google Scholar] [CrossRef] [PubMed]
  151. Ge, R.; Ji, Y.; Ding, Y.; Huang, C.; He, H.; Yu, D.-G. Electrospun self-emulsifying core-shell nanofibers for effective delivery of paclitaxel. Front. Bioeng. Biotechnol. 2023, 11, 1112338. [Google Scholar] [CrossRef] [PubMed]
  152. Yu, D.-G.; Yang, C.; Jin, M.; Williams, G.R.; Zou, H.; Wang, X.; Bligh, S.W.A. Medicated Janus fibers fabricated using a Teflon-coated side-by-side spinneret. Colloids Surf. B Biointerfaces 2016, 138, 110–116. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, M.; Ge, R.; Zhao, P.; Williams, G.R.; Yu, D.-G.; Bligh, S.A. Exploring wettability difference-driven wetting by utilizing electrospun chimeric Janus microfiber comprising cellulose acetate and polyvinylpyrrolidone. Mat. Des. 2023, 226, 111652. [Google Scholar] [CrossRef]
  154. Wang, M.; Li, D.; Li, J.; Li, S.; Chen, Z.; Yu, D.-G.; Liu, Z.; Guo, J.Z. Electrospun Janus zein–PVP nanofibers provide a two-stage controlled release of poorly water-soluble drugs. Mater. Des. 2020, 196, 109075. [Google Scholar] [CrossRef]
  155. van Duong, T.; van den Mooter, G. The role of the carrier in the formulation of pharmaceutical solid dispersions. Part I: Crystalline and semi-crystalline carriers. Expert Opin. Drug Deliv. 2016, 13, 1583–1594. [Google Scholar] [CrossRef] [PubMed]
  156. Justen, A.; Kurth, C.; Schaldach, G.; Thommes, M. Preparation of micron and submicron particles via spray drying and electrostatic precipitation. Chem. Eng. Technol. 2023, 46, 343–349. [Google Scholar] [CrossRef]
  157. Thommes, M.; Ely, D.R.; Carvajal, M.T.; Pinal, R. Improvement of the dissolution rate of poorly soluble drugs by solid crystal suspensions. Mol. Pharm. 2011, 8, 727–735. [Google Scholar] [CrossRef] [PubMed]
  158. Karimi-Jafari, M.; Padrela, L.; Walker, G.M.; Croker, D.M. Creating Cocrystals: A review of pharmaceutical cocrystal preparation routes and applications. Cryst. Growth Des. 2018, 18, 6370–6387. [Google Scholar] [CrossRef]
  159. Franco, P.; Pessolano, E.; Belvedere, R.; Petrella, A.; Marco, I. Supercritical impregnation of mesoglycan into calcium alginate aerogel for wound healing. J. Supercrit. Fluids 2020, 157, 104711. [Google Scholar] [CrossRef]
  160. Marco, I.; Reverchon, E. Starch aerogel loaded with poorly water-soluble vitamins through supercritical CO2 adsorption. Chem. Eng. Res. Des. 2017, 119, 221–230. [Google Scholar] [CrossRef]
  161. Chin, S.-F.; Jimmy, F.B.; Pang, S.-C. Fabrication of cellulose aerogel from sugarcane bagasse as drug delivery carriers. J. Phys. Sci. 2016, 27, 159–168. [Google Scholar] [CrossRef]
  162. Khalbas, A.H.; Albayati, T.M.; Ali, N.S.; Salih, I.K. Drug loading methods and kinetic release models using of mesoporous silica nanoparticles as a drug delivery system: A review. S. Afr. J. Chem. Eng. 2024, 50, 261–280. [Google Scholar] [CrossRef]
  163. Khalbas, A.H.; Albayati, T.M.; Saady, N.M.C.; Zendehboudi, S.; Salih, I.K.; Tofah, M.L. Insights into drug loading techniques with mesoporous silica nanoparticles: Optimization of operating conditions and assessment of drug stability. J. Drug Deliv. Sci. Technol. 2024, 96, 105698. [Google Scholar] [CrossRef]
  164. Zarinwall, A.; Maurer, V.; Pierick, J.; Oldhues, V.M.; Porsiel, J.C.; Finke, J.H.; Garnweitner, G. Amorphization and modified release of ibuprofen by post-synthetic and solvent-free loading into tailored silica aerogels. Drug Deliv. 2022, 29, 2086–2099. [Google Scholar] [CrossRef] [PubMed]
  165. Bugnone, C.A.; Ronchetti, S.; Manna, L.; Banchero, M. An emulsification/internal setting technique for the preparation of coated and uncoated hybrid silica/alginate aerogel beads for controlled drug delivery. J. Supercrit. Fluids 2018, 142, 1–9. [Google Scholar] [CrossRef]
  166. Kazemzadeh, P.; Sayadi, K.; Toolabi, A.; Sayadi, J.; Zeraati, M.; Chauhan, N.P.S.; Sargazi, G. Structure-property relationship for different mesoporous silica nanoparticles and its drug delivery applications: A review. Front. Chem. 2022, 10, 823785. [Google Scholar] [CrossRef] [PubMed]
  167. Trivedi, R.; Chatterjee, B.; Kalave, S.; Pandya, M. Role of fine silica as amorphous solid dispersion carriers for enhancing drug load and preventing recrystallization- A comprehensive review. Curr. Drug Deliv. 2023, 20, 694–707. [Google Scholar] [CrossRef] [PubMed]
  168. Li, C.; Zhou, K.; Chen, D.; Xu, W.; Tao, Y.; Pan, Y.; Meng, K.; Shabbir, M.A.B.; Liu, Q.; Huang, L.; et al. Solid lipid nanoparticles with enteric coating for improving stability, palatability, and oral bioavailability of enrofloxacin. Int. J. Nanomed. 2019, 14, 1619–1631. [Google Scholar] [CrossRef] [PubMed]
  169. Ulker, Z.; Erkey, C. An emerging platform for drug delivery: Aerogel based systems. J. Control. Release 2014, 177, 51–63. [Google Scholar] [CrossRef] [PubMed]
  170. García-González, C.A.; Sosnik, A.; Kalmár, J.; Marco, I.; Erkey, C.; Concheiro, A.; Alvarez-Lorenzo, C. Aerogels in drug delivery: From design to application. J. Control. Release 2021, 332, 40–63. [Google Scholar] [CrossRef] [PubMed]
  171. Singh, N.; Vinjamur, M.; Mukhopadhyay, M. In vitro release kinetics of drugs from silica aerogels loaded by different modes and conditions using supercritical CO2. J. Supercrit. Fluids 2021, 170, 105142. [Google Scholar] [CrossRef]
  172. Singh, N.; Vinjamur, M.; Mukhopadhyay, M. Influence of drug properties on loadings and release kinetics of drugs from silica aerogels loaded in supercritical CO2. J. Supercrit. Fluids 2022, 181, 105510. [Google Scholar] [CrossRef]
  173. Singh, N.; Vinjamur, M.; Mukhopadhyay, M. Insights into adsorptive drug loading on silica aerogels from supercritical CO2. Langmuir 2022, 38, 13075–13083. [Google Scholar] [CrossRef] [PubMed]
  174. Smirnova, I.; Suttiruengwong, S.; Arlt, W. Feasibility study of hydrophilic and hydrophobic silica aerogels as drug delivery systems. J. Non-Cryst. Solids 2004, 350, 54–60. [Google Scholar] [CrossRef]
  175. Smirnova, I.; Suttiruengwong, S.; Seiler, M.; Arlt, W. Dissolution rate enhancement by adsorption of poorly soluble drugs on hydrophilic silica aerogels. Pharm. Dev. Technol. 2004, 9, 443–452. [Google Scholar] [CrossRef] [PubMed]
  176. Smirnova, I.; Suttiruengwong, S.; Arlt, W. Aerogels: Tailor-made Carriers for Immediate and Prolonged Drug Release. KONA Powder Part. J. 2005, 23, 86–97. [Google Scholar] [CrossRef]
  177. Uejo, F.; Limwikrant, W.; Moribe, K.; Yamamoto, K. Dissolution improvement of fenofibrate by melting inclusion in mesoporous silica. Asian J. Pharm. Sci. 2013, 8, 329–335. [Google Scholar] [CrossRef]
  178. Baumgartner, A.; Dobaj, N.; Planinšek, O. Investigating the influence of processing conditions on dissolution and physical stability of solid dispersions with fenofibrate and mesoporous silica. Pharmaceutics 2024, 16, 575. [Google Scholar] [CrossRef] [PubMed]
  179. Limnell, T.; Santos, H.A.; Mäkilä, E.; Heikkilä, T.; Salonen, J.; Murzin, D.Y.; Kumar, N.; Laaksonen, T.; Peltonen, L.; Hirvonen, J. Drug delivery formulations of ordered and nonordered mesoporous silica: Comparison of three drug loading methods. J. Pharm. Sci. 2011, 100, 3294–3306. [Google Scholar] [CrossRef] [PubMed]
  180. Shen, S.-C.; Ng, W.K.; Hu, J.; Letchmanan, K.; Ng, J.; Tan, R.B.H. Solvent-free direct formulation of poorly-soluble drugs to amorphous solid dispersion via melt-absorption. Adv. Powder Technol. 2017, 28, 1316–1324. [Google Scholar] [CrossRef]
  181. Aerts, C.A.; Verraedt, E.; Depla, A.; Follens, L.; Froyen, L.; van Humbeeck, J.; Augustijns, P.; van den Mooter, G.; Mellaerts, R.; Martens, J.A. Potential of amorphous microporous silica for ibuprofen controlled release. Int. J. Pharm. 2010, 397, 84–91. [Google Scholar] [CrossRef] [PubMed]
  182. Zůza, D.; Šoltys, M.; Mužík, J.; Lizoňová, D.; Lhotka, M.; Ulbrich, P.; Kašpar, O.; Štěpánek, F. Silica particles with three levels of porosity for efficient melt amorphisation of drugs. Microporous Mesoporous Mater. 2019, 274, 61–69. [Google Scholar] [CrossRef]
  183. Mužík, J.; Lizoňová, D.; Zadražil, A.; Štěpánek, F. Drug amorphisation by fluid bed hot-melt impregnation of mesoporous silica carriers. Chem. Eng. J. 2020, 392, 123754. [Google Scholar] [CrossRef]
  184. Hussain, T.; Waters, L.J.; Parkes, G.M.B.; Shahzad, Y. Microwave processed solid dispersions for enhanced dissolution of gemfibrozil using non-ordered mesoporous silica. Colloids Surf. A Physicochem. Eng. Asp. 2017, 520, 428–435. [Google Scholar] [CrossRef]
  185. Ahern, R.J.; Hanrahan, J.P.; Tobin, J.M.; Ryan, K.B.; Crean, A.M. Comparison of fenofibrate-mesoporous silica drug-loading processes for enhanced drug delivery. Eur. J. Pharm. Sci. 2013, 50, 400–409. [Google Scholar] [CrossRef] [PubMed]
  186. Zolotov, S.A.; Demina, N.B.; Zolotova, A.S.; Shevlyagina, N.V.; Buzanov, G.A.; Retivov, V.M.; Kozhukhova, E.I.; Zakhoda, O.Y.; Dain, I.A.; Filatov, A.R.; et al. Development of novel darunavir amorphous solid dispersions with mesoporous carriers. Eur. J. Pharm. Sci. 2021, 159, 105700. [Google Scholar] [CrossRef] [PubMed]
  187. Baán, A.; Adriaensens, P.; Lammens, J.; Delgado Hernandez, R.; Vanden Berghe, W.; Pieters, L.; Vervaet, C.; Kiekens, F. Dry amorphisation of mangiferin, a poorly water-soluble compound, using mesoporous silica. Eur. J. Pharm. Biopharm. 2019, 141, 172–179. [Google Scholar] [CrossRef] [PubMed]
  188. Bahl, D.; Bogner, R.H. Amorphization of indomethacin by co-grinding with neusilin US2: Amorphization kinetics, physical stability and mechanism. Pharm. Res. 2006, 23, 2317–2325. [Google Scholar] [CrossRef] [PubMed]
  189. Grobelny, P.; Kazakevich, I.; Zhang, D.; Bogner, R. Amorphization of itraconazole by inorganic pharmaceutical excipients: Comparison of excipients and processing methods. Pharm. Dev. Technol. 2015, 20, 118–127. [Google Scholar] [CrossRef] [PubMed]
  190. Mellaerts, R.; Jammaer, J.A.G.; van Speybroeck, M.; Chen, H.; van Humbeeck, J.; Augustijns, P.; van den Mooter, G.; Martens, J.A. Physical state of poorly water soluble therapeutic molecules loaded into SBA-15 ordered mesoporous silica carriers: A case study with itraconazole and ibuprofen. Langmuir 2008, 24, 8651–8659. [Google Scholar] [CrossRef] [PubMed]
  191. Heikkilä, T.; Salonen, J.; Tuura, J.; Kumar, N.; Salmi, T.; Murzin, D.Y.; Hamdy, M.S.; Mul, G.; Laitinen, L.; Kaukonen, A.M.; et al. Evaluation of mesoporous TCPSi, MCM-41, SBA-15, and TUD-1 materials as API carriers for oral drug delivery. Drug Deliv. 2007, 14, 337–347. [Google Scholar] [CrossRef] [PubMed]
  192. Lovskaya, D.D.; Lebedev, A.E.; Menshutina, N.V. Aerogels as drug delivery systems: In vitro and in vivo evaluations. J. Supercrit. Fluids 2015, 106, 115–121. [Google Scholar] [CrossRef]
  193. Kiss, T.; Katona, G.; Mérai, L.; Janovák, L.; Deák, Á.; Kozma, G.; Kónya, Z.; Ambrus, R. Development of a hydrophobicity-controlled delivery system containing levodopa methyl ester hydrochloride loaded into a mesoporous silica. Pharmaceutics 2021, 13, 39. [Google Scholar] [CrossRef] [PubMed]
  194. Vraníková, B.; Niederquell, A.; Šklubalová, Z.; Kuentz, M. Relevance of the theoretical critical pore radius in mesoporous silica for fast crystallizing drugs. Int. J. Pharm. 2020, 591, 120019. [Google Scholar] [CrossRef] [PubMed]
  195. Hentzschel, C.M.; Alnaief, M.; Smirnova, I.; Sakmann, A.; Leopold, C.S. Enhancement of griseofulvin release from liquisolid compacts. Eur. J. Pharm. Biopharm. 2012, 80, 130–135. [Google Scholar] [CrossRef] [PubMed]
  196. Hentzschel, C.M.; Alnaief, M.; Smirnova, I.; Sakmann, A.; Leopold, C.S. Tableting properties of silica aerogel and other silicates. Drug Dev. Ind. Pharm. 2012, 38, 462–467. [Google Scholar] [CrossRef] [PubMed]
  197. Wang, X.; Wang, X.; Liu, L.; Bai, L.; An, H.; Zheng, L.; Yi, L. Preparation and characterization of carbon aerogel microspheres by an inverse emulsion polymerization. J. Non-Cryst. Solids 2011, 357, 793–797. [Google Scholar] [CrossRef]
  198. Edinger, M.; Bar-Shalom, D.; Sandler, N.; Rantanen, J.; Genina, N. QR encoded smart oral dosage forms by inkjet printing. Int. J. Pharm. 2018, 536, 138–145. [Google Scholar] [CrossRef] [PubMed]
  199. Katsiotis, C.S.; Tikhomirov, E.; Leliopoulos, C.; Strømme, M.; Welch, K. Development of a simple paste for 3D printing of drug formulations containing a mesoporous material loaded with a poorly water-soluble drug. Eur. J. Pharm. Biopharm. 2024, 198, 114270. [Google Scholar] [CrossRef] [PubMed]
  200. Wang, Y.; Gou, K.; Guo, X.; Ke, J.; Li, S.; Li, H. Advances in regulating physicochemical properties of mesoporous silica nanocarriers to overcome biological barriers. Acta Biomater. 2021, 123, 72–92. [Google Scholar] [CrossRef] [PubMed]
  201. Barkat, A.; Beg, S.; Panda, S.K.; Alharbi, K.S.; Rahman, M.; Ahmed, F.J. Functionalized mesoporous silica nanoparticles in anticancer therapeutics. Semin. Cancer Biol. 2021, 69, 365–375. [Google Scholar] [CrossRef] [PubMed]
  202. Zhuang, J.; Yu, Y.; Lu, R. Mesoporous silica nanoparticles as carrier to overcome bacterial drug resistant barriers. Int. J. Pharm. 2023, 631, 122529. [Google Scholar] [CrossRef] [PubMed]
  203. Charman, W.N. Lipids, lipophilic drugs, and oral drug delivery—Some emerging concepts. J. Pharm. Sci. 2000, 89, 967–978. [Google Scholar] [CrossRef] [PubMed]
  204. Lim, S.B.; Banerjee, A.; Önyüksel, H. Improvement of drug safety by the use of lipid-based nanocarriers. J. Control. Release 2012, 163, 34–45. [Google Scholar] [CrossRef] [PubMed]
  205. Pouton, C.W. Formulation of poorly water-soluble drugs for oral administration: Physicochemical and physiological issues and the lipid formulation classification system. Eur. J. Pharm. Sci. 2006, 29, 278–287. [Google Scholar] [CrossRef] [PubMed]
  206. Talegaonkar, S.; Bhattacharyya, A. Potential of lipid nanoparticles (SLNs and NLCs) in enhancing oral bioavailability of drugs with poor intestinal permeability. AAPS PharmSciTech 2019, 20, 121. [Google Scholar] [CrossRef] [PubMed]
  207. Crounse, R.G. Human Pharmacology of Griseofulvin: The Effect of Fat Intake on Gastrointestinal Absorption11From the Department of Dermatology, University of Miami Medical School, Miami, Florida. J. Investig. Dermatol. 1961, 37, 529–533. [Google Scholar] [CrossRef] [PubMed]
  208. Gangavarapu, A.; Tapia-Lopez, L.V.; Sarkar, B.; Pena-Zacarias, J.; Badruddoza, A.Z.M.; Nurunnabi, M. Lipid nanoparticles for enhancing oral bioavailability. Nanoscale 2024, 16, 18319–18338. [Google Scholar] [CrossRef] [PubMed]
  209. Feeney, O.M.; Crum, M.F.; McEvoy, C.L.; Trevaskis, N.L.; Williams, H.D.; Pouton, C.W.; Charman, W.N.; Bergström, C.A.S.; Porter, C.J.H. 50 years of oral lipid-based formulations: Provenance, progress and future perspectives. Adv. Drug Deliv. Rev. 2016, 101, 167–194. [Google Scholar] [CrossRef] [PubMed]
  210. Chen, X.Q.; Gudmundsson, O.S.; Hageman, M.J. Application of lipid-based formulations in drug discovery. J. Med. Chem. 2012, 55, 7945–7956. [Google Scholar] [CrossRef] [PubMed]
  211. Bangham, A.D.; Standish, M.M.; Watkins, J.C. Diffusion of univalent ions across the lamellae of swollen phospholipids. J. Mol. Biol. 1965, 13, 238-IN227. [Google Scholar] [CrossRef] [PubMed]
  212. Gregoriadis, G.; Ryman, B.E. Liposomes as carriers of enzymes or drugs: A new approach to the treatment of storage diseases. Biochem. J. 1971, 124, 58. [Google Scholar] [CrossRef] [PubMed]
  213. Gregoriadis, G. Drug entrapment in liposomes. FEBS Lett. 1973, 36, 292–296. [Google Scholar] [CrossRef] [PubMed]
  214. Gregoriadis, G. The carrier potential of liposomes in biology and medicine (first of two parts). N. Engl. J. Med. 1976, 295, 704–710. [Google Scholar] [CrossRef] [PubMed]
  215. Gregoriadis, G. The carrier potential of liposomes in biology and medicine (second of two parts). N. Engl. J. Med. 1976, 295, 765–770. [Google Scholar] [CrossRef] [PubMed]
  216. Lopez-Berestein, G.; Fainstein, V.; Hopfer, R.; Mehta, K.; Sullivan, M.P.; Keating, M.; Rosenblum, M.G.; Mehta, R.; Luna, M.; Hersh, E.M.; et al. Liposomal amphotericin B for the treatment of systemic fungal infections in patients with cancer: A preliminary study. J. Infect. Dis. 1985, 151, 704–710. [Google Scholar] [CrossRef] [PubMed]
  217. Gabizon, A.; Peretz, T.; Sulkes, A.; Amselem, S.; Ben-Yosef, R.; Ben-Baruch, N.; Catane, R.; Biran, S.; Barenholz, Y. Systemic administration of doxorubicin-containing liposomes in cancer patients: A phase I study. Eur. J. Cancer Clin. Oncol. 1989, 25, 1795–1803. [Google Scholar] [CrossRef] [PubMed]
  218. Davidson, R.N.; Martino, L.D.; Gradoni, L.; Giacchino, R.; Russo, R.; Gaeta, G.B.; Pempinello, R.; Scott, S.; Raimondi, F.; Cascio, A.; et al. Liposomal amphotericin B (AmBisome) in Mediterranean visceral leishmaniasis: A multi-centre trial. QJM 1994, 87, 75–81. [Google Scholar] [PubMed]
  219. James, N.D.; Coker, R.J.; Tomlinson, D.; Harris, J.R.; Gompels, M.; Pinching, A.J.; Stewart, J.S. Liposomal doxorubicin (Doxil): An effective new treatment for Kaposi’s sarcoma in AIDS. Clin. Oncol. 1994, 6, 294–296. [Google Scholar] [CrossRef] [PubMed]
  220. van Hoogevest, P.; Wendel, A. The use of natural and synthetic phospholipids as pharmaceutical excipients. Eur. J. Lipid Sci. Technol. 2014, 116, 1088–1107. [Google Scholar] [CrossRef] [PubMed]
  221. Lee, M.-K. Liposomes for enhanced bioavailability of water-insoluble drugs: In vivo evidence and recent approaches. Pharmaceutics 2020, 12, 264. [Google Scholar] [CrossRef] [PubMed]
  222. Luiz, H.; Oliveira Pinho, J.; Gaspar, M.M. Advancing medicine with lipid-nased nanosystems-The successful case of liposomes. Biomedicines 2023, 11, 435. [Google Scholar] [CrossRef] [PubMed]
  223. Riaz, M.K.; Riaz, M.A.; Zhang, X.; Lin, C.; Wong, K.H.; Chen, X.; Zhang, G.; Lu, A.; Yang, Z. Surface functionalization and targeting strategies of liposomes in solid tumor therapy: A review. Int. J. Mol. Sci. 2018, 19, 195. [Google Scholar] [CrossRef] [PubMed]
  224. Massing, U.; Cicko, S.; Ziroli, V. Dual asymmetric centrifugation (DAC)—A new technique for liposome preparation. J. Control. Release 2008, 125, 16–24. [Google Scholar] [CrossRef] [PubMed]
  225. Allen, T.M.; Cullis, P.R. Liposomal drug delivery systems: From concept to clinical applications. Adv. Drug Deliv. Rev. 2013, 65, 36–48. [Google Scholar] [CrossRef] [PubMed]
  226. Yu, J.; Zhou, S.; Li, J.; Wang, Y.; Su, Y.; Chi, D.; Wang, J.; Wang, X.; He, Z.; Lin, G.; et al. Simple weak-acid derivatives of paclitaxel for remote loading into liposomes and improved therapeutic effects. RSC Adv. 2020, 10, 27676–27687. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, X.; Zhao, F.; Feng, J.; Chen, C.; He, Z.; Sun, M.; Sun, J. Transforming a toxic non-ionizable drug into an efficacious liposome via ionizable prodrug and remote loading strategies against malignant breast tumors. Mol. Pharm. 2023, 20, 2642–2649. [Google Scholar] [CrossRef] [PubMed]
  228. Yu, J.; Wang, Y.; Zhou, S.; Li, J.; Wang, J.; Chi, D.; Wang, X.; Lin, G.; He, Z.; Wang, Y. Remote loading paclitaxel–doxorubicin prodrug into liposomes for cancer combination therapy. Acta Pharm. Sin. B 2020, 10, 1730–1740. [Google Scholar] [CrossRef] [PubMed]
  229. Moore, B.; Wilson, F.P.; Hutchinson, L. A Contribution to the Bio-Chemistry of Haemolysis:-(a) Changes in solubility of the lipoids in presence of one another, and of certain unsaturated organic substances. (b) The balancing action of certain pairs of Haemolysers in preventing Haemolysis. (c) The protective action of serum proteins against Haemolysers. (d) The effects of oxydising and reducing agents upon Haemolysis. Biochem. J. 1909, 4, 346–368. [Google Scholar] [PubMed]
  230. Martin, G.P.; Marriott, C. Membrane damage by bile salts: The protective function of phospholipids. J. Pharm. Pharmacol. 1981, 33, 754–759. [Google Scholar] [CrossRef] [PubMed]
  231. Tan, Y.; Qi, J.; Lu, Y.; Hu, F.; Yin, Z.; Wu, W. Lecithin in mixed micelles attenuates the cytotoxicity of bile salts in CaCo-2 cells. Toxicol. In Vitro 2013, 27, 714–720. [Google Scholar] [CrossRef] [PubMed]
  232. Okazaki, M.; Hara, I.; Kobayashi, T.; Hayashi, M. Effect of phosphatidyl choline on the hemolytic activities of bile salts. J. Oleo Sci. 1980, 29, 743–747. [Google Scholar] [CrossRef]
  233. Steffen, H.; Schmidt, D. Injektionslösungen. German Patent Application DE2730570C2, 19 February 1989. [Google Scholar]
  234. Alkan-Onyuksel, H.; Ramakrishnan, S.; Chai, H.B.; Pezzuto, J.M. A mixed micellar formulation suitable for the parenteral administration of taxol. Pharm. Res. 1994, 11, 206–212. [Google Scholar] [CrossRef] [PubMed]
  235. Hammad, M.A.; Müller, B.W. Increasing drug solubility by means of bile salt-phosphatidylcholine-based mixed micelles. Eur. J. Pharm. Biopharm. 1998, 46, 361–367. [Google Scholar] [CrossRef] [PubMed]
  236. Hammad, M.A.; Müller, B.W. Solubility and stability of lorazepam in bile salt/soya phosphatidylcholine-mixed micelles. Drug Dev. Ind. Pharm. 1999, 25, 409–417. [Google Scholar] [CrossRef] [PubMed]
  237. Brajtburg, J.; Elberg, S.; Travis, S.J.; Kobayashi, G.S. Treatment of murine candidiasis and cryptococcosis with amphotericin B incorporated into egg lecithin-bile salt mixed micelles. Antimicrob. Agents Chemother. 1994, 38, 294–299. [Google Scholar] [CrossRef] [PubMed]
  238. van Hoogevest, P. Review—An update on the use of oral phospholipid excipients. Eur. J. Pharm. Sci. 2017, 108, 1–12. [Google Scholar] [CrossRef] [PubMed]
  239. Schmitt, G. Safety of excipients in pediatric formulations-A call for toxicity studies in juvenile animals? Children 2015, 2, 191–197. [Google Scholar] [CrossRef] [PubMed]
  240. Naik, S.; Chougule, M.; Padhi, B.K.; Misra, A. Development of novel lyophilized mixed micelle amphotericin B formulation for treatment of systemic fungal infection. Curr. Drug Deliv. 2005, 2, 177–184. [Google Scholar] [CrossRef] [PubMed]
  241. Lv, Q.; Li, X.; Shen, B.; Xu, H.; Shen, C.; Dai, L.; Bai, J.; Yuan, H.; Han, J. Application of spray granulation for conversion of mixed phospholipid-bile salt micelles to dry powder form: Influence of drug hydrophobicity on nanoparticle reagglomeration. Int. J. Nanomed. 2014, 9, 505–515. [Google Scholar] [CrossRef] [PubMed]
  242. Lv, Q.; Shen, C.; Li, X.; Shen, B.; Yu, C.; Xu, P.; Xu, H.; Han, J.; Yuan, H. Mucoadhesive buccal films containing phospholipid-bile salts-mixed micelles as an effective carrier for Cucurbitacin B delivery. Drug Deliv. 2015, 22, 351–358. [Google Scholar] [CrossRef] [PubMed]
  243. Dong, F.; Xie, Y.; Qi, J.; Hu, F.; Lu, Y.; Li, S.; Wu, W. Bile salt/phospholipid mixed micelle precursor pellets prepared by fluid-bed coating. Int. J. Nanomed. 2013, 8, 1653–1663. [Google Scholar]
  244. Singh, Y.; Meher, J.G.; Raval, K.; Khan, F.A.; Chaurasia, M.; Jain, N.K.; Chourasia, M.K. Nanoemulsion: Concepts, development and applications in drug delivery. J. Control. Release 2017, 252, 28–49. [Google Scholar] [CrossRef] [PubMed]
  245. Berton-Carabin, C.C.; Coupland, J.N.; Elias, R.J. Effect of the lipophilicity of model ingredients on their location and reactivity in emulsions and solid lipid nanoparticles. Colloids Surf. A Physicochem. Eng. Asp. 2013, 431, 9–17. [Google Scholar] [CrossRef]
  246. Kupetz, E.; Bunjes, H. Lipid nanoparticles: Drug localization is substance-specific and achievable load depends on the size and physical state of the particles. J. Control. Release 2014, 189, 54–64. [Google Scholar] [CrossRef] [PubMed]
  247. Driscoll, D.F. Lipid injectable emulsions: Pharmacopeial and safety issues. Pharm. Res. 2006, 23, 1959–1969. [Google Scholar] [CrossRef] [PubMed]
  248. Hormann, K.; Zimmer, A. Drug delivery and drug targeting with parenteral lipid nanoemulsions—A review. J. Control. Release 2016, 223, 85–98. [Google Scholar] [CrossRef] [PubMed]
  249. Gupta, A. Nanoemulsions. In Nanoparticles for Biomedical Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 371–384. [Google Scholar]
  250. Tayeb, H.H.; Sainsbury, F. Nanoemulsions in drug delivery: Formulation to medical application. Nanomedicine 2018, 13, 2507–2525. [Google Scholar] [CrossRef] [PubMed]
  251. Schmidt, J.; Damm, C.; Romeis, S.; Peukert, W. Formation of nanoemulsions in stirred media mills. Chem. Eng. Sci. 2013, 102, 300–308. [Google Scholar] [CrossRef]
  252. Steiner, D.; Bunjes, H. Influence of process and formulation parameters on the preparation of solid lipid nanoparticles by dual centrifugation. Int. J. Pharm. X 2021, 3, 100085. [Google Scholar] [CrossRef] [PubMed]
  253. Yukuyama, M.N.; Ghisleni, D.D.; Pinto, T.J.; Bou-Chacra, N.A. Nanoemulsion: Process selection and application in cosmetics—A review. Int. J. Cosmet. Sci. 2016, 38, 13–24. [Google Scholar] [CrossRef] [PubMed]
  254. Goke, K.; Bunjes, H. Drug solubility in lipid nanocarriers: Influence of lipid matrix and available interfacial area. Int. J. Pharm. 2017, 529, 617–628. [Google Scholar] [CrossRef] [PubMed]
  255. Zhang, Y.; Shang, Z.; Gao, C.; Du, M.; Xu, S.; Song, H.; Liu, T. Nanoemulsion for solubilization, stabilization, and in vitro release of pterostilbene for oral delivery. AAPS PharmSciTech 2014, 15, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  256. Stewart, K.D.; Johnston, J.A.; Matza, L.S.; Curtis, S.E.; Havel, H.A.; Sweetana, S.A.; Gelhorn, H.L. Preference for pharmaceutical formulation and treatment process attributes. Patient Prefer. Adherence 2016, 10, 1385–1399. [Google Scholar] [CrossRef] [PubMed]
  257. Singh, K.K.; Vingkar, S.K. Formulation, antimalarial activity and biodistribution of oral lipid nanoemulsion of primaquine. Int. J. Pharm. 2008, 347, 136–143. [Google Scholar] [CrossRef] [PubMed]
  258. Wang, X.; Jiang, Y.; Wang, Y.W.; Huang, M.T.; Ho, C.T.; Huang, Q. Enhancing anti-inflammation activity of curcumin through O/W nanoemulsions. Food Chem. 2008, 108, 419–424. [Google Scholar] [CrossRef] [PubMed]
  259. Tiwari, S.B.; Amiji, M.M. Improved oral delivery of paclitaxel following administration in nanoemulsion formulations. J. Nanosci. Nanotechnol. 2006, 6, 3215–3221. [Google Scholar] [CrossRef] [PubMed]
  260. Ma, Y.; Li, H.; Guan, S. Enhancement of the oral bioavailability of breviscapine by nanoemulsions drug delivery system. Drug Dev. Ind. Pharm. 2015, 41, 177–182. [Google Scholar] [CrossRef] [PubMed]
  261. Yu, H.; Huang, Q. Improving the oral bioavailability of curcumin using novel organogel-based nanoemulsions. J. Agric. Food Chem. 2012, 60, 5373–5379. [Google Scholar] [CrossRef] [PubMed]
  262. Choi, Y.K.; Poudel, B.K.; Marasini, N.; Yang, K.Y.; Kim, J.W.; Kim, J.O.; Choi, H.-G.; Yong, C.S. Enhanced solubility and oral bioavailability of itraconazole by combining membrane emulsification and spray drying technique. Int. J. Pharm. 2012, 434, 264–271. [Google Scholar] [CrossRef] [PubMed]
  263. Dollo, G.; Le Corre, P.; Guerin, A.; Chevanne, F.; Burgot, J.L.; Leverge, R. Spray-dried redispersible oil-in-water emulsion to improve oral bioavailability of poorly soluble drugs. Eur. J. Pharm. Sci. 2003, 19, 273–280. [Google Scholar] [CrossRef] [PubMed]
  264. Steiner, D.; Schumann, L.V.; Bunjes, H. Processing of Lipid Nanodispersions into Solid Powders by Spray Drying. Pharmaceutics 2022, 14, 2464. [Google Scholar] [CrossRef] [PubMed]
  265. Groves, M.J.; Mustafa, R.M.A. Measurement of the ‘spontaneity’ of self-emulsifiable oils. J. Pharm. Pharmacol. 1974, 26, 671–681. [Google Scholar] [CrossRef] [PubMed]
  266. Pouton, C.W. Self-emulsifying drug delivery systems: Assessment of the efficiency of emulsification. Int. J. Pharm. 1985, 27, 335–348. [Google Scholar] [CrossRef]
  267. Gershanik, T.; Benita, S. Self-dispersing lipid formulations for improving oral absorption of lipophilic drugs. Eur. J. Pharm. Biopharm. 2000, 50, 179–188. [Google Scholar] [CrossRef] [PubMed]
  268. Grune, L.; Bunjes, H. Self-dispersing formulations for the delivery of poorly soluble drugs—Miscibility of phosphatidylcholines with oils and fats. Eur. J. Pharm. Biopharm. 2020, 151, 209–219. [Google Scholar] [CrossRef] [PubMed]
  269. Grune, L.; Bunjes, H. Solubility of poorly soluble drugs in phosphatidylcholine-based drug delivery systems: Comparison of the loading capacity in the bulk formulation and its dispersed state. Pharmaceuticals 2024, 17, 400. [Google Scholar] [CrossRef] [PubMed]
  270. Froelich, A.; Osmalek, T.; Jadach, B.; Puri, V.; Michniak-Kohn, B. Microemulsion-based media in nose-to-brain drug delivery. Pharmaceutics 2021, 13, 201. [Google Scholar] [CrossRef] [PubMed]
  271. Kuentz, M. Lipid-based formulations for oral delivery of lipophilic drugs. Drug Discov. Today Technol. 2012, 9, e97–e104. [Google Scholar] [CrossRef] [PubMed]
  272. Xiao, L.; Yi, T.; Liu, Y.; Huan, D.; He, J.K. Preparation and characterization of self-microemulsifying oral fast dissolving films of total ginkgo flavonoid. Chin. Tradit. Herb. Drugs 2011, 42, 1517–1522. [Google Scholar]
  273. Xiao, L.; Yi, T.; Liu, Y. A new self-microemulsifying mouth dissolving film to improve the oral bioavailability of poorly water soluble drugs. Drug Dev. Ind. Pharm. 2013, 39, 1284–1290. [Google Scholar] [CrossRef] [PubMed]
  274. Talekar, S.D.; Haware, R.V.; Dave, R.H. Evaluation of self-nanoemulsifying drug delivery systems using multivariate methods to optimize permeability of captopril oral films. Eur. J. Pharm. Sci. 2019, 130, 215–224. [Google Scholar] [CrossRef] [PubMed]
  275. Abdalla, A.; Mader, K. Preparation and characterization of a self-emulsifying pellet formulation. Eur. J. Pharm. Biopharm. 2007, 66, 220–226. [Google Scholar] [CrossRef] [PubMed]
  276. Vithani, K.; Goyanes, A.; Jannin, V.; Basit, A.W.; Gaisford, S.; Boyd, B.J. A proof of concept for 3D printing of solid lipid-based formulations of poorly water-soluble drugs to control formulation dispersion kinetics. Pharm. Res. 2019, 36, 102. [Google Scholar] [CrossRef] [PubMed]
  277. Barber, B.W.; Dumont, C.; Caisse, P.; Simon, G.P.; Boyd, B.J. A 3D-printed polymer-lipid-hybrid tablet towards the development of bespoke SMEDDS formulations. Pharmaceutics 2021, 13, 2107. [Google Scholar] [CrossRef] [PubMed]
  278. Johannesson, J.; Khan, J.; Hubert, M.; Teleki, A.; Bergstrom, C.A.S. 3D-printing of solid lipid tablets from emulsion gels. Int. J. Pharm. 2021, 597, 120304. [Google Scholar] [CrossRef] [PubMed]
  279. Meirinho, S.; Rodrigues, M.; Santos, A.O.; Falcao, A.; Alves, G. Self-emulsifying drug delivery systems: An alternative approach to improve brain bioavailability of poorly water-soluble drugs through intranasal administration. Pharmaceutics 2022, 14, 1487. [Google Scholar] [CrossRef] [PubMed]
  280. Lucks, J.S.; Müller, R.H.; König, B. Solid lipid nanoparticles (SLN)—An alternative parenteral drug delivery system. Eur. J. Pharm. Biopharm. 1992, 38, 33. [Google Scholar]
  281. Kumar, R.; Singh, A.; Sharma, K.; Dhasmana, D.; Garg, N.; Siril, P.F. Preparation, characterization and in vitro cytotoxicity of Fenofibrate and Nabumetone loaded solid lipid nanoparticles. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 106, 110184. [Google Scholar] [CrossRef] [PubMed]
  282. Silva, A.C.; Gonzalez-Mira, E.; Garcia, M.L.; Egea, M.A.; Fonseca, J.; Silva, R.; Santos, D.; Souto, E.B.; Ferreira, D. Preparation, characterization and biocompatibility studies on risperidone-loaded solid lipid nanoparticles (SLN): High pressure homogenization versus ultrasound. Colloids Surf. B. Biointerfaces 2011, 86, 158–165. [Google Scholar] [CrossRef] [PubMed]
  283. Bunjes, H.; Steiniger, F.; Richter, W. Visualizing the structure of triglyceride nanoparticles in different crystal modifications. Langmuir 2007, 23, 4005–4011. [Google Scholar] [CrossRef] [PubMed]
  284. Helgason, T.; Awad, T.S.; Kristbergsson, K.; McClements, D.J.; Weiss, J. Effect of surfactant surface coverage on formation of solid lipid nanoparticles (SLN). J. Colloid Interface Sci. 2009, 334, 75–81. [Google Scholar] [CrossRef] [PubMed]
  285. Westesen, K.; Bunjes, H.; Koch, M.H.J. Physiochemical characterization of lipid nanoparticles and evaluation of their drug loading capacity and sustained release potential. J. Control. Release 1997, 48, 223–236. [Google Scholar] [CrossRef]
  286. Müller, R.H.; Mäder, K.; Gohla, S. Solid lipid nanoparticles (SNL) for controlled drug delivery—A review of the state of the art. Eur. J. Pharm. Biopharm. 2000, 50, 161–177. [Google Scholar] [CrossRef] [PubMed]
  287. Illing, A.; Unruh, T. Investigation on the flow behavior of dispersions of solid triglyceride nanoparticles. Int. J. Pharm. 2004, 284, 123–131. [Google Scholar] [CrossRef] [PubMed]
  288. Bayon-Cordero, L.; Alkorta, I.; Arana, L. Application of solid lipid nanoparticles to improve the efficiency of anticancer drugs. Nanomaterials 2019, 9, 474. [Google Scholar] [CrossRef] [PubMed]
  289. Arana, L.; Gallego, L.; Alkorta, I. Incorporation of antibiotics into solid lipid nanoparticles: A promising approach to reduce antibiotic resistance emergence. Nanomaterials 2021, 11, 1251. [Google Scholar] [CrossRef] [PubMed]
  290. Santonocito, D.; Sarpietro, M.G.; Castelli, F.; Lauro, M.R.; Torrisi, C.; Russo, S.; Puglia, C. Development of solid lipid nanoparticles as dry powder: Characterization and formulation considerations. Molecules 2023, 28, 1545. [Google Scholar] [CrossRef] [PubMed]
  291. Gaspar, D.P.; Gaspar, M.M.; Eleuterio, C.V.; Grenha, A.; Blanco, M.; Goncalves, L.M.D.; Taboada, P.; Almeida, A.J.; Remunan-Lopez, C. Microencapsulated solid lipid nanoparticles as a hybrid platform for pulmonary antibiotic delivery. Mol. Pharm. 2017, 14, 2977–2990. [Google Scholar] [CrossRef] [PubMed]
  292. Steiner, D.; Emmendörffer, J.F.; Bunjes, H. Orodispersible Films: A Delivery Platform for Solid Lipid Nanoparticles? Pharmaceutics 2021, 13, 2162. [Google Scholar] [CrossRef] [PubMed]
  293. Jones, E.; Ojewole, E.; Kalhapure, R.; Govender, T. In vitro comparative evaluation of monolayered multipolymeric films embedded with didanosine-loaded solid lipid nanoparticles: A potential buccal drug delivery system for ARV therapy. Drug Dev. Ind. Pharm. 2014, 40, 669–679. [Google Scholar] [CrossRef] [PubMed]
  294. Tzanova, M.M.; Hagesaether, E.; Tho, I. Solid lipid nanoparticle-loaded mucoadhesive buccal films—Critical quality attributes and in vitro safety & efficacy. Int. J. Pharm. 2021, 592, 120100. [Google Scholar] [PubMed]
  295. Hirun, N.; Mahadlek, J.; Limmatvapirat, S.; Sriamornsak, P.; Yonemochi, E.; Furuishi, T.; Kraisit, P. Fabrication and characterization of pectin films containing solid lipid nanoparticles for buccal delivery of fluconazole. Int. J. Mol. Sci. 2024, 25, 5413. [Google Scholar] [CrossRef] [PubMed]
  296. Hazzah, H.A.; Farid, R.M.; Nasra, M.M.; El-Massik, M.A.; Abdallah, O.Y. Lyophilized sponges loaded with curcumin solid lipid nanoparticles for buccal delivery: Development and characterization. Int. J. Pharm. 2015, 492, 248–257. [Google Scholar] [CrossRef] [PubMed]
  297. Casadei, M.A.; Cerreto, F.; Cesa, S.; Giannuzzo, M.; Feeney, M.; Marianecci, C.; Paolicelli, P. Solid lipid nanoparticles incorporated in dextran hydrogels: A new drug delivery system for oral formulations. Int. J. Pharm. 2006, 325, 140–146. [Google Scholar] [CrossRef] [PubMed]
  298. Eugster, R.; Luciani, P. Liposomes: Bridging the gap from lab to pharmaceuticals. Curr. Opin. Colloid Interface Sci. 2025, 75, 101875. [Google Scholar] [CrossRef]
  299. Tomberg, T.; Hämäläinen, I.; Strachan, C.J.; van Veen, B. Dynamic phase behavior of amorphous solid dispersions revealed with in situ stimulated Raman scattering microscopy. Mol. Pharm. 2024, 21, 6444–6457. [Google Scholar] [CrossRef] [PubMed]
  300. Vora, L.K.; Gholap, A.D.; Jetha, K.; Thakur, R.R.S.; Solanki, H.K.; Chavda, V.P. Artificial intelligence in pharmaceutical technology and drug delivery design. Pharmaceutics 2023, 15, 1916. [Google Scholar] [CrossRef] [PubMed]
  301. Dong, J.; Zheng, W.; Huanle, X.; Defang, O. FormulationAI: A novel web-based platform for drug formulation design driven by artificial intelligence. Brief. Bioinform. 2024, 25, 1–10. [Google Scholar] [CrossRef] [PubMed]
  302. Wang, N.; Dong, J.; Ouyang, D. AI-directed formulation strategy design initiates rational drug development. J. Control. Release 2025, 378, 619–636. [Google Scholar] [CrossRef] [PubMed]
  303. Dong, J.; Gao, H.; Ouyang, D. PharmSD: A novel AI-based computational platform for solid dispersion formulation design. Int. J. Pharm. 2021, 604, 120705. [Google Scholar] [CrossRef] [PubMed]
  304. Bekker, J.; Davis, J. Learning from positive and unlabeled data: A survey. Mach. Learn. 2020, 109, 719–760. [Google Scholar] [CrossRef]
Figure 1. Simplified structure of an ASD.
Figure 1. Simplified structure of an ASD.
Pharmaceuticals 18 01089 g001
Figure 2. Different manufacturing technologies used for the preparation of ASDs, according to [85].
Figure 2. Different manufacturing technologies used for the preparation of ASDs, according to [85].
Pharmaceuticals 18 01089 g002
Figure 3. Enthalpy and volume of crystalline drugs in various states as a function of the temperature, according to [83].
Figure 3. Enthalpy and volume of crystalline drugs in various states as a function of the temperature, according to [83].
Pharmaceuticals 18 01089 g003
Figure 4. Phase diagram of a drug/polymer formulation. The Tg line is shown as a dashed black line, and the solubility line as a solid black line. The gray area denotes the amorphous–amorphous phase region. Below the solubility line, recrystallization occurs, according to [87].
Figure 4. Phase diagram of a drug/polymer formulation. The Tg line is shown as a dashed black line, and the solubility line as a solid black line. The gray area denotes the amorphous–amorphous phase region. Below the solubility line, recrystallization occurs, according to [87].
Pharmaceuticals 18 01089 g004
Figure 5. Approaches to the preparation of ASDs from filaments made from amorphous formulations or crystalline suspensions using FDM.
Figure 5. Approaches to the preparation of ASDs from filaments made from amorphous formulations or crystalline suspensions using FDM.
Pharmaceuticals 18 01089 g005
Figure 6. Different methods using electrospinning for ASD preparation.
Figure 6. Different methods using electrospinning for ASD preparation.
Pharmaceuticals 18 01089 g006
Figure 7. Exemplary illustration of drug-loaded lipid-based formulations.
Figure 7. Exemplary illustration of drug-loaded lipid-based formulations.
Pharmaceuticals 18 01089 g007
Table 1. Advantages and disadvantages of solution (SES), melt (MES), and emulsion electrospinning (EES).
Table 1. Advantages and disadvantages of solution (SES), melt (MES), and emulsion electrospinning (EES).
AdvantagesDisadvantages
SES
  • Processing of many polymers and drugs without restriction
  • Simple equipment
  • Adjustable release profiles (coaxial, triaxial, and side-by-side)
  • Loading capacity is limited by solubility
  • Compatible solvent–drug combination is necessary
  • Limited thickness of the fiber mat
MES
  • High drug-loading capacity
  • No solvent usage or residue
  • Wide temperature range
  • Well-defined architecture via melt electrowriting (size, shape, and volume)
  • Heat-sensitive substances (e.g., proteins and nucleic acids) cannot be processed
  • More complex device setup
  • Limited output
EES
  • Encapsulation of hydrophilic drugs/bioactive agents in a hydrophobic polymer matrix
  • Simple way to create core–shell structures
  • Enhanced stability of drug
  • Higher encapsulation efficiency
  • Difficult to scale-up
  • Required surfactants can cause toxicity
  • Stability problems during electrospinning
Table 2. Overview of the different technologies and their formulation principles, focusing on their potential application for poorly water-soluble BCS class II or IV drugs, as well as the benefits and drawbacks of the technologies.
Table 2. Overview of the different technologies and their formulation principles, focusing on their potential application for poorly water-soluble BCS class II or IV drugs, as well as the benefits and drawbacks of the technologies.
TechnologyPrincipleBCS ClassBenefitsDrawbacks
Nanomilling of drug particlesNanonizationII (IV)
  • Well-established technique
  • Process scale-up possible
  • Further processing into solid form established
  • Product contamination by grinding beads
  • Nanoparticle agglomeration
Precipitation of nanoparticlesNanonizationII (IV)
  • Continuous processing possible
  • Residues of solvents in product
  • Low production rate
  • Challenging scale-up
  • Further processing to solid form
Spray drying with protein carriersASD
formation
II
  • Improved stability and drug loading observed
  • New method, limited knowledge
  • Low bulk density
HMEASD
formation
II
  • Well established
  • Continuous process
  • Solvent-free
  • Extensive mixing
  • High processing temperatures
  • Powder flowability and feedability can be limiting
HME with CO2ASD
formation
II
  • Continuous process
  • Solvent-free
  • Intensified mixing
  • Moderate temperatures compared with conventional HME
  • New method, limited knowledge
  • Powder flowability and feedability can be limiting
HME coupled FDMASD
formation
II
  • In situ ASD formation possible
  • Enables individualization
  • Dissolution adjustment via geometric adjustments
  • Point-of-care manufacturing possible
  • Second heating step
  • Requires feedstock material
  • Specific feedstock mechanical properties required
ElectrospinningASD
formation
II
  • Low thermal stress (EES and SES)
  • High surface area
  • Versatility in formulation (hydrophilic/hydrophobic drugs and polymers)
  • Scale-up limitations (esp., MES)
  • High solvent consumption (EES)
  • Solvent residue concerns (EES, SES)
  • Process complexity
Mesoporous systems and aerogelsDrug amorphizationII (IV)
  • Stabilization of amorphous drugs
  • Controlled and/or targeted drug release
  • Dry and low-temperature loading processes possible
  • Carriers can negatively influence properties of, for example, tablets
  • Physically limited loading capacity
LiposomesDrug
solubilization
II, IV
  • Controlled and/or targeted drug release
  • Applicable for hydrophilic and hydrophobic drugs
  • Low loading capacity for lipophilic drugs
  • High production cost
Mixed micellesDrug
solubilization
II, IV
  • High biocompatibility and low toxicity
  • Simple production technology
  • Low drug-loading capacity
  • Less suitable for large molecules
Lipid nanoemulsionsDissolution in
lipids
II, IV
  • Suitable for highly lipophilic drugs
  • Positive impact of lipid
  • Low thermal stress
  • Limited drug-loading capacity
  • Unstable systems
Solid-lipid-based formulationsDissolution in
lipids
II, IV
  • Suitable for highly lipophilic drugs
  • Positive impact of lipid
  • Low drug-loading capacity
  • Lipids can undergo polymorphic transformations
  • Preparation at high temperatures
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Quodbach, J.; Preis, E.; Karkossa, F.; Winck, J.; Finke, J.H.; Steiner, D. Novel Strategies for the Formulation of Poorly Water-Soluble Drug Substances by Different Physical Modification Strategies with a Focus on Peroral Applications. Pharmaceuticals 2025, 18, 1089. https://doi.org/10.3390/ph18081089

AMA Style

Quodbach J, Preis E, Karkossa F, Winck J, Finke JH, Steiner D. Novel Strategies for the Formulation of Poorly Water-Soluble Drug Substances by Different Physical Modification Strategies with a Focus on Peroral Applications. Pharmaceuticals. 2025; 18(8):1089. https://doi.org/10.3390/ph18081089

Chicago/Turabian Style

Quodbach, Julian, Eduard Preis, Frank Karkossa, Judith Winck, Jan Henrik Finke, and Denise Steiner. 2025. "Novel Strategies for the Formulation of Poorly Water-Soluble Drug Substances by Different Physical Modification Strategies with a Focus on Peroral Applications" Pharmaceuticals 18, no. 8: 1089. https://doi.org/10.3390/ph18081089

APA Style

Quodbach, J., Preis, E., Karkossa, F., Winck, J., Finke, J. H., & Steiner, D. (2025). Novel Strategies for the Formulation of Poorly Water-Soluble Drug Substances by Different Physical Modification Strategies with a Focus on Peroral Applications. Pharmaceuticals, 18(8), 1089. https://doi.org/10.3390/ph18081089

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop