Next Article in Journal
Exploring Diverse Bioactive Secondary Metabolites from Marine Microorganisms Using Co-Culture Strategy
Previous Article in Journal
Synthesis and Biological Activities of Luminescent 5,6-Membered Bis(Metallacyclic) Platinum(II) Complexes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Herbal Medicine Nanocrystals: A Potential Novel Therapeutic Strategy

1
Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
2
Department of Critical Care Medicine, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
3
First Affiliated Hospital of the Medical College, Shihezi University, Shihezi 832008, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(17), 6370; https://doi.org/10.3390/molecules28176370
Submission received: 15 July 2023 / Revised: 11 August 2023 / Accepted: 25 August 2023 / Published: 31 August 2023

Abstract

:
Herbal medicines have gained recognition among physicians and patients due to their lower adverse effects compared to modern medicines. They are extensively used to treat various diseases, including cancer, cardiovascular issues, chronic inflammation, microbial contamination, diabetes, obesity, and hepatic disorders, among others. Unfortunately, the clinical application of herbal medicines is limited by their low solubility and inadequate bioavailability. Utilizing herbal medicines in the form of nanocrystals (herbal medicine nanocrystals) has shown potential in enhancing solubility and bioavailability by reducing the particle size, increasing the specific surface area, and modifying the absorption mechanisms. Multiple studies have demonstrated that these nanocrystals significantly improve drug efficacy by reducing toxicity and increasing bioavailability. This review comprehensively examines therapeutic approaches based on herbal medicine nanocrystals. It covers the preparation principles, key factors influencing nucleation and polymorphism control, applications, and limitations. The review underscores the importance of optimizing delivery systems for successful herbal medicine nanocrystal therapeutics. Furthermore, it discusses the main challenges and opportunities in developing herbal medicine nanocrystals for the purpose of treating conditions such as cancer, inflammatory diseases, cardiovascular disorders, mental and nervous diseases, and antimicrobial infections. In conclusion, we have deliberated regarding the hurdles and forthcoming outlook in the realm of nanotoxicity, in vivo kinetics, herbal ingredients as stabilizers of nanocrystals, and the potential for surmounting drug resistance through the utilization of nanocrystalline formulations in herbal medicine. We anticipate that this review will offer innovative insights into the development of herbal medicine nanocrystals as a promising and novel therapeutic strategy.

1. Introduction

Herbal medicines have multiple active ingredients that exert therapeutic properties, such as antioxidant, anti-inflammatory, antitumor, antibacterial, and antiproliferative activities, and have been widely used to treat various diseases, including cancer, cardiovascular disease, chronic inflammatory disease, microbial contamination, diabetes, obesity, and liver disease (Figure 1) [1,2]. Therefore, herbal medicines provide active molecules for clinical therapeutic drugs [3], and almost one-third of the top-selling pharmaceuticals are derived from plants or other natural sources [3,4]. A large number of active ingredients of herbal medicines have been discovered and used in the clinic with the development of separation and extraction techniques. For example, the active ingredient paclitaxel is a natural secondary metabolite isolated and purified from the bark of Taxus chinensis (Pilger) Rehd and is widely used in the treatment of breast cancer, ovarian cancer, and non-small-cell lung cancer. Curcumin, another extensively researched active ingredient, is extracted from the root of Curcuma longa L. (Zingiberaceae family). It has favorable anti-inflammatory and antitumor effectiveness, and more than three hundred clinical trials have been conducted for its use in cancer [5] and atherosclerosis cardiovascular diseases [6]. The clinical application of herbal medicines is greatly limited by their low solubility and membrane permeability, short half-life, and inadequate bioavailability. Traditionally, herbal medicines have been prepared as tablets, capsules, and decoctions; unfortunately, these strategies did not improve the bioavailability of herbal medicines. Excitingly, nanotechnology has shown great achievements in enhancing the efficacy of herbal medicines and overcoming biological barriers by improving their bioavailability [7,8].
Nanocrystals have emerged as a popular nanodrug delivery system for hydrophobic drugs since the 20th century [9]. They are nanosized pure drug particles dispersed in an aqueous medium with particle sizes ranging from 10 to 1000 nm. Differently from other nanoformulations, nanocrystals do not require a carrier, which can minimize carrier-related toxicity. At the same time, the high drug load is beneficial for scaling up and increasing patient compliance. The most important advantages of nanocrystals for enhancing oral bioavailability include (1) increased saturation solubility and dissolution rate of poorly water-soluble drugs [10]; (2) enhanced adhesion to biological membranes, which prolongs the retention time in the gastrointestinal tract [11,12]; and (3) nanocrystals that can also be absorbed through intestinal epithelial cells [13] and the lymphatic pathway [14] with intact crystalline particles. However, during the preparation and storage processes, aggregation, Ostwald ripening, and sedimentation are prone to occurring due to the high surface energy and polymorphic transition, resulting in physical instability [15,16]. To solve these problems, key preparation processes and stabilizers such as surfactants and polymers should be considered to control nucleation and crystal growth and prevent collisions and aggregation between particles [15]. The preparation techniques for nanocrystals can be categorized into three main approaches: bottom-up methods, top-down methods, and combinations thereof. Following their preparation, nanocrystals can be employed either as liquid suspensions or advanced into tablet and capsule forms through solidification processes. Presently, a diverse range of nanocrystal-based drugs has entered the market. As per the available data, the US Food and Drug Administration (FDA) has received an excess of 80 applications encompassing Abbreviated New Drug Applications (ANDA), New Drug Applications (NDA), and Investigational New Drug (IND) submissions, all related to drug products incorporating nanocrystals [17]. This versatility allows for delivery via diverse routes of administration and application across a spectrum of diseases [17].
Given that a majority of the active compounds found in herbal medicines fall under the classification of the Biopharmaceutics Classification System (BCS) II or BCS IV, nano-crystals have emerged as a prominent tool for enhancing their oral bioavailability. This review succinctly encapsulates the existing benefits of herbal medicine nanocrystals, encompassing their preparation methodologies, pivotal factors governing nucleation and polymorphism, dissolution characteristics, and pharmacokinetic attributes, as well as recent advancements across diverse disease contexts. Ultimately, we delve into the challenges and future prospects entailing nanotoxicity, in vivo fate, and the potential for herbal medicine nanocrystals to overcome drug resistance.

2. Preparation Methods of Herbal Medicine Nanocrystals

The preparation techniques for herbal medicine nanocrystals encompass bottom-up methods, top-down methods, and their amalgamation. We have synthesized these preparation methods, along with their respective merits and demerits, and they are shown in Figure 2 and Table 1.

2.1. Bottom-Up Methods

Bottom-up methods, often referred to as precipitation techniques, operate on the fundamental principle of precipitating drug nanocrystals from a solution that has reached a state of supersaturation. This process entails two distinct stages: nucleation followed by subsequent crystal growth [18]. The advantages of bottom-up methods are (1) simple to operate, (2) most cost-effective, and (3) more suitable for intravenous administration of anticancer compounds [18]. However, residual organic solvents may cause toxicity, and they are not suitable for drugs that are difficult to dissolve in organic solvents. In addition, the nucleation and crystal growth processes are difficult to control. The bottom-up methods primarily include antisolvent precipitation, supercritical fluid technology (SCF), and the evaporative precipitation into aqueous solution (EPAS) method.

2.1.1. Antisolvent Precipitation

The antisolvent precipitation method entails the amalgamation of a drug solution, typically dissolved within an organic solvent compatible with water, with an aqueous antisolvent (often comprising stabilizers). This fusion engenders a state of supersaturation within the solution. Subsequently, the drug undergoes nucleation and precipitates, giving rise to nanoscale drug crystals. Stabilizers such as surfactants and polymers used to be added to limit the growth and aggregation of crystals. Curcumin (CUR) is a hydrophobic compound categorized within the BCS IV class, and it exhibits inherent constraints concerning solubility and bioavailability. Hyeonmin Lee et al. employed the antisolvent precipitation technique to fabricate curcumin nanocrystals, designated as TA-CUR-NSP. This process involved the utilization of stabilizers such as sodium dodecyl sulfate (SDS) and tannic acid-modified polyvinylpyrrolidone/vinyl acetate (PVP/VA). The curcumin (CUR) nanocrystals were successfully generated, achieving a particle size of 127.7 ± 1.3 nm. Notably, the mucoadhesion assay revealed that TA-CUR-NSP exhibited enhanced mucus affinity across all pH conditions. Furthermore, the drug’s plasma concentration for TA-CUR-NSP demonstrated a remarkable 7.2-fold increase in comparison to the pure drug formulation [19].

2.1.2. Supercritical Fluid Technology

Supercritical fluid technology (SCF) involves the dissolution of drugs within a supercritical fluid. By pressurizing and atomizing the supercritical fluid through a nozzle with a small aperture, rapid vaporization occurs, leading to drug crystallization and the formation of drug nanocrystals. Carbon dioxide (CO2) stands as the predominant supercritical fluid choice due to its critical temperature of 31 °C and minimum critical pressure of 73.8 bar, facilitating its conversion into a supercritical state. This method is environmentally friendly, characterized by low costs, high safety, and facile removal of the supercritical fluid [20].

2.1.3. Evaporative Precipitation into Aqueous Solution Method

The evaporative precipitation into aqueous solution (EPAS) method was first developed by Sarkari et al. [21] and is now widely used to fabricate nanocrystals of poorly water-soluble drugs [22,23]. It employs rapid phase separation to initiate nucleation and foster the growth of nanocrystals for poorly soluble drugs. In essence, the drug is dissolved in an organic solvent with a low boiling temperature, such as dichloromethane. This solution is then sprayed through a fine nozzle into a heated aqueous solution. Rapid evaporation of the organic solvent occurs, leading to the drug’s precipitation alongside polymeric surfactants acting as stabilizers [21]. Artemisinin (ART) is a highly potent antimalarial medication effective against multi-drug-resistant Plasmodium falciparum malaria strains. Unfortunately, its clinical efficacy is hindered by its limited solubility. In a study by Mitali Kakran et al., an artemisinin nanocrystal was engineered through the evaporative precipitation into aqueous solution (EPAS) technique, employing PEG as a stabilizing agent. The investigation revealed that the crystalline nature of artemisinin in the nanocrystals diminished proportionally with the escalating polymer concentrations. Notably, the drug’s solubility and dissolution rate exhibited significant enhancements in comparison to the original drug powder [24].

2.2. Top-Down Methods

Top-down methods involve the fragmentation of large crystals into nanoscale particles through the application of mechanical force. The notable benefits of employing these techniques include the elimination of the requirement for organic solvents and the simplicity of scaling up production processes [25]. These primarily encompass the wet media milling (WMM) method and the high-pressure homogenization (HPH) method.

2.2.1. Wet Media Milling

In the early 1990s, Merisko Liversidge et al. introduced the wet media milling method (i.e., NanocrystalTM, Alkermes PLC, Waltham, MA, USA) to reduce the insoluble drug particle size [26]. This technology has witnessed rapid development owing to its uncomplicated preparation procedure and minimal excipient requirement. Typically, a liquid blend of the crude drug and stabilizers, in specified proportions, is introduced into a chamber filled with grinding media. These grinding media materials commonly encompass zirconia, glass, agate, or beads coated with polystyrene resin. Through high-speed rotation, interactions arise among drug particles, grinding media, and the container walls, engendering persistent and robust pressure and shear forces. These forces supply the necessary energy for the micronization of the drug particles. This method presents several advantages, including its straightforward preparation process and the attainment of a narrow particle size distribution for the resultant nanocrystals. These features render it particularly suitable for drugs that exhibit insolubility in both water and organic solvents. Moreover, its noteworthy benefits extend to various drug delivery pathways, including oral and intravenous administration. Among these, oral administration stands out, attracting the most attention due to its pronounced advantages. Denisa Lizoňová and her team successfully fabricated curcumin nanocrystals through the utilization of a wet media milling technique, yielding a particle size range of 40–90 nm. They opted for a synergistic blend of phospholipids and polyethylene glycol as stabilizers, which proved to be the most effective combination, bestowing a steric stabilization effect. This formulation resulted in colloidal stability across diverse media and exhibited remarkable anticancer potential in comparison to widely employed commercial cytostatic agents [27].

2.2.2. High-Pressure Homogenization

High-pressure homogenization (HPH) stands as the second-generation nanocrystal preparation technology, having been pioneered by Müller et al. in the 1990s, succeeding techniques such as NanopureTM (SkyePharma PLC, Saint-Quentin-Fallavier, France). This method can be categorized into microfluidization technology (e.g., Insoluble Drug Delivery Particles, IDD-PTM) and piston-gap homogenization technology (DissoCubesTM, Abbott GmbH &Co., KG, Wiesbaden, Germany).
In microfluidization technology, a drug suspension is swiftly propelled through the homogenization chamber using jet air, subjecting it to repeated directional shifts within the pipeline. This intricate process invokes effects like cavitation, impact, and shear, effectively reducing the sizes of the drug particles.
In piston-gap homogenization technology, an insoluble drug previously subjected to micronization is transformed into a crude suspension. This suspension is then propelled through a high-pressure homogenizer pump, passing through the homogenization valve gap at high speed. This process culminates in the formation of final nanocrystals. The sizes of the resultant particles can be precisely controlled by optimizing the high-pressure homogenization cycle, which often requires several iterations.
High-pressure homogenization finds extensive application in herbal medicines. For instance, Lou et al. successfully developed oridonin nanocrystals (ORI-N) through high-pressure homogenization. ORI-N displayed a particle size of 897.2 ± 14.2 nm and a zeta potential of −21.8 ± 0.8 mV. Notably, ORI-N exhibited potent inhibition against SMMC-7721 cell proliferation, leading to a higher apoptotic rate compared to the bulk ORI solution. Furthermore, ORI-N demonstrated superior antitumor efficacy, underscoring nanocrystal formation as a promising strategy for enhancing the therapeutic potential of oridonin in tumor treatment [28].

2.3. Combinative Technology

Attaining nanocrystals with exceedingly reduced particle sizes via a sole technology presents challenges. Consequently, combined techniques have been devised. These integrated approaches amalgamate the bottom-up and top-down methodologies. Typically, the precipitation method is applied to create initial coarse suspensions, followed by the milling method for precise particle size regulation and uniformity in the ultimate nanocrystals.
Prominent examples of such combinative technologies include the NanoEdge™, H69, H42, and H96 technologies. These techniques capitalize on the synergy between the strengths of both bottom-up and top-down approaches, ultimately enhancing the control and optimization of nanocrystal properties.

2.3.1. NanoEdge™

NanoEdge™ (Baxter, IL, USA), also referred to as the precipitation homogenization method, stands as one of the pioneering combined technologies for the preparation of nanocrystals. In this process, the drug undergoes both nucleation and crystal growth. The majority of crystals formed at this stage tend to be imperfect. Some may adopt an amorphous or partially crystalline state, a quality advantageous for subsequent high-pressure homogenization procedures.
A notable example is the work of Yang et al., who successfully developed a 10-hydroxycamptothecin (10-HCPT) nanocrystal. They employed a modified acid–base microprecipitation approach coupled with a high-pressure homogenization technique, resulting in nanocrystals with particle sizes of approximately 130 nm. This method showcases the potential of combining distinct approaches to tailor nanocrystal properties [29]. This nanocrystal had higher cellular uptake and antiproliferative activity than the 10-HCPT injections. It also enhanced the antitumor efficacy.

2.3.2. H69 Technology

H69 technology integrates the cavitation precipitation and high-pressure homogenization methods. A key distinction between H69 technology and NanoEdge™ lies in the timing of events. In the H69 process, precipitation and high-pressure homogenization take place almost concurrently. As drug crystals emerge during the precipitation stage, they are immediately exposed to cavitation, enduring the impact force and shear force of the homogenizer. This process serves to finely regulate crystal growth and enhance the uniformity of the resulting nanocrystals.

2.3.3. H42 Technology

H42 technology merges the principles of spray drying and high-pressure homogenization. In the H42 process, the hydrophobic drug is initially dispersed within stabilizers via a spray drying procedure. Subsequently, this drug–stabilizer mixture is dispersed into an aqueous solution, leading to the formation of a coarse suspension. This suspension then undergoes high-pressure homogenization, which regulates the particle size. While H42 technology finds extensive application in commercial production, it is worth noting that its yield tends to be low, and it may not be suitable for drugs sensitive to heat.

2.3.4. H96 Technology

H96 technology encompasses the combination of freeze drying and high-pressure homogenization. Analogous to H42 technology, the initial step involves the pretreatment of the crude drug via freeze drying. This process imparts a loose and porous structure to the drug crystals, rendering them more susceptible to further fragmentation. A noteworthy feature of this method is the ability to bypass the requirement for organic solvents while still achieving optimal nanocrystal formation under relatively mild conditions. However, it is important to acknowledge that the freeze-drying procedure does demand a considerable amount of time.

2.4. Key Factors to Control Nucleation and Polymorphism

2.4.1. Polymorphism Transformation in the Top-Down Method

Polymorphism transformation significantly affects the dissolution characteristics of nanocrystals. Amorphous nanocrystals inherently exhibit favorable dissolution rates without necessitating the overcoming of lattice energy. In the context of the “top-down” approach, milling processes are primarily propelled by factors such as cavitation forces, shear forces, and inter-particle collisions within the chamber. The mechanical energy’s pressure can disturb crystal lattices, potentially leading to the transformation of the drug crystal into an amorphous state or a metastable form [30]. The process of polymorphic transformation during mechanical milling typically involves two steps: (1) the amorphization of the impacted fraction and (2) rapid recrystallization guided by surrounding crystallites, favoring the γ or α forms [31]. Notably, Mazel et al. observed that anhydrous caffeine underwent a transformation from form I to a metastable state following grinding, eventually transitioning into form II over a period of days [32].
Milling temperature, along with the heat generated during milling, can further heighten the probability of crystal transformation. Amorphization becomes likely when the milling temperature substantially deviates from the glass transition temperature (Tg). Near Tg, recrystallization may manifest on the surface due to heightened molecular mobility, surpassing that within the bulk. Additionally, temperatures exceeding Tg tend to induce the emergence of new crystal forms [30,33].
While particle size reduction and amorphization are anticipated to enhance nanocrystal dissolution behavior, they can also introduce instability concerns. The creation of crystal surface defects and amorphization during milling leads to elevated specific surface energy in nanocrystals. Stabilizers can be introduced to embed within defect lattices and amorphous drug molecules, mitigating excessive surface energy and preserving particle stability. Consequently, optimizing preparation parameters such as milling speed, pressure, time, cycles, temperature, and stabilizer incorporation becomes pivotal to achieving optimal outcomes.

2.4.2. Nucleation and Polymorphism Transformation in the Bottom-Up Method

The “bottom-up” method engenders nanocrystals at the molecular level of the drug within a solution. This process involves the sequential occurrences of nucleation and crystal growth. The pivotal factors governing nucleation and crystal growth encompass molecular diffusion, supersaturation, surface tension, temperature [34], solvent [35] and stabilizers [36]. When an insoluble drug is mixed with an antisolvent solvent, a high metastable state situated at a supersaturation level is achieved [37,38]. This condition leads to swift nucleation bursts characterized by gradual molecular diffusion and pronounced interfacial tension. The abundance of nuclei produces results in a heightened surface area, accompanied by elevated free energy. To minimize the total surface energy, particles tend to amalgamate through flocculation aggregation and crystal growth. However, this process introduces impurities, demands high energy consumption, and carries the potential for polymorphic transformation [39]. The meticulous optimization of organic solvents and stabilizers is crucial to controlling nucleation and crystal growth. Stabilizers, including polymers and surfactants, have the capacity to attach to the surfaces of nuclei, restraining crystal growth. These stabilizers additionally enhance stability through spatial hindrance and electrostatic repulsion. The specific type and concentration of stabilizers dictate the crystallinity and melting point. Lennart et al. demonstrated that even a minute concentration of PVP could significantly diminish the crystal growth rate and modify the crystal morphology [40].
Upon simultaneous implementation of the “top-down” and “bottom-up” methods, distinct and controlled nuclei are generated, preventing them from maturing into larger nuclei.

3. Characterization of Nanocrystals

3.1. Particle Size

The Noyes–Whitney equation (Equation (1)) establishes a direct relationship between the dissolution rate of a drug and the surface area of its particles. As particle size decreases, the dissolution rate increases. Thus, particle size stands out as a pivotal attribute for assessing nanocrystals. In a study by Wang et al., the impact of particle size on the in vitro and in vivo behavior of astilbin nanocrystals (AT-NSs) was investigated. They formulated three AT-NSs with particle sizes of 215.17 ± 4.83 nm, 496.51 ± 12.94 nm, and 803.34 ± 9.06 nm. The formulation featuring a particle size of 215.17 nm exhibited a notably enhanced dissolution rate. However, the formulation with a particle size of 496.51 nm displayed the highest bioavailability. This disparity could be attributed to larger AT-NSs being sequestered by the reticuloendothelial system, accumulating in the lung and liver [41].
In another investigation, two silybin nanocrystals were prepared, with particle sizes of 637 ± 9.4 nm (SN-A) and 132 ± 4.8 nm (SN-B). Intriguingly, SN-A, when intravenously administered, exhibited a preference for targeting the liver and spleen [42]. SN-B, on the other hand, demonstrated superior induction of PC-3 cell apoptosis compared to SN-A [43]. These findings underscore the significant influence of particle size not only on the dissolution rate, but also on the biodistribution of nanocrystals.
dC/dt = AD/d(Cs − Cx)
where dC/dt is the dissolution rate; A is the surface area of the drug particle; D is the diffusion coefficient; d is the diffusion layer thickness; Cs is the saturation solubility of the drug; and Cx is the mass concentration of the drug in the solvent.

3.2. Solid State

During the preparation of nanocrystals, the drug crystals can exist in either a crystalline or amorphous state [44]. In the case of amorphous drug nanocrystals, the need to overcome lattice energy upon drug molecule dissolution is eliminated, resulting in a heightened dissolution rate. However, the amorphous state represents a high-energy condition, potentially leading to physical and chemical stability issues [45]. Gye Hwa Shin et al. successfully created an amorphous curcumin-TPGS nanocrystal through ultrasonic homogenization to enhance curcumin’s solubility. This formulation showcased a remarkable 433-fold improvement in water solubility compared to crude curcumin [46]. In another study, Liu et al. examined the solid-state characteristics of resveratrol nanocrystals prepared using different methods, including spray drying, rotary evaporation, and quench cooling, all coupled with high-pressure homogenization. Their findings highlighted that spray-drying pretreatment marginally reduced the crystallinity and altered the morphology of resveratrol nanocrystals, ultimately leading to the smallest particle size [47].

3.3. Morphology

The morphology of nanocrystals varies based on the preparation methods or stabilizers employed. Stabilizers have been shown to influence crystal growth rates in distinct directions [48], yielding diverse morphologies, such as needle-like, spherical, flaky, cubic, and rod-like crystals. This phenomenon significantly impacts the dissolution and bioavailability of nanocrystals [11].

4. Application of Nanocrystals in Herbal Medicines

This section offers an overview of the application of herbal medicine nanocrystals for diverse diseases, including cancer, inflammatory diseases, cardiovascular diseases, mental and nervous diseases, and antimicrobial diseases. Figure 3 illustrates the chemical structures of several active ingredients found in herbal medicines.

4.1. Cancer Therapy

In recent decades, herbal medicines have garnered substantial attention due to their potent biological activities, including antiproliferation, antioxidant effects, apoptosis induction, and antitumor properties, rendering them promising agents for cancer therapy. Nonetheless, many active constituents in herbal medicines fall under the BCS II or BCS IV categories, characterized by poor water solubility and membrane permeability which hinder therapeutic efficacy. Nanocrystal technology emerges as a promising strategy to address these challenges. The application of nanocrystals in herbal medicines for cancer therapy is outlined in Table 2.
Annonaceous acetogenins (ACGs) are prominent antitumor agents extracted from seeds of the Annonaceae species [49]. They exhibit significant antitumor activity against various cancer cell lines, including A549, MCF-7, L1210, SMMC7721, HeLa, MKN-45, and HepG2 [49,50,51,52,53]. Despite their efficacy, ACGs may cause acute toxicity upon oral administration, for example, liver and kidney damage [54,55]. To enhance ACGs’ antitumor effectiveness while mitigating toxicity, Hong et al. produced an ACG nanocrystal (ACG-NSps) using an antisolvent sonoprecipitation technique. Employing mPEG2000-PCL2000 as a stabilizer, the resulting ACG-NSps exhibited particle sizes of 123.2 ± 3.54 nm and a zeta potential of −20.17 mV. This formulation demonstrated enhanced cytotoxicity against 4T1, MCF-7, and HeLa cells, superior to the free drug. Notably, ACG-NSps exhibited efficient tumor accumulation and achieved enhanced therapeutic efficacy with a mere 1/10th of the dose compared to the drug oil solution [50].
Gambogenic acid, a key component of Garcinia hanburyi, displays a broad antineoplastic spectrum across breast cancer, lung cancer, and multiple myeloma [56,57,58,59]. It holds the potential to reverse P-gp-mediated multidrug resistance and influence apoptosis through ROS-dependent activation of IRE1α/JNK [60]. To enhance the bioavailability and antitumor efficiency of gambogenic acid, Yuan et al. developed gambogenic acid nanocrystals (GNA-NSs) with PVP K30 and PEG2000 as stabilizers, resulting in particles sized at 183.7 nm with a zeta potential of −22.8 mV. The AUC0–∞ and t1/2 of GNA-NSs were increased by 2.63-fold and 1.77-fold compared with those of the reference formulation, along with elevated cytotoxicity in HepG2 cells [61].
10-Hydroxycamptothecin is an alkaloid extracted from the seeds or roots of Camptotheca acuminata Decne. It can selectively inhibit topoisomerase I, interfering with DNA replication [62], and is widely exploited in clinical practice [63]. Although it has poor solubility in water, it can easily dissolve in a basic solution and precipitate after adding an acidic solution. Utilizing a modified acid–base microprecipitation technique coupled with high-pressure homogenization, Yang et al. created a 10-hydroxycamptothecin nanocrystal free of organic solvents. With a spherical particle size of around 130 nm, this nanocrystal demonstrated improved tumor accumulation and anticancer efficacy compared to 10-hydroxycamptothecin injection [29]. Zhou et al. compared the anticancer activities of hydroxycamptothecin nanocrystals with different shapes (nanorods and nanoparticles). They proved that nanorods showed higher and longer-term toxicity than nanoparticles [64], indicating that particle shape is also an important characteristic for nanocrystals.
Resveratrol, an extract from grapes, has gained attention as a potential antitumor drug and chemopreventive agent [65] due to its cancer initiation, promotion, and progression effects [65]. It was reported that resveratrol could inhibit cell proliferation by blocking PI3k/Akt/mTOR [66]. It could also downregulate CD8+CD122+ Tregs to inhibit hepatocellular carcinoma. Wang et al. crafted a folate-modified resveratrol nanocrystal, enhancing cytotoxicity against A549 cells and amplifying antitumor efficacy [67]. Additionally, a recent study revealed that resveratrol nanocrystals, when compared to their non-nanocrystalline forms, demonstrated significant reductions in tumor cell proliferation, fewer blood vessels in the peritoneum, and diminished systemic toxicity (such as hepatocellular necrosis and apoptosis, hepatic fibrosis, and minor fatty changes) in Ehrlich ascites tumor-bearing mice [68].
Table 2. Summary of nanocrystal formulations used in herbal medicines for cancer therapy.
Table 2. Summary of nanocrystal formulations used in herbal medicines for cancer therapy.
Extract/CompoundsStabilizersPreparation MethodsParticle SizeBioavailabilityAdvantagesApplicationsRef.
Annonaceous acetogeninsmPEG2000–PCL2000Antisolvent sonoprecipitation123.2 ± 3.54 nm/Nanocrystal achieved much better therapeutic efficacy than the traditional dosage form (oil solution)Antitumor[50]
OridoninLecithin, HPMC, and PVPHigh pressure homogenization912.5 ± 17.6 nm/Significantly inhibited the proliferation of PC-3 cells; enhanced growth suppression; and induced apoptosis of PC-3 cells, higher antitumor efficacy, lower toxicityProstatic carcinoma[28,69]
Gambogenic acidPVPK30 and PEG2000Antisolvent precipitation183.7 nmAUC and t1/2 of GNA-NS were increased 2.63- and 1.77-fold than that of the reference formulationExhibited superior cytotoxicity compared with GNA solution toward HepG2 cellsAntitumor[61,70]
IsoliquiritigeninHPC SSL and PVP K30Wet media milling238.1 ± 4.9 nm and 354.1 ± 9.1 nm/Improved the solubility; enhanced the cytotoxicityAntitumor[71]
Oleanolic acidSucrose monolaurate and sucrose monopalmitateO/W emulsion and organic solvent evaporation methods~100 nmOral bioavailability of the oleanolic acid nanocrystal was 6–7-times higher than that of the oleanolic acid coarse suspensionNS group had significantly higher bioavailability (6- to 7-fold) than the suspension groupAnticancer[72]
SilibininLecithin and poloxamer 188High pressure homogenization641.8 ± 14.7 nm 127 ± 1.9 nm/Showed better apoptosis effect on PC-3 cellsProstatic carcinoma[43]
CelastrolP 188Antisolvent precipitation147.9 nm/Displayed a significantly enhanced tumor inhibition rate and therapeutic efficacy in comparison with that of the CSL suspension.Breast cancer[73]
10-hydroxycamptot-hecinP 188A modified acid–base microprecipitation combined with a high-pressure homogenization technique~130 nmThe AUC0–24 value was 2.81-fold, as high as the injections group. Meanwhile, the mean residence time of the 10-HCPT nanocrystal was significantly higher than that of the 10-HCPT injections group.10-HCPT nanocrystals exhibited much higher drug levels in the plasma and tissues of the test mice than the marketed 10-HCPT injections, and significantly improved the antitumor therapeutic effect.Antitumor[29,74]
QuercetinPolysorbate 80Nanoprecipitation and high-pressure homogenization393.5 nmExhibited a significant reduction in clearance rate and increase in AUC compared with the control suspension.The solubility of QT in nanocrystals was approximately 70-fold that of crude QT, and the dissolution of QT from QT-NS was increased as compared to that of the original QT powder.Antitumor[75]
Andrographolide analogue (3A.1)Chitosan derivativesAntisolvent precipitation220–270 nm/Increased solubility and pharmacological effectiveness with the induction of apoptosis; had the strongest anticancer effect compared to the drug solutionColorectal cancer[76]
ResveratrolTPGS and DSPE-PEG-FAAntisolvent precipitation100–200 nm/Higher antitumor efficacy due to reduced tumor volume and weightAntitumor[67]
CurcuminmPEG2000-DSPE and soybean lecithinAntisolvent sonoprecipitation186.33 ± 2.73 nmAfter i.v. administration, the AUC0–24 of CUR-NSps was 4.50 times that of the CUR injections; t1/2 of CUR-NSps was approximately 35.95 times that of CUR injections.
The mean residence time of CUR-NSps was 18.90-fold longer than that of CUR solution.
CUR-NSps exhibited a significantly greater AUC0–24 and prolonged MRT compared to CUR injections after i.v. administration.
Higher biodistribution in the liver, kidney, brain, and tumor for CUR-NSps compared to CUR injections.
Antitumor[77]
CurcuminTPGSTrituration followed by ultraturrax homogenization and high-pressure homogenization210.2 nmAUC0–∞ of CUR-NS was approximately 3.8-fold greater than CUR solution; the mean residence time was 11.2-fold longer.CUR-NS showed greater AUC0–∞ and prolonged MRT compared to CUR solution in rabbits after i.v. administration.Antitumor[78]

4.2. Inflammatory Diseases

Inflammation serves as a key pathological basis or complication in various disorders, including osteoarthritis, pulmonary ailments, and hepatic conditions. Herbal medicines often possess anti-inflammatory properties, making them valuable candidates for intervention. For instance, naringenin demonstrates the capacity to regulate intracellular cytokine degradation, aiding in the amelioration of acute inflammation [79]. Icariin, on the other hand, inhibits the HO-1/Nrf2 and NF-κB signaling pathways to mitigate carrageenan-induced acute inflammation [80]. Additionally, silymarin acts as a hepatoprotective agent, and is capable of mitigating liver damage induced by diet or drugs [81,82]. The summary of herbal medicine nanocrystals for inflammatory diseases was listed in Table 3.
Gera et al. reported on naringenin nanocrystals (NRG-NS) produced through antisolvent sonoprecipitation, resulting in a particle size of 117 ± 5 nm. These nanocrystals exhibited heightened ALP levels compared to NRG and demonstrated superior structural repair of the cortical and trabecular bone architecture [83]. In a separate study, NRG nanocrystals were developed via the wet media milling technique with TPGS to address postinfectious cough [84]. The NRG nanocrystal showcased enhanced antitussive effects, yielding a 3-fold and 1.6-fold reduction in cough frequency compared to the blank model and crude NRG, respectively.
Icariin, a BCS IV flavonoid glycoside, faces solubility and membrane permeability limitations that hinder oral absorption. Icaritin, the bioactive aglycone form and a major intestinal metabolite of icariin, falls under BCS II compounds, boasting better potency than icariin in terms of osteoblast differentiation and proliferation. Li et al. crafted icaritin nanocrystals (ICTN) using the antisolvent precipitation method, yielding rod-shaped particles with sizes of 216.6 ± 12.4 nm and an approximate 50% decrease in crystallinity. Bioavailability studies unveiled significantly elevated Cmax (4.7-fold higher) and AUC0–12 (2.0-fold higher), along with decreased Tmax, compared to unformulated ICT. The study on anti-osteoporosis activity further demonstrated that ICTN improved osteoblast proliferation and differentiation, whereas ICT did not elicit such stimulation [85].
Li et al. engineered tetramethylpyrazine dihydroxynaphthalenate nanocrystals (TMP-NS) for intra-articular injection to target osteoarthritis [86]. TMP-NS exhibited retention lasting five times longer in the articular cavity, elevated TMP concentrations in the joints, and more pronounced anti-osteoarthritic efficacy compared to the TMP solution.
Table 3. Summary of nanocrystal formulations used in herbal medicines for inflammatory diseases.
Table 3. Summary of nanocrystal formulations used in herbal medicines for inflammatory diseases.
Extract/CompoundsStabilizersPreparation MethodsParticle SizeBioavailabilityAdvantagesApplicationsRef.
CurcuminP188Wet media milling200–240 nm//Bronchial Asthma[87]
NaringeninPVP K90Antisolvent sonoprecipitation117 ± 5 nm/NRG-NS showed a decrease in the levels of acid phosphatase and inorganic phosphorus compared to plain NRGAnti-osteoporotic[83]
NaringeninTPGSWet media milling~182.2 nm/Nanocrystal showed an excellent antitussive effect.
The cough frequency decreased by threefold compared with the blank model, with an inhibition rate of 66.7%.
Showed a good cough-relieving effect, decreasing IL-6 and MDA, increasing SOD, and reducing lung damage
Post-infectious cough[84]
lipophilic aglycone icaritinHPMC E3Antisolvent precipitation216.6 ± 12.4 nm2.0-fold in AUC0–12 and 4.7-fold in Cmax ICTN exhibited a faster dissolution rate, significantly faster absorption.
Enhanced proliferation and differentiation activities.
Treats impotence and prevent osteoporosis[85]
Phyllanthus amarus extract1.5% PVA Nanoprecipitation method243 ± 9.7 nm/The levels of the serum enzymes and bile salt were significantly lower.Hepatic disorders[88]
SilymarinPVAAntisolvent sonoprecipitation277.3 ± 10.4 nm/Exhibited faster dissolution rate, higher drug content, and pronounced enhancement of saturation solubility.Liver disorders, for instance, acute and chronic viral hepatitis, cirrhosis, toxic hepatitis and fatty liver[89]
Tetramethylpyrazine0.2% PVP K30 and 0.8% HPMCWet media milling588 nmThe t1/2 of TMP-NS was approximately three times longer than in the TMP group.
The average retention time of TMP-NS was approximately five times longer than TMP.
After intra-articular injection in rats, NS had a longer retention time in the articular cavity, higher TMP concentrations in the joints, and greater anti-osteoarthritic efficacy than the TMP solution.Osteoarthritis[86]
AndrographolideTPGSWet media milling244.6 ± 3.0 nmThe AUC0–t and Cmax of the freeze-dried ADG-NS were threefold and twofold higher than the ADG coarse powder.
The AUC0–t of the freeze-dried ADG-NS was 54.3% higher than the freeze-dried ADG-NS without TPGS.
The AUC0–t of the freeze-dried ADG-NS (with or without TPGS) was 1.38- or 2.14-fold higher than that of the ADG dripping pills.
ADG-NS showed higher permeability and plasma exposure.
ADG-NS were more effective in reducing the rate of paw swelling and producing a greater increase in the serum levels of nitric oxide (NO), interleukin-1 (IL-1), and tumor necrosis factor-α (TNF-α), as well as an increase in superoxide dismutase activity
Anti-inflammatory[90]

4.3. Cardiovascular Diseases

Herbal medicines have a long history of utilization in addressing cardiovascular diseases, offering various benefits such as blood pressure reduction, lipid lowering, glucose regulation, myocardial protection, and anti-platelet aggregation effects [91,92,93]. To enhance the bioavailability and therapeutic efficacy of insoluble active ingredients, nanocrystals have been harnessed. For instance, honokiol is derived from the stem bark of Magnolia officinalis Rehd. Et Wils, and exhibits platelet aggregation inhibitory effects by acting as a specific collagen receptor glycoprotein VI antagonist on human platelets [94]. Additionally, it plays a role in regulating mitochondrial substrate utilization and cellular fatty acid metabolism in diabetic mouse hearts [95,96]. To improve its oral bioavailability and distribution within the cardiovascular and cerebrovascular systems, Han et al. developed honokiol nanocrystals (HK-NSps) with particle sizes of 116.2 ± 1.77 nm [97]. Using PVP and BSA as stabilizers, they achieved an approximate 3.94-fold increase in Cmax and a 2.2-fold increase in AUC0–t of HK-NSps compared to Honokiol coarse suspension after oral administration at 20 mg/kg body weight. This enhancement stemmed from improved dissolution, adhesion between nanoparticles and intestinal villi, and specific absorption pathways like lymphatic transport. Biodistribution investigations indicated heightened Cmax and AUC0–t in the liver and kidney following oral administration of HK-NSps. Intravenous administration of HK-NSps further elevated their distribution in the heart and brain, signifying the promising potential of HK-NSps in the treatment of cardio-cerebro-vascular ailments. Various herbal medicine nanocrystals for cardiovascular diseases are summarized in Table 4.

4.4. Mental and Nervous Diseases

Addressing brain disorders, including mental and nervous diseases, poses significant challenges. Neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD) involve progressive neurodegeneration in the brain. Existing drugs for these conditions often exhibit limited efficacy and substantial side effects, which are less than ideal for clinical use. Herbal medicines have promise for the treatment of neurodegenerative diseases by regulating mitochondrial dysfunction-induced cell apoptosis [103] and influencing critical signaling pathways like PI3K, NF-κB, and AMPK, which are associated with oxidative and inflammatory stress in neurons [104]. Nevertheless, due to the poor water solubility of active components in herbal medicines, their transport across the blood–brain barrier (BBB) to achieve effective concentrations is challenging. Nanocrystals present a strategy by which to increase drug accumulation in the brain by enhancing blood drug concentrations. The application of nanocrystals in mental and nervous system diseases is presented in Table 5.
Ginkgolide B (GB) is a potent anti-Parkinsonism compound known for its ability to reduce reactive oxygen species [105] and inhibit nerve cell apoptosis [106]. Given GB’s low solubility (2.5 × 10−4 mol/L), its in vivo drug concentration is often insufficient. To address this, Liu et al. developed ginkgolide B nanocrystals (GB-NCs) with particle sizes of 83.48 ± 1.77 nm. In rat models, GB-NCs exhibited elevated drug plasma levels and enhanced neuronal drug distribution (see Table 5), leading to shorter Tmax and t1/2 compared to coarse drugs. Due to its transport across the gastrointestinal tract and BBB, GB-NCs demonstrated a much longer MRT in the brain compared to plasma. This favorable trait of GB-NCs improved behavioral deficits and reduced dopaminergic deficiency, highlighting their potential as an efficient means for facilitating brain delivery of GB [107].
Alzheimer’s disease, a prevalent neurodegenerative condition among the elderly, is characterized by a high incidence rate. Stahr et al. developed hesperetin nanocrystals with varying particle sizes and examined the impact of particle size on Alzheimer’s disease treatment. Decreasing the particle size, particularly below 200 nm, led to an increased dissolution rate [108].
Table 5. Summary of nanocrystal formulations used in herbal medicines for mental and nervous system diseases.
Table 5. Summary of nanocrystal formulations used in herbal medicines for mental and nervous system diseases.
Extract/CompoundsStabilizersPreparation MethodsParticle SizeBioavailabilityAdvantagesApplicationsRef.
Quercetin/Evaporative precipitation120 nm/Increased antioxidant enzyme activitiesParkinson’s disease[109,110]
Schisantherin A0.1% HPMC E3Antisolvent precipitation∼160 nmIn plasma, SA-NC increased 7.88-fold in Cmax and 6.37-fold in AUC0–t.
In the brain, SA-NC increased by 5.47-fold in Cmax and 6.32-fold in AUC0–t.
A fast dissolution rate in vitro,
Higher concentration in plasma and brain;
higher efficiency in reversing MPTP-induced dopaminergic neuronal loss and locomotion deficiency in zebrafish, as well as the MPP+-induced damage of neuronal cell culture model;
stronger neuroprotective effect
Parkinson’s disease[111]
Ginkgolide B0.05% HPMC E5Antisolvent precipitation83.48 ± 1.77 nmIn plasma, GB-NCs increased by 13-fold in Cmax and by 5-fold in AUC0–t. GBNCs had a shorter Tmax and t1/2.
In the brain, GB-NCs increased by threefold in Cmax, and by 2.5-fold in AUC0–t. GBNC increased Tmax and delayed t1/2.
The MRT0–t of the GB-NCs in the brain were 2.11-fold longer than those of the plasma.
GB-NCs have high rates of dissolution, enhanced cellular uptake, and permeability;
higher concentrations in the plasma and brain;
remain for longer times in the brain; and possess
higher efficiency in protecting neurons against cytotoxicity induced by MPP+,
The GB-NCs can protect neurons against cytotoxicity induced by MPP+, improve behavior, reduce dopamine deficiency, and elevate dopamine metabolite levels.
Parkinson’s disease[107]
HesperetinPlantacare 2000High-pressure homogenization
and wet media milling
Between 200 and 800 nm/Increased dissolution rate and kinetic solubility, higher antioxidant capacityAlzheimer’s disease[108,112]
Curcumin didecanoate5% F68Wet media milling517 ± 9 nm3.73-fold in Cmax, 4.7-fold in AUC0–t.Higher concentration in brainAntidepressant[113]

4.5. Antimicrobial Treatment

Microbial infections are common clinical conditions, and antibiotics like penicillin, cephalosporin, tetracyclines, and carbapenems are effective in inhibiting microbial reproduction. However, the widespread use of antibiotics has led to the emergence of drug-resistant microbes in significant numbers. Herbal medicines offer unique natural advantages in treating microbial infections, particularly drug-resistant ones. Additionally, they have played a crucial role in the management of the COVID-19 pandemic [114]. The application of nanocrystals for antimicrobial agents is outlined in Table 6.
Berberine, an isoquinoline alkaloid found in Berberis aristata, is frequently employed in the treatment of bacterial diarrhea [115]. Due to its poor water solubility and limited permeability through intestinal epithelial cells, its oral bioavailability is around 5%. To enhance its solubility, Sahibzada et al. developed two berberine nanocrystals using the evaporation precipitation method (EPN) and antisolvent precipitation with a syringe pump method (APSP). Following nanocrystallization, berberine was in a semicrystalline state, with particle sizes of 90–110 nm for APSP and 65–75 nm for EPN. Both formulations exhibited significantly improved dissolution rates compared to crude berberine. Antimicrobial assays demonstrated that berberine nanocrystals had more potent antibacterial activity than unprocessed berberine [116].
Herpetrione (HPE), extracted from Herpetospermum caudigerum, has demonstrated potent antiviral properties. Guo et al. developed HPE nanocrystals (HPE NS) using a high-pressure homogenization method. The HPE NS exhibited a substantial increase in dissolution rate. After oral administration in rats, Cmax and AUC0–t increased by 2.45-fold and 2.49-fold, respectively, compared to crude drugs. Reduced Tmax and MRT suggested rapid intestinal absorption. Activity assessment of HPE NS indicated enhanced inhibition of HBsAg and HBeAg replication and expression, as well as more significant inhibitory effects on HBV-DNA [117].
While improving oral bioavailability is a significant advantage of nanocrystals for herbal medicines, not all drugs experience improved efficacy after conversion into nanocrystals. Thymol, known for its broad-spectrum antibacterial and antifungal activities and used in treating coughs and diarrhea [118], was examined by Zhou et al. They produced a caseinate-stabilized thymol nanocrystal through a pH-driven approach to enhance water solubility. X-ray diffraction analysis revealed that thymol nanocrystals were in an amorphous state. Unexpectedly, in antibacterial activity studies, the MICs and MBCs of thymol nanocrystals were higher than those of free thymol against four bacterial strains. This outcome contrasted with the conventional understanding and might be attributed to caseinate coating preventing thymol release, thereby reducing its contact with bacteria [119].
Table 6. Summary of nanocrystal formulations used in herbal medicines for antimicrobial treatment.
Table 6. Summary of nanocrystal formulations used in herbal medicines for antimicrobial treatment.
Extract/CompoundsStabilizersPreparation MethodsParticle SizeBioavailabilityAdvantagesApplicationsRef.
Berberine1% HPMC and 1% PGEvaporative precipitation of nanocrystal (EPN) and anti-solvent precipitation with a syringe pump (APSP)71.53 nm for EPN method, 102.62 nm for APSP method/BBR NPs prepared by the EPN method showed higher solubility and dissolution rate
BBR NPs produced by both APSP and EPN methods showed promising activities against gram-positive and gram-negative bacteria, as well as yeasts, with NPs prepared by the EPN method showing superior results compared to those made with the APSP method.
Antimicrobial[116]
Herpetrione0.2% SLS and 0.3% PVP K30High-pressure homogenization286 ± 1.3 nm2.45-fold in Cmax and 2.49-fold in AUC0–t, and decrease in Tmax and MRTHPE NS increased dissolution velocity markedly; was more effective in reducing the replication and expression of HBsAg and HBeAg; and had a more significant inhibitory effect on HBV-DNAAntiviral[117]
TretinoinBean lecithinAntisolvent precipitation324 nm/Improved drug permeation and UV irradiation stabilityAcne vulgaris[120]
CurcuminF127 and CTAB Wet media milling~ 150 nm/Solubility and dissolution rate of CUR were significantly enhanced
CUR NCs displayed low cytotoxicity against normal human kidney-2 cells
CUR NCs with high positive surface charge exhibited excellent antibacterial activity compared to free CUR against E. coli and S. aureus.
Antimicrobial[121]
Nigella sativa L. extract1.5% PVAEvaporative precipitation//Nanocrystals showed higher antioxidant activity than the extract
Higher biofilm inhibition activity against Escherichia coli than the extract and ciprofloxacin
Antimicrobial[122]
ThymolCaseinateA modified acid–base microprecipitation79.4 nm/No enhancement in antimicrobial activity against four common pathogenic bacteria (Salmomella enterca, Staphylococcus aureus, Escherichia coli, and Listeria monocytogenes)Antimicrobial[119]

5. Challenges and Future Perspectives

5.1. Nanotoxicity of Nanocrystals

Nanotoxicity is a critical consideration in the clinical development of nanoformulations. In general, nanocrystals can potentially reduce the toxicity of drugs by increasing systemic exposure and decreasing the required dosage [123,124]. For instance, research by Gao et al. demonstrated that the LD50 of paclitaxel nanosuspensions stabilized with TPGs was twice that of the marketed paclitaxel for injection (98.63 mg/kg vs. 41.46 mg/kg), indicating a good tolerance of paclitaxel nanosuspensions [123]. Similarly, Zahra Karami et al. studied the oral toxicity of acetaminophen nanosuspension with a repeated dose of 100 mg/kg for 28 days, and no deaths and no abnormalities in clinical signs, body weights, or necropsy findings were observed [124].
While nanosuspensions can mitigate the inherent toxicity of drug molecules, the potential nanotoxicity of the formulation itself must also be considered. Nanoparticles offer advantages such as improved cellular uptake and targeted delivery to specific tissues, like tumors, through functionalization. For oral administration, nanocrystals tend to dissolve in the gastrointestinal fluids, and the lipophilic drug molecules are absorbed by passive diffusion. The non-absorbed stabilizers are then discharged from the body. However, functionalized nanomaterials can also interact with biological systems in unforeseen ways, potentially leading to adverse effects. For example, some stabilizers used in nanocrystal formulations, such as chitosan (open the tight junction of intestinal epithelial cells) and N-acetylcysteine (cleave the disulfide bond of mucin proteins), may interfere with intestinal mucosa function, increasing the risk of intestinal bacterial infections and inflammation. In the case of intravenous administration, non-biodegradable stabilizers can accumulate in the body, causing oxidative stress, immune reactions, and irreversible damage [125,126,127]. To mitigate these risks, biodegradable stabilizers are being explored for nanocrystal formulations.
Particle size also plays a crucial role in nanotoxicity. Particles larger than 1000 nm or micrometers can be phagocytosed by specific cells, like macrophages, in the liver, spleen, lung, and Peyer’s patches [127], resulting in limited toxic risk. In contrast, nanoparticles below 100 nm can be taken up by most cells, potentially leading to cell damage or systemic effects. Nanoparticles with sizes between 100 nm and 1000 nm are easily internalized by cells, but may be challenging to clear from the body through renal excretion. Rainer H. Müller et al. proposed a nanotoxicological classification system (NCS) to differentiate potential nanotoxicological risks that warrant careful investigation. For drug nanocrystals, via oral and external administration, the dissolution process often separates the relationship between particle size and clearance. However, for intravenous injection, when the particle size of a nanocrystal is greater than 5 μm, there is a risk of clogging capillaries.
In recent years, the cytotoxicity induced by particle shape has gained attention. For instance, needle-shaped particles were found to induce transient disruption of cell membranes, although cell recovery was identified after 48 h [128]. Shape effects on physiological responses were studied by Zhang et al. using PLGA-PEG nanoparticles, revealing that needle-shaped nanoparticles caused DNA fragmentation and cell death by disrupting lysosomes and activating apoptosis pathways, which was not observed with spherical nanoparticles [129]. Shape heterogeneity is a feature of nanocrystals due to their unique crystal lattices. Drug nanocrystals can be developed into various shapes, including spherical, rod-like, flaky, and oblong [11], which may impact their toxicological profiles. Additionally, different shapes may influence the maximum tolerated doses, affecting long-term toxicity. It has been reported that PEGylated hydroxycamptothecin nanorods have a higher lethal dose than PEGylated hydroxycamptothecin nanoparticles [64]. While research on the nanotoxicity caused by different shapes of drug nanocrystals is limited, it is an aspect that should not be ignored during the development of herbal medicine nanocrystals.

5.2. In Vivo Fate of Herbal Medicine Nanocrystals

Understanding the in vivo fate of herbal medicine nanocrystals is crucial for their successful application. While the primary purpose of nanocrystal technology is to enhance drug dissolution and bioavailability, research has shown that nanocrystals can be absorbed intact through intestinal epithelial cells [11] and lymphatic transport [14], as well as through systemic circulation after intravenous administration. To track nanocrystals in vivo, innovative approaches, such as fluorescent labeled hybrid nanocrystals, have been developed. For example, ACQ probes have water-quenching sensitivity, which means that the fluorescent signal can be detected when the nanocrystals are intact, and the signal disappears when the nanocrystals are dissolved [130]. The use of aggregation-caused quenching (ACQ) fluorescent probes embedded within drug nanocrystals allows for real-time monitoring of their dissolution and fates. Studies with CUR hybrid nanocrystals have found that they are mainly captured by the mononuclear phagocytic system, accumulating in the liver and lungs, with a portion persisting in the blood for up to 48 h before accumulating in reticuloendothelial organs and tissues. The prolonged circulation may result from the surface coating of F68. Surface modification, particle size, and particle shape have all been identified as factors that can alter the in vivo fate of nanocrystals. In the case of quercetin hybrid nanocrystals (QT-HNCs) via the oral route [131], QT-HNCs can be retained in the gastrointestinal tract for 12–16 h, and then distribute in the liver, spleen, lung, and kidney with particle size-related discriminating biodistribution. At 12 h, the biodistribution of QT-HNCs in different tissues reached its maximum. In our previous study, we proposed that particle shape was also a non-ignored factor that can alter the in vivo fate of nanocrystals [12]. We found that rod-shaped nanocrystals had longer retention times in the gastrointestinal tract. To sum up, surface modification, particle size, and particle shape should be fully considered in order to optimize the biodistribution and therapeutic effects of herbal medicine nanocrystals.

5.3. Potential Advantages of Overcoming Drug Resistance

Drug resistance is a significant challenge in cancer and antimicrobial treatments. Compared with chemotherapeutic drugs, herbal medicines offer advantages in terms of reversing multidrug resistance (MDR) in tumors, regulating immunity, and reducing non-target toxicity. Qiao et al. developed a hybrid nanocrystal containing hydroxycamptothecin (HCPT, an antitumor natural ingredient) and quercetin (QUR, a natural flavonoid compound with anti-MDR and immunomodulatory effects) [132]. The hybrid nanocrystals (HQ-NPs) were quasi-spherical in morphology, which was different from the HCPT nanosuspension (spherical-like) and QUR suspension (rodlike). The molecular dynamics study showed that the integration of HCPT and QUR was attributed to the strong π–π stacking and hydrophobic interactions of the planar structures. In H22 tumor-bearing mice, HQ-NPs showed similar accumulation of HCPT in tumor sites and antitumor effects compared with the HCPT nanosuspension. However, the concentration of HCPT in HQ-NPs was only 3/5 of that in the HCPT nanosuspension, indicating that HQ-NPs had enormous potential to overcome MDR. This result may be because QUR accelerated the cellular uptake of HQ-NPs by inhibiting P-gp, although the antitumor effect of QUR was weak.
Antibiotic resistance is another urgent challenge to be solved worldwide. The emergence of antibiotic resistance is mainly due to the widespread abuse of antibiotics. It has been reported that the most promising herbal medicines to be used alone against multidrug-resistant bacteria are Piper betle, Glycyrrhiza glabra, and berberine [133]. Nanocrystals have exhibited increased antibacterial effects for these drugs compared to unprocessed ones, as described in Section 4.5. In other situations, herbal medicines in combination with antibiotics have expanded the spectrum against multidrug-resistant bacteria [133]. The underlying mechanisms may be that herbal medicines could inhibit the efflux pump; modify the bacterial cell wall and/or membrane; inhibit the cell division protein-filamenting, temperature-sensitive Z-ring; and inhibit protein synthesis and gene expression. Harsha Kathpalia et al. investigated the effect of a combination of atovaquone nanocrystals–proguanil–artesunate on antimalarial therapy. Atovaquone nanocrystals were proven to be effective alone at 1/80th the therapeutic dose. In the triple combination, a complete cure was observed with atovaquone nanosuspension in combination with proguanil at 1/80th the therapeutic dose of each and 1/5th the therapeutic dose of artesunate, indicating that nanocrystals can reduce the therapeutic dose and delay the occurrence of antibiotic resistance [134]. Even though the therapeutic dose was dramatically reduced when applying nanocrystal technology, the optimal dosing ratio for herbal medicines and antibiotics needs to be further determined.

5.4. Herbal Ingredients as Stabilizers of Nanocrystals

Due to the potential toxicity of synthetic polymers or surfactant-based stabilizers, natural sources of stabilizers have attracted increasing attention. Saponins extracted from natural plants are widely used in food and beverages as foaming agents and emulsifiers [135] due to their amphipathic structure, with hydrophobic triterpene groups and hydrophilic sugar chains. Glycyrrhizin is a natural triterpenoid saponin. Chen et al. used glycyrrhizin to stabilize andrographolide nanocrystals (AGE-NS). Under the interfacial properties and electrostatic effects of glycyrrhizin, AGE-NS could be entrapped in its network structure [136]. Li’s group prepared hesperidin nanocrystals using tea saponins as a novel multifunctional stabilizer [137]. Compared with synthetic stabilizers, tea saponins have stabilization efficiency at low concentrations. In addition, they could prevent the aggregation of nanocrystals in the freeze-drying process by acting as a cryoprotectant. In another study, the researchers used gypenosides, a tetracyclic triterpenoid compound extracted from Gynostemma pentaphyllum, to stabilize quercetin nanocrystals (QUE-NS) [138]. Compared with glycyrrhizin, tea saponins, and other synthetic stabilizers, QUE-NS stabilized with gypenosides showed the smallest particle size and a narrow size distribution (PDI < 0.1). Lentinan is a natural β–glucan with various bioactivities, such as antitumor effects. Suo et al. proved that lentinan could stabilize regorafenib nanocrystals by forming hydrogen bonds and hydrophobic interactions between the two molecules. They also found that lentinan could enhance the in vitro anticancer activity and oral bioavailability of regorafenib [139]. With the discovery of an increasing number of natural stabilizers, the dual roles of herbal active ingredients have been explored. However, whether natural stabilizers are superior to synthetic stabilizers in terms of therapeutic efficacy is not yet fully understood. Meanwhile, the toxicity of natural stabilizers needs further investigation.

6. Conclusions

In conclusion, herbal medicine’s appeal in disease therapy is rising due to its diverse pharmacological effects and multi-target capabilities. However, the poor water solubility and potential toxicity of many active herbal ingredients have hindered their clinical application. Nanocrystals offer a promising solution for delivering insoluble drugs, offering benefits such as high drug loading, improved solubility, enhanced bioavailability, and reduced systemic toxicity. The successful commercialization of products like Emend®, Tricor®, Rapamune®, and Celevrex® underscores the potential and conversion opportunities of nanocrystals. However, the application of nanocrystals in herbal medicine is still in its early stages.
Currently, the evaluation of herbal medicine nanocrystals often focuses on physical characterization, in vitro dissolution, and bioavailability. However, aspects such as physical and chemical stability, absorption, and therapeutic mechanisms deserve more attention. Integrating the clinical theories of herbal medicine with nanocrystal technology is essential for its comprehensive development. Moreover, research on the in vivo fate of herbal medicine nanocrystals has primarily been concentrated on oral and intravenous administration, with limited exploration in pulmonary, ophthalmic, and intranasal delivery methods. Nanotoxicity is an important consideration in the development of herbal medicine nanocrystals, necessitating thorough investigation to ensure their safety. To avoid the toxicity caused by synthetic stabilizers, naturally derived stabilizers may be a novel development direction for the next generation of nanocrystals.
In conclusion, the combination of herbal medicines with nanocrystals holds promise for overcoming drug resistance and improving therapeutic outcomes. However, thorough investigations are needed in order to understand the interactions between nanocrystals, herbal medicines, and biological systems to ensure safe and effective treatment.

Author Contributions

M.G. contributed to the drafting of the paper; S.W. contributed to the literature collection and sorting; M.S. and H.Y. contributed to the figure drawing; S.Q. and X.W. contributed to the content revision; M.G., P.F. and Z.J. contributed to the conception and design; Z.J. contributed to the final approval of the version to be published. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key Clinical Specialties Construction Program, the Youth Fund of National Natural Science Foundation of China (No. 82104081), the Sichuan Province Science and Technology Support Program (No. 2023NSFSC1682), and the Science and Technology Project of Xinjiang Production and Construction Corps (No. 2022AB020).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

10-HCPT 10-hydroxycamptothecin
ADG Andrographolide
ANDA Abbreviated New Drug Application
ART Artemisinin
BCS Biopharmaceutics classification system
BSA Bovine serum albumin
CUR Curcumin
DSPE 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine
EPAS Evaporative precipitation into aqueous solution
GNA Gambogenic acid
HPC Hydroxypropyl cellulose
HPH High-pressure homogenization
HPMC Hydroxypropyl Methyl Cellulose
ICT Icaritin
IND Investigational New Drug
MDA Malondialdehyde
NDA New Drug Application
NRG Naringenin
P188 Poloxamer 188
PG Propylene glycol
PVA Polyvinyl acetate
PVP Polyvinylpyrrolidone
PVP/VA Polyvinylpyrrolidone/vinyl acetate
QT Quercetin
SCF Supercritical fluid technology
SDS Sodium dodecyl sulfate
SOD Superoxide dismutase
TPGS D-α-Tocopherol polyethylene glycol succinate
WMM Wet media milling

References

  1. Ekor, M. The growing use of herbal medicines: Issues relating to adverse reactions and challenges in monitoring safety. Front. Pharmacol. 2014, 4, 177. [Google Scholar] [CrossRef] [PubMed]
  2. Caesar, L.K.; Cech, N.B. Synergy and antagonism in natural product extracts: When 1 + 1 does not equal 2. Nat. Prod. Rep. 2019, 36, 869–888. [Google Scholar] [CrossRef] [PubMed]
  3. Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.; Heiss, E.H.; et al. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv. 2015, 33, 1582–1614. [Google Scholar] [CrossRef]
  4. Teja, P.K.; Mithiya, J.; Kate, A.S.; Bairwa, K.; Chauthe, S.K. Herbal nanomedicines: Recent advancements, challenges, opportunities and regulatory overview. Phytomedicine 2022, 96, 153890. [Google Scholar] [CrossRef]
  5. Giordano, A.; Tommonaro, G. Curcumin and Cancer. Nutrients 2019, 11, 2376. [Google Scholar] [CrossRef] [PubMed]
  6. Singh, L.; Sharma, S.; Xu, S.; Tewari, D.; Fang, J. Curcumin as a Natural Remedy for Atherosclerosis: A Pharmacological Review. Molecules 2021, 26, 4036. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, J.; Hu, K.; Di, L.; Wang, P.; Liu, Z.; Zhang, J.; Yue, P.; Song, W.; Zhang, J.; Chen, T.; et al. Traditional herbal medicine and nanomedicine: Converging disciplines to improve therapeutic efficacy and human health. Adv. Drug Deliv. Rev. 2021, 178, 113964. [Google Scholar] [CrossRef]
  8. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef]
  9. Singh, S.K.; Vaidya, Y.; Gulati, M.; Bhattacharya, S.; Garg, V.; Pandey, N.K. Nanosuspension: Principles, Perspectives and Practices. Curr. Drug Deliv. 2016, 13, 1222–1246. [Google Scholar] [CrossRef]
  10. 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]
  11. Guo, M.; Wei, M.; Li, W.; Guo, M.; Guo, C.; Ma, M.; Wang, Y.; Yang, Z.; Li, M.; Fu, Q.; et al. Impacts of particle shapes on the oral delivery of drug nanocrystals: Mucus permeation, transepithelial transport and bioavailability. J. Control. Release 2019, 307, 64–75. [Google Scholar] [CrossRef]
  12. Guo, M.R.; Chen, H.N.; Zhang, C.; Zhang, G.S.; Wang, Y.Z.; Li, P.C.; Fu, Q. Probing the particle shape effects on the biodistribution and antihyperlipidemic efficiency for oral lovastatin nanocrystals. J. Mol. Liq. 2021, 324, 114700. [Google Scholar] [CrossRef]
  13. Zhang, G.; Wang, Y.; Zhang, Z.; He, Z.; Liu, Y.; Fu, Q. FRET imaging revealed that nanocrystals enhanced drug oral absorption by dissolution rather than endocytosis: A case study of coumarin 6. J. Control. Release 2021, 332, 225–232. [Google Scholar] [CrossRef] [PubMed]
  14. Fu, Q.; Sun, J.; Ai, X.; Zhang, P.; Li, M.; Wang, Y.; Liu, X.; Sun, Y.; Sui, X.; Sun, L.; et al. Nimodipine nanocrystals for oral bioavailability improvement: Role of mesenteric lymph transport in the oral absorption. Int. J. Pharm. 2013, 448, 290–297. [Google Scholar] [CrossRef] [PubMed]
  15. Guan, W.H.; Ma, Y.Y.; Ding, S.J.; Liu, Y.C.; Song, Z.M.; Liu, X.; Tang, L.L.; Wang, Y.C. The technology for improving stability of nanosuspensions in drug delivery. J. Nanopart. Res. 2022, 24, 14. [Google Scholar] [CrossRef]
  16. Wang, Y.; Zheng, Y.; Zhang, L.; Wang, Q.; Zhang, D. Stability of nanosuspensions in drug delivery. J. Control. Release 2013, 172, 1126–1141. [Google Scholar] [CrossRef]
  17. Chen, M.L.; John, M.; Lee, S.L.; Tyner, K.M. Development Considerations for Nanocrystal Drug Products. AAPS J. 2017, 19, 642–651. [Google Scholar] [CrossRef]
  18. Fan, M.; Geng, S.; Liu, Y.; Wang, J.; Wang, Y.; Zhong, J.; Yan, Z.; Yu, L. Nanocrystal Technology as a Strategy to Improve Drug Bioavailability and Antitumor Efficacy for the Cancer Treatment. Curr. Pharm. Des. 2018, 24, 2416–2424. [Google Scholar] [CrossRef]
  19. Lee, H.; Bang, J.B.; Na, Y.G.; Lee, J.Y.; Cho, C.W.; Baek, J.S.; Lee, H.K. Development and Evaluation of Tannic Acid-Coated Nanosuspension for Enhancing Oral Bioavailability of Curcumin. Pharmaceutics 2021, 13, 1460. [Google Scholar] [CrossRef]
  20. Murakami, Y.; Shimoyama, Y. Production of nanosuspension functionalized by chitosan using supercritical fluid extraction of emulsion. J. Supercrit. Fluids 2017, 128, 121–127. [Google Scholar] [CrossRef]
  21. Sarkari, M.; Brown, J.; Chen, X.; Swinnea, S.; Williams, R.O., III; Johnston, K.P. Enhanced drug dissolution using evaporative precipitation into aqueous solution. Int. J. Pharm. 2002, 243, 17–31. [Google Scholar] [CrossRef] [PubMed]
  22. Gao, L.; Liu, G.; Kang, J.; Niu, M.; Wang, Z.; Wang, H.; Ma, J.; Wang, X. Paclitaxel nanosuspensions coated with P-gp inhibitory surfactants: I. Acute toxicity and pharmacokinetics studies. Colloids Surf. B Biointerfaces 2013, 111, 277–281. [Google Scholar] [CrossRef] [PubMed]
  23. Bosselmann, S.; Nagao, M.; Chow, K.T.; Williams, R.O., III. Influence of formulation and processing variables on properties of itraconazole nanoparticles made by advanced evaporative precipitation into aqueous solution. AAPS PharmSciTech 2012, 13, 949–960. [Google Scholar] [CrossRef] [PubMed]
  24. Kakran, M.; Sahoo, N.G.; Li, L.; Judeh, Z. Dissolution of artemisinin/polymer composite nanoparticles fabricated by evaporative precipitation of nanosuspension. J. Pharm. Pharmacol. 2010, 62, 413–421. [Google Scholar] [CrossRef] [PubMed]
  25. Lu, Y.; Chen, Y.; Gemeinhart, R.A.; Wu, W.; Li, T. Developing nanocrystals for cancer treatment. Nanomedicine 2015, 10, 2537–2552. [Google Scholar] [CrossRef]
  26. 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]
  27. Lizonova, D.; Hladek, F.; Chvila, S.; Balaz, A.; Stankova, S.; Stepanek, F. Surface stabilization determines macrophage uptake, cytotoxicity, and bioactivity of curcumin nanocrystals. Int. J. Pharm. 2022, 626, 122133. [Google Scholar] [CrossRef]
  28. Lou, H.; Gao, L.; Wei, X.; Zhang, Z.; Zheng, D.; Zhang, D.; Zhang, X.; Li, Y.; Zhang, Q. Oridonin nanosuspension enhances anti-tumor efficacy in SMMC-7721 cells and H22 tumor bearing mice. Colloids Surf. B Biointerfaces 2011, 87, 319–325. [Google Scholar] [CrossRef]
  29. Yang, X.; Liu, Y.; Zhao, Y.; Han, M.; Guo, Y.; Kuang, H.; Wang, X. A stabilizer-free and organic solvent-free method to prepare 10-hydroxycamptothecin nanocrystals: In vitro and in vivo evaluation. Int. J. Nanomed. 2016, 11, 2979–2994. [Google Scholar] [CrossRef]
  30. Meng, T.T.; Qiao, F.X.; Ma, S.J.; Gao, T.; Li, L.; Hou, Y.H.; Yang, J.H. Exploring the influence factors and improvement strategies of drug polymorphic transformation combined kinetic and thermodynamic perspectives during the formation of nanosuspensions. Drug Dev. Ind. Pharm. 2021, 47, 1867–1880. [Google Scholar] [CrossRef]
  31. Dupont, A.; Guerain, M.; Danede, F.; Paccou, L.; Guinet, Y.; Hedoux, A.; Willart, J.F. Kinetics and mechanism of polymorphic transformation of sorbitol under mechanical milling. Int. J. Pharm. 2020, 590, 119902. [Google Scholar] [CrossRef] [PubMed]
  32. Mazel, V.; Delplace, C.; Busignies, V.; Faivre, V.; Tchoreloff, P.; Yagoubi, N. Polymorphic transformation of anhydrous caffeine under compression and grinding: A re-evaluation. Drug Dev. Ind. Pharm. 2011, 37, 832–840. [Google Scholar] [CrossRef]
  33. Descamps, M.; Willart, J.F. Perspectives on the amorphisation/milling relationship in pharmaceutical materials. Adv. Drug Deliv. Rev. 2016, 100, 51–66. [Google Scholar] [CrossRef]
  34. Aleandri, S.; Schonenberger, M.; Niederquell, A.; Kuentz, M. Temperature-Induced Surface Effects on Drug Nanosuspensions. Pharm. Res. 2018, 35, 69. [Google Scholar] [CrossRef] [PubMed]
  35. Nandi, S.; Padrela, L.; Tajber, L.; Collas, A. Nucleation kinetics-based solvent selection for the liquid antisolvent crystallization of a lipophilic intermediate. J. Mol. Liq. 2023, 375, 121306. [Google Scholar] [CrossRef]
  36. Shariare, M.H.; Sharmin, S.; Jahan, I.; Reza, H.M.; Mohsin, K. The impact of process parameters on carrier free paracetamol nanosuspension prepared using different stabilizers by antisolvent precipitation method. J. Drug Deliv. Sci. Technol. 2018, 43, 122–128. [Google Scholar] [CrossRef]
  37. Chen, Z.J.; Wu, W.; Lu, Y. What is the future for nanocrystal-based drug-delivery systems? Ther. Deliv. 2020, 11, 225–230. [Google Scholar] [CrossRef] [PubMed]
  38. Ahmadi Tehrani, A.; Omranpoor, M.M.; Vatanara, A.; Seyedabadi, M.; Ramezani, V. Formation of nanosuspensions in bottom-up approach: Theories and optimization. Daru 2019, 27, 451–473. [Google Scholar] [CrossRef]
  39. Yang, X.; Ong, T.C.; Michaelis, V.K.; Heng, S.; Griffin, R.G.; Myerson, A.S. Formation of Organic Molecular Nanocrystals under Soft Confinement. CrystEngComm 2015, 17, 6044–6052. [Google Scholar] [CrossRef]
  40. Lindfors, L.; Forssen, S.; Westergren, J.; Olsson, U. Nucleation and crystal growth in supersaturated solutions of a model drug. J. Colloid Interface Sci. 2008, 325, 404–413. [Google Scholar] [CrossRef]
  41. Wang, X.H.; Liu, Y.; Shen, C.Y.; Shen, B.D.; Zhong, R.N.; Yuan, H.L. Effect of particle size on in vitro and in vivo behavior of astilbin nanosuspensions. J. Drug Deliv. Sci. Technol. 2019, 52, 778–783. [Google Scholar] [CrossRef]
  42. Wang, Y.; Wang, L.; Liu, Z.; Zhang, D.; Zhang, Q. In vivo evaluation of silybin nanosuspensions targeting liver. J. Biomed. Nanotechnol. 2012, 8, 760–769. [Google Scholar] [CrossRef] [PubMed]
  43. Zheng, D.; Wang, Y.; Zhang, D.; Liu, Z.; Duan, C.; Jia, L.; Wang, F.; Liu, Y.; Liu, G.; Hao, L.; et al. In vitro antitumor activity of silybin nanosuspension in PC-3 cells. Cancer Lett. 2011, 307, 158–164. [Google Scholar] [CrossRef] [PubMed]
  44. Shelar, D.B.; Pawar, S.K.; Vavia, P.R. Fabrication of isradipine nanosuspension by anti-solvent microprecipitation-high-pressure homogenization method for enhancing dissolution rate and oral bioavailability. Drug Deliv. Transl. Res. 2013, 3, 384–391. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, T.; Muller, R.H.; Moschwitzer, J.P. Effect of drug physico-chemical properties on the efficiency of top-down process and characterization of nanosuspension. Expert Opin. Drug Deliv. 2015, 12, 1741–1754. [Google Scholar] [CrossRef]
  46. Shin, G.H.; Li, J.; Cho, J.H.; Kim, J.T.; Park, H.J. Enhancement of Curcumin Solubility by Phase Change from Crystalline to Amorphous in Cur-TPGS Nanosuspension. J. Food Sci. 2016, 81, N494–N501. [Google Scholar] [CrossRef]
  47. Liu, T.; Muller, R.H.; Moschwitzer, J.P. Consideration of the solid state for resveratrol nanocrystal production. Powder Technol. 2018, 332, 63–69. [Google Scholar] [CrossRef]
  48. Bellucci, M.A.; Marx, A.; Wang, B.; Fang, L.; Zhou, Y.; Greenwell, C.; Li, Z.; Becker, A.; Sun, G.; Brandenburg, J.G.; et al. Effect of Polymer Additives on the Crystal Habit of Metformin HCl. Small Methods 2023, 7, e2201692. [Google Scholar] [CrossRef]
  49. Nakanishi, Y.; Chang, F.R.; Liaw, C.C.; Wu, Y.C.; Bastow, K.F.; Lee, K.H. Acetogenins as selective inhibitors of the human ovarian 1A9 tumor cell line. J. Med. Chem. 2003, 46, 3185–3188. [Google Scholar] [CrossRef]
  50. Hong, J.; Li, Y.; Li, Y.; Xiao, Y.; Kuang, H.; Wang, X. Annonaceous acetogenins nanosuspensions stabilized by PCL-PEG block polymer: Significantly improved antitumor efficacy. Int. J. Nanomed. 2016, 11, 3239–3253. [Google Scholar] [CrossRef]
  51. Hopp, D.C.; Alali, F.Q.; Gu, Z.M.; McLaughlin, J.L. Three new bioactive bis-adjacent THF-ring acetogenins from the bark of Annona squamosa. Bioorg. Med. Chem. 1998, 6, 569–575. [Google Scholar] [CrossRef] [PubMed]
  52. Ahammadsahib, K.I.; Hollingworth, R.M.; Mcgovren, J.P.; Hui, Y.H.; Mclaughlin, J.L. Mode of Action of Bullatacin—A Potent Antitumor and Pesticidal Annonaceous Acetogenin. Life Sci. 1993, 53, 1113–1120. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, Y.; Chen, J.W.; Li, X. Cytotoxic Bistetrahydrofuran Annonaceous Acetogenins from the Seeds of Annona squamosa. J. Nat. Prod. 2011, 74, 2477–2481. [Google Scholar] [CrossRef] [PubMed]
  54. Oberlies, N.H.; Croy, V.L.; Harrison, M.L.; McLaughlin, J.L. The Annonaceous acetogenin bullatacin is cytotoxic against multidrug-resistant human mammary adenocarcinoma cells. Cancer Lett. 1997, 115, 73–79. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, Y.; Chen, J.W.; Zhai, J.H.; Wang, Y.; Wang, S.L.; Li, X. Antitumor activity and toxicity relationship of annonaceous acetogenins. Food Chem. Toxicol. 2013, 58, 394–400. [Google Scholar] [CrossRef] [PubMed]
  56. Fan, Y.; Peng, A.H.; He, S.C.; Shao, X.M.; Nie, C.L.; Chen, L.J. Isogambogenic acid inhibits tumour angiogenesis by suppressing Rho GTPases and vascular endothelial growth factor receptor 2 signalling pathway. J. Chemother. 2013, 25, 298–308. [Google Scholar] [CrossRef]
  57. Zhou, J.; Luo, Y.H.; Wang, J.R.; Lu, B.B.; Wang, K.M.; Tian, Y. Gambogenic Acid Induction of Apoptosis in a Breast Cancer Cell Line. Asian Pac. J. Cancer Prev. 2013, 14, 7601–7605. [Google Scholar] [CrossRef]
  58. Wang, M.; Chen, D.; Cheng, H.; Li, B.; Yan, F.G.; Su, J.J.; Peng, C.; Sun, M.L.; Hu, Y.W.; Wang, X.S.; et al. Gambogenic Acid Kills Lung Cancer Cells through Aberrant Autophagy. PLoS ONE 2014, 9, e83604. [Google Scholar] [CrossRef]
  59. Chen, R.; Zhang, H.; Liu, P.; Wu, X.; Chen, B. Gambogenic acid synergistically potentiates bortezomib-induced apoptosis in multiple myeloma. J. Cancer 2017, 8, 839–851. [Google Scholar] [CrossRef]
  60. Xu, Q.; Guo, J.; Chen, W. Gambogenic acid reverses P-glycoprotein mediated multidrug resistance in HepG2/Adr cells and its underlying mechanism. Biochem. Biophys. Res. Commun. 2019, 508, 882–888. [Google Scholar] [CrossRef]
  61. Yuan, H.; Li, X.; Zhang, C.; Pan, W.; Liang, Y.; Chen, Y.; Chen, W.; Liu, L.; Wang, X. Nanosuspensions as delivery system for gambogenic acid: Characterization and in vitro/in vivo evaluation. Drug Deliv. 2016, 23, 2772–2779. [Google Scholar] [CrossRef] [PubMed]
  62. Li, Q.Y.; Zu, Y.G.; Shi, R.Z.; Yao, L.P. Review Camptothecin: Current Perspectives. Curr. Med. Chem. 2006, 13, 2021–2039. [Google Scholar] [CrossRef] [PubMed]
  63. Yao, Z.; Wang, W.; Ning, J.; Zhang, X.; Zheng, W.; Qian, Y.; Fan, C. Hydroxycamptothecin Inhibits Peritendinous Adhesion via the Endoplasmic Reticulum Stress-Dependent Apoptosis. Front. Pharmacol. 2019, 10, 967. [Google Scholar] [CrossRef]
  64. Zhou, M.; Zhang, X.; Yu, C.; Nan, X.; Chen, X.; Zhang, X. Shape regulated anticancer activities and systematic toxicities of drug nanocrystals in vivo. Nanomedicine 2016, 12, 181–189. [Google Scholar] [CrossRef]
  65. Huang, X.; Zhu, H.L. Resveratrol and its analogues: Promising antitumor agents. Anti-Cancer Agents Med. Chem. 2011, 11, 479–490. [Google Scholar] [CrossRef]
  66. Bian, P.; Hu, W.; Liu, C.; Li, L. Resveratrol potentiates the anti-tumor effects of rapamycin in papillary thyroid cancer: PI3K/AKT/mTOR pathway involved. Arch. Biochem. Biophys. 2020, 689, 108461. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, L.; Liu, Y.; Zhao, J.; Li, C.; Zhou, Y.; Du, J.; Wang, Y. In vitro and in vivo evaluation of targeting tumor with folate-based amphiphilic multifunctional stabilizer for resveratrol nanosuspensions. Colloids Surf. B Biointerfaces 2017, 160, 462–472. [Google Scholar] [CrossRef]
  68. Ancic, D.; Orsolic, N.; Odeh, D.; Tomasevic, M.; Pepic, I.; Ramic, S. Resveratrol and its nanocrystals: A promising approach for cancer therapy? Toxicol. Appl. Pharmacol. 2022, 435, 115851. [Google Scholar] [CrossRef]
  69. Zhang, Z.; Zhang, X.; Xue, W.; Yangyang, Y.; Xu, D.; Zhao, Y.; Lou, H. Effects of oridonin nanosuspension on cell proliferation and apoptosis of human prostatic carcinoma PC-3 cell line. Int. J. Nanomed. 2010, 5, 735–742. [Google Scholar] [CrossRef]
  70. Tang, X.; Sun, J.; Ge, T.; Zhang, K.; Gui, Q.; Zhang, S.; Chen, W. PEGylated liposomes as delivery systems for Gambogenic acid: Characterization and in vitro/in vivo evaluation. Colloids Surf. B Biointerfaces 2018, 172, 26–36. [Google Scholar] [CrossRef]
  71. Qiao, F.; Zhao, Y.; Mai, Y.; Guo, J.; Dong, L.; Zhang, W.; Yang, J. Isoliquiritigenin Nanosuspension Enhances Cytostatic Effects in A549 Lung Cancer Cells. Planta Med. 2020, 86, 538–547. [Google Scholar] [CrossRef] [PubMed]
  72. Li, W.; Das, S.; Ng, K.Y.; Heng, P.W. Formulation, biological and pharmacokinetic studies of sucrose ester-stabilized nanosuspensions of oleanolic Acid. Pharm. Res. 2011, 28, 2020–2033. [Google Scholar] [CrossRef] [PubMed]
  73. Huang, T.; Wang, Y.; Shen, Y.; Ao, H.; Guo, Y.; Han, M.; Wang, X. Preparation of high drug-loading celastrol nanosuspensions and their anti-breast cancer activities in vitro and in vivo. Sci. Rep 2020, 10, 8851. [Google Scholar] [CrossRef] [PubMed]
  74. Yang, L.; Hong, J.; Di, J.; Guo, Y.; Han, M.; Liu, M.; Wang, X. 10-Hydroxycamptothecin (HCPT) nanosuspensions stabilized by mPEG(1000)-HCPT conjugate: High stabilizing efficiency and improved antitumor efficacy. Int. J. Nanomed. 2017, 12, 3681–3695. [Google Scholar] [CrossRef]
  75. Sun, M.; Gao, Y.; Pei, Y.; Guo, C.; Li, H.; Cao, F.; Yu, A.; Zhai, G. Development of nanosuspension formulation for oral delivery of quercetin. J. Biomed. Nanotechnol. 2010, 6, 325–332. [Google Scholar] [CrossRef]
  76. Pengnam, S.; Charoensuksai, P.; Yingyongnarongkul, B.E.; Saeeng, R.; Uludag, H.; Patrojanasophon, P.; Opanasopit, P.; Plianwong, S. siRNA Targeting Mcl-1 Potentiates the Anticancer Activity of Andrographolide Nanosuspensions via Apoptosis in Breast Cancer Cells. Pharmaceutics 2022, 14, 1196. [Google Scholar] [CrossRef]
  77. Hong, J.Y.; Liu, Y.Y.; Xiao, Y.; Yang, X.F.; Su, W.J.; Zhang, M.Z.; Liao, Y.H.; Kuang, H.X.; Wang, X.T. High drug payload curcumin nanosuspensions stabilized by mPEG-DSPE and SPC: In vitro and in vivo evaluation. Drug Deliv. 2017, 24, 109–120. [Google Scholar] [CrossRef]
  78. Gao, Y.; Li, Z.G.; Sun, M.; Li, H.L.; Guo, C.Y.; Cui, J.; Li, A.G.; Cao, F.L.; Xi, Y.W.; Lou, H.X.; et al. Preparation, characterization, pharmacokinetics, and tissue distribution of curcumin nanosuspension with TPGS as stabilizer. Drug Dev. Ind. Pharm. 2010, 36, 1225–1234. [Google Scholar] [CrossRef]
  79. Jin, L.; Zeng, W.; Zhang, F.; Zhang, C.; Liang, W. Naringenin Ameliorates Acute Inflammation by Regulating Intracellular Cytokine Degradation. J. Immunol. 2017, 199, 3466–3477. [Google Scholar] [CrossRef]
  80. El-Shitany, N.A.; Eid, B.G. Icariin modulates carrageenan-induced acute inflammation through HO-1/Nrf2 and NF-kB signaling pathways. Biomed. Pharmacother. 2019, 120, 109567. [Google Scholar] [CrossRef]
  81. Guo, Y.; Wang, S.; Wang, Y.; Zhu, T. Silymarin improved diet-induced liver damage and insulin resistance by decreasing inflammation in mice. Pharm. Biol. 2016, 54, 2995–3000. [Google Scholar] [CrossRef] [PubMed]
  82. Yardim, A.; Kucukler, S.; Ozdemir, S.; Comakli, S.; Caglayan, C.; Kandemir, F.M.; Celik, H. Silymarin alleviates docetaxel-induced central and peripheral neurotoxicity by reducing oxidative stress, inflammation and apoptosis in rats. Gene 2021, 769, 145239. [Google Scholar] [CrossRef] [PubMed]
  83. Gera, S.; Sampathi, S.; Maddukuri, S.; Dodoala, S.; Junnuthula, V.; Dyawanapelly, S. Therapeutic Potential of Naringenin Nanosuspension: In Vitro and In Vivo Anti-Osteoporotic Studies. Pharmaceutics 2022, 14, 1449. [Google Scholar] [CrossRef] [PubMed]
  84. Dong, Z.Q.; Wang, X.T.; Wang, M.Y.; Wang, R.; Meng, Z.; Wang, X.T.; Yu, B.; Han, M.H.; Guo, Y.F. Optimization of Naringenin Nanoparticles to Improve the Antitussive Effects on Post-Infectious Cough. Molecules 2022, 27, 3736. [Google Scholar] [CrossRef]
  85. Li, Y.; Sun, S.P.; Chang, Q.; Zhang, L.; Wang, G.N.; Chen, W.X.; Miao, X.Q.; Zheng, Y. A Strategy for the Improvement of the Bioavailability and Antiosteoporosis Activity of BCS IV Flavonoid Glycosides through the Formulation of Their Lipophilic Aglycone into Nanocrystals. Mol. Pharm. 2013, 10, 2534–2542. [Google Scholar] [CrossRef]
  86. Li, H.M.; Zhuo, H.Y.; Yin, D.; Li, W.; Zhang, Y.M.; Li, P.; Zou, L. Intra-Articular Injection of a Nanosuspension of Tetramethylpyrazine Dihydroxynaphthalenate for Stronger and Longer-Lasting Effects Against Osteoarthritis. J. Biomed. Nanotechnol. 2021, 17, 1199–1207. [Google Scholar] [CrossRef]
  87. Casula, L.; Lai, F.; Pini, E.; Valenti, D.; Sinico, C.; Cardia, M.C.; Marceddu, S.; Ailuno, G.; Fadda, A.M. Pulmonary Delivery of Curcumin and Beclomethasone Dipropionate in a Multicomponent Nanosuspension for the Treatment of Bronchial Asthma. Pharmaceutics 2021, 13, 1300. [Google Scholar] [CrossRef]
  88. Mishra, S.B.; Pandey, H.; Pandey, A.C. Nanosuspension of Phyllanthus amarus extract for improving oral bioavailability and prevention of paracetamol induced hepatotoxicity in Sprague-Dawley rats. Adv. Nat. Sci. Nanosci. Nanotechnol. 2013, 4, 035007. [Google Scholar] [CrossRef]
  89. Ibrahim, A.H.; Rosqvist, E.; Smatt, J.H.; Ibrahim, H.M.; Ismael, H.R.; Afouna, M.I.; Samy, A.M.; Rosenholm, J.M. Formulation and optimization of lyophilized nanosuspension tablets to improve the physicochemical properties and provide immediate release of silymarin. Int. J. Pharm. 2019, 563, 217–227. [Google Scholar] [CrossRef]
  90. Qiao, H.Z.; Chen, L.H.; Rui, T.Q.; Wang, J.X.; Chen, T.; Fu, T.M.; Li, J.S.; Di, L.Q. Fabrication and in vitro/in vivo evaluation of amorphous andrographolide nanosuspensions stabilized by D-alpha-tocopheryl polyethylene glycol 1000 succinate/sodium lauryl sulfate. Int. J. Nanomed. 2017, 12, 1033–1046. [Google Scholar] [CrossRef]
  91. Koonrungsesomboon, N.; Karbwang, J. Ethical considerations in clinical research on herbal medicine for prevention of cardiovascular disease in the ageing. Phytomedicine 2016, 23, 1090–1094. [Google Scholar] [CrossRef] [PubMed]
  92. Shaito, A.; Thuan, D.T.B.; Phu, H.T.; Nguyen, T.H.D.; Hasan, H.; Halabi, S.; Abdelhady, S.; Nasrallah, G.K.; Eid, A.H.; Pintus, G. Herbal Medicine for Cardiovascular Diseases: Efficacy, Mechanisms, and Safety. Front. Pharmacol. 2020, 11, 422. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, M.Q.; Wang, S.Z.; Goel, V.; Ling, L.; Shan, J. Anti-Oxidant Activity of Different Herbal Extracts Used in Traditional Chinese Medicine for the Treatment of Cardiovascular Disease. Pharm. Biol. 2010, 48, 29–30. [Google Scholar]
  94. Lee, T.Y.; Chang, C.C.; Lu, W.J.; Yen, T.L.; Lin, K.H.; Geraldine, P.; Li, J.Y.; Sheu, J.R. Honokiol as a specific collagen receptor glycoprotein VI antagonist on human platelets: Functional ex vivo and in vivo studies. Sci. Rep. 2017, 7, 40002. [Google Scholar] [CrossRef] [PubMed]
  95. Jayakumari, N.R.; Rajendran, R.S.; Sivasailam, A.; Parambil, S.T.; Reghuvaran, A.C.; Sreelatha, H.V.; Gopala, S. Honokiol regulates mitochondrial substrate utilization and cellular fatty acid metabolism in diabetic mice heart. Eur. J. Pharmacol. 2021, 896, 173918. [Google Scholar] [CrossRef] [PubMed]
  96. Elbarbry, F.; Moshirian, N. The Modulation of Arachidonic Acid Metabolism and Blood Pressure-Lowering Effect of Honokiol in Spontaneously Hypertensive Rats. Molecules 2022, 27, 3396. [Google Scholar] [CrossRef]
  97. Han, M.H.; Yu, X.; Guo, Y.F.; Wang, Y.H.; Kuang, H.X.; Wang, X.T. Honokiol nanosuspensions: Preparation, increased oral bioavailability and dramatically enhanced biodistribution in the cardio-cerebro-vascular system. Colloids Surf. B Biointerfaces 2014, 116, 114–120. [Google Scholar] [CrossRef]
  98. Wang, Z.P.; Wu, J.B.; Zhou, Q.; Wang, Y.F.; Chen, T.S. Berberine Nanosuspension Enhances Hypoglycemic Efficacy on Streptozotocin Induced Diabetic C57BL/6 Mice. Evid.-Based Complement. Altern. Med. 2015, 2015, 239749. [Google Scholar] [CrossRef]
  99. Rui, T.Q.; Zhang, L.; Qiao, H.Z.; Huang, P.; Qian, S.; Li, J.S.; Chen, Z.P.; Fu, T.M.; Di, L.Q.; Cai, B.C. Preparation and Physicochemical and Pharmacokinetic Characterization of Ginkgo Lactone Nanosuspensions for Antiplatelet Aggregation. J. Pharm. Sci. 2016, 105, 242–249. [Google Scholar] [CrossRef]
  100. Zhao, X.; Wang, W.; Zu, Y.; Zhang, Y.; Li, Y.; Sun, W.; Shan, C.; Ge, Y. Preparation and characterization of betulin nanoparticles for oral hypoglycemic drug by antisolvent precipitation. Drug Deliv. 2014, 21, 467–479. [Google Scholar] [CrossRef]
  101. Singh, A.K.; Pandey, H.; Ramteke, P.W.; Mishra, S.B. Nano-suspension of ursolic acid for improving oral bioavailability and attenuation of type II diabetes: A histopathological investigation. Biocatal. Agric. Biotechnol. 2019, 22, 101433. [Google Scholar] [CrossRef]
  102. Zhang, T.; Li, X.X.; Xu, J.W.; Shao, J.B.; Ding, M.H.; Shi, S.L. Preparation, Characterization, and Evaluation of Breviscapine Nanosuspension and Its Freeze-Dried Powder. Pharmaceutics 2022, 14, 923. [Google Scholar] [CrossRef] [PubMed]
  103. Li, R.L.; Wang, L.Y.; Duan, H.X.; Zhang, Q.; Guo, X.; Wu, C.; Peng, W. Regulation of mitochondrial dysfunction induced cell apoptosis is a potential therapeutic strategy for herbal medicine to treat neurodegenerative diseases. Front. Pharmacol. 2022, 13, 937289. [Google Scholar] [CrossRef] [PubMed]
  104. Yin, R.; Xue, J.; Tan, Y.; Fang, C.; Hu, C.; Yang, Q.; Mei, X.; Qi, D. The Positive Role and Mechanism of Herbal Medicine in Parkinson’s Disease. Oxidative Med. Cell. Longev. 2021, 2021, 9923331. [Google Scholar] [CrossRef] [PubMed]
  105. Li, L.; Zhang, Q.G.; Lai, L.Y.; Wen, X.J.; Zheng, T.; Cheung, C.W.; Zhou, S.Q.; Xu, S.Y. Neuroprotective Effect of Ginkgolide B on Bupivacaine-Induced Apoptosis in SH-SY5Y Cells. Oxidative Med. Cell. Longev. 2013, 2013, 159864. [Google Scholar] [CrossRef]
  106. Hua, J.; Yin, N.; Yang, B.B.; Zhang, J.; Ding, J.H.; Fan, Y.; Hu, G. Ginkgolide B and bilobalide ameliorate neural cell apoptosis in alpha-synuclein aggregates. Biomed. Pharmacother. 2017, 96, 792–797. [Google Scholar] [CrossRef]
  107. Liu, Y.; Liu, W.; Xiong, S.; Luo, J.S.; Li, Y.; Zhao, Y.Y.; Wang, Q.; Zhang, Z.X.; Chen, X.J.; Chen, T.K. Highly stabilized nanocrystals delivering Ginkgolide B in protecting against the Parkinson’s disease. Int. J. Pharm. 2020, 577, 119053. [Google Scholar] [CrossRef]
  108. Stahr, P.L.; Grewal, R.; Eckert, G.P.; Keck, C.M. Investigating hesperetin nanocrystals with tailor-made sizes for the prevention and treatment of Alzheimer’s disease. Drug Deliv. Transl. Res. 2021, 11, 659–674. [Google Scholar] [CrossRef]
  109. Kakran, M.; Sahoo, N.G.; Li, L. Dissolution enhancement of quercetin through nanofabrication, complexation, and solid dispersion. Colloids Surf. B Biointerfaces 2011, 88, 121–130. [Google Scholar] [CrossRef]
  110. Ghaffafi, F.; Moghaddam, A.H.; Zare, M. Neuroprotective Effect of Quercetin Nanocrystal in a 6-Hydroxydopamine Model of Parkinson Disease: Biochemical and Behavioral Evidence. Basic Clin. Neurosci. 2018, 9, 317–324. [Google Scholar] [CrossRef]
  111. Chen, T.K.; Li, C.W.; Li, Y.; Yi, X.; Lee, S.M.Y.; Zheng, Y. Oral Delivery of a Nanocrystal Formulation of Schisantherin A with Improved Bioavailability and Brain Delivery for the Treatment of Parkinson’s Disease. Mol. Pharm. 2016, 13, 3864–3875. [Google Scholar] [CrossRef] [PubMed]
  112. Babylon, L.; Grewal, R.; Stahr, P.L.; Eckert, R.W.; Keck, C.M.; Eckert, G.P. Hesperetin Nanocrystals Improve Mitochondrial Function in a Cell Model of Early Alzheimer Disease. Antioxidants 2021, 10, 1003. [Google Scholar] [CrossRef] [PubMed]
  113. Wei, X.L.; Han, Y.R.; Quan, L.H.; Liu, C.Y.; Liao, Y.H. Oily nanosuspension for long-acting intramuscular delivery of curcumin didecanoate prodrug: Preparation, characterization and in vivo evaluation. Eur. J. Pharm. Sci. 2013, 49, 286–293. [Google Scholar] [CrossRef]
  114. Ang, L.; Song, E.; Zhang, J.; Lee, H.W.; Lee, M.S. Herbal medicine for COVID-19: An overview of systematic reviews and meta-analysis. Phytomedicine 2022, 102, 154136. [Google Scholar] [CrossRef]
  115. Yu, M.; Jin, X.; Liang, C.; Bu, F.; Pan, D.; He, Q.; Ming, Y.; Little, P.; Du, H.; Liang, S.; et al. Berberine for diarrhea in children and adults: A systematic review and meta-analysis. Ther. Adv. Gastroenterol. 2020, 13, 1756284820961299. [Google Scholar] [CrossRef] [PubMed]
  116. Sahibzada, M.U.K.; Sadiq, A.; Faidah, H.S.; Khurram, M.; Amin, M.U.; Haseeb, A.; Kakar, M. Berberine nanoparticles with enhanced in vitro bioavailability: Characterization and antimicrobial activity. Drug Des. Dev. Ther. 2018, 12, 303–312. [Google Scholar] [CrossRef] [PubMed]
  117. Guo, J.J.; Yue, P.F.; Lv, J.L.; Han, J.; Fu, S.S.; Jin, S.X.; Jin, S.Y.; Yuan, H.L. Development and in vivo/in vitro evaluation of novel herpetrione nanosuspension. Int. J. Pharm. 2013, 441, 227–233. [Google Scholar] [CrossRef]
  118. Marchese, A.; Orhan, I.E.; Daglia, M.; Barbieri, R.; Di Lorenzo, A.; Nabavi, S.F.; Gortzi, O.; Izadi, M.; Nabavi, S.M. Antibacterial and antifungal activities of thymol: A brief review of the literature. Food Chem. 2016, 210, 402–414. [Google Scholar] [CrossRef]
  119. Zhou, W.; Zhang, Y.; Li, R.Y.; Peng, S.F.; Ruan, R.; Li, J.H.; Liu, W. Fabrication of Caseinate Stabilized Thymol Nanosuspensions via the pH-Driven Method: Enhancement in Water Solubility of Thymol. Foods 2021, 10, 1074. [Google Scholar] [CrossRef]
  120. Lai, F.; Pireddu, R.; Corrias, F.; Fadda, A.M.; Valenti, D.; Pini, E.; Sinico, C. Nanosuspension improves tretinoin photostability and delivery to the skin. Int. J. Pharm. 2013, 458, 104–109. [Google Scholar] [CrossRef]
  121. Zong, R.; Ruan, H.A.; Zhu, W.Z.; Zhang, P.; Feng, Z.J.; Liu, C.M.; Fan, S.H.; Liang, H.M.; Li, J. Curcumin nanocrystals with tunable surface zeta potential: Preparation, characterization and antibacterial study. J. Drug Deliv. Sci. Technol. 2022, 76, 103771. [Google Scholar] [CrossRef]
  122. Sahar, P.; Ali, T.; Naeem, M.; Hussain, F. Nanotechnology approach for exploring the enhanced bioactivities, biochemical characterisation and phytochemistry of freshly prepared Mentha arvensis L. nanosuspensions. Phytochem. Anal. 2022. [Google Scholar] [CrossRef] [PubMed]
  123. Fujimura, H.; Komasaka, T.; Tomari, T.; Kitano, Y.; Takekawa, K. Nanosuspension formulations of poorly water-soluble compounds for intravenous administration in exploratory toxicity studies: In vitro and in vivo evaluation. J. Appl. Toxicol. 2016, 36, 1259–1267. [Google Scholar] [CrossRef] [PubMed]
  124. Karami, Z.; Bidgoli, S.A.; Zanjani, M.S.; Arabshahi, P.; Gazori, T.; Hamidi, M. Pharmacokinetics and repeated dose 28-day oral toxicity studies of acetaminophen nanosuspension. J. Biomed. Mater. Res. Part B 2023, 111, 1687–1696. [Google Scholar] [CrossRef] [PubMed]
  125. Khanna, P.; Ong, C.; Bay, B.H.; Baeg, G.H. Nanotoxicity: An Interplay of Oxidative Stress, Inflammation and Cell Death. Nanomaterials 2015, 5, 1163–1180. [Google Scholar] [CrossRef] [PubMed]
  126. Ng, C.T.; Yu, L.E.; Ong, C.N.; Bay, B.H.; Baeg, G.H. The use of Drosophila melanogaster as a model organism to study immune-nanotoxicity. Nanotoxicology 2019, 13, 429–446. [Google Scholar] [CrossRef]
  127. Muller, R.H.; Gohla, S.; Keck, C.M. State of the art of nanocrystals—Special features, production, nanotoxicology aspects and intracellular delivery. Eur. J. Pharm. Biopharm. 2011, 78, 1–9. [Google Scholar] [CrossRef]
  128. Doshi, N.; Mitragotri, S. Needle-shaped polymeric particles induce transient disruption of cell membranes. J. R. Soc. Interface 2010, 7, S403–S410. [Google Scholar] [CrossRef]
  129. Zhang, B.K.; Lung, P.S.; Zhao, S.S.; Chu, Z.Q.; Chrzanowski, W.; Li, Q. Shape dependent cytotoxicity of PLGA-PEG nanoparticles on human cells. Sci. Rep. 2017, 7, 7315. [Google Scholar] [CrossRef]
  130. Wang, T.; Qi, J.; Ding, N.; Dong, X.; Zhao, W.; Lu, Y.; Wang, C.; Wu, W. Tracking translocation of self-discriminating curcumin hybrid nanocrystals following intravenous delivery. Int. J. Pharm. 2018, 546, 10–19. [Google Scholar] [CrossRef]
  131. Shen, C.; Yang, Y.; Shen, B.; Xie, Y.; Qi, J.; Dong, X.; Zhao, W.; Zhu, W.; Wu, W.; Yuan, H.; et al. Self-discriminating fluorescent hybrid nanocrystals: Efficient and accurate tracking of translocation via oral delivery. Nanoscale 2017, 10, 436–450. [Google Scholar] [CrossRef]
  132. Qiao, Y.H.; Wei, Z.H.; Qin, T.T.; Song, R.F.; Yu, Z.Q.; Yuan, Q.; Du, J.; Zeng, Q.B.; Zong, L.L.; Duan, S.F.; et al. Combined nanosuspensions from two natural active ingredients for cancer therapy with reduced side effects. Chin. Chem. Lett. 2021, 32, 2877–2881. [Google Scholar] [CrossRef]
  133. Herman, A.; Herman, A.P. Herbal Products and Their Active Constituents Used Alone and in Combination with Antibiotics against Multidrug-Resistant Bacteria. Planta Med. 2022, 89, 168–182. [Google Scholar] [CrossRef] [PubMed]
  134. Kathpalia, H.; Juvekar, S.; Mohanraj, K.; Apsingekar, M.; Shidhaye, S. Investigation of pre-clinical pharmacokinetic parameters of atovaquone nanosuspension prepared using a pH-based precipitation method and its pharmacodynamic properties in a novel artemisinin combination. J. Glob. Antimicrob. Resist. 2020, 22, 248–256. [Google Scholar] [CrossRef]
  135. Siram, K.; Raghavan, C.V.; Marslin, G.; Rahman, H.; Selvaraj, D.; Balakumar, K.; Franklin, G. Quillaja saponin: A prospective emulsifier for the preparation of solid lipid nanoparticles. Colloids Surf. B Biointerfaces 2016, 147, 274–280. [Google Scholar] [CrossRef]
  136. Chen, Y.C.; Liu, Y.; Xu, J.N.; Xie, Y.B.; Zheng, Q.; Yue, P.F.; Yang, M. A Natural Triterpenoid Saponin as Multifunctional Stabilizer for Drug Nanosuspension Powder. Aaps Pharmscitech 2017, 18, 2744–2753. [Google Scholar] [CrossRef]
  137. Long, J.Y.; Song, J.W.; Zhang, X.M.; Deng, M.; Xie, L.; Zhang, L.L.; Li, X.F. Tea saponins as natural stabilizers for the production of hesperidin nanosuspensions. Int. J. Pharm. 2020, 583, 119406. [Google Scholar] [CrossRef]
  138. Chen, H.J.; Deng, M.; Xie, L.; Liu, K.; Zhang, X.M.; Li, X.F. Preparation and characterization of quercetin nanosuspensions using gypenosides as novel stabilizers. J. Drug Deliv. Sci. Technol. 2022, 67, 102962. [Google Scholar] [CrossRef]
  139. Suo, Z.L.; Sun, Q.M.; Peng, X.; Zhang, S.S.; Gan, N.; Zhao, L.D.; Yuan, N.; Zhang, Y.K.; Li, H. Lentinan as a natural stabilizer with bioactivities for preparation of drug-drug nanosuspensions. Int. J. Biol. Macromol. 2021, 184, 101–108. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Application of herbal medicine nanocrystals in various diseases.
Figure 1. Application of herbal medicine nanocrystals in various diseases.
Molecules 28 06370 g001
Figure 2. Nanocrystal preparation methods.
Figure 2. Nanocrystal preparation methods.
Molecules 28 06370 g002
Figure 3. Chemical structures of some of the main herbal medicine active ingredients.
Figure 3. Chemical structures of some of the main herbal medicine active ingredients.
Molecules 28 06370 g003
Table 1. Advantages and limitations of the preparation method of herbal medicine nanocrystals.
Table 1. Advantages and limitations of the preparation method of herbal medicine nanocrystals.
Preparation MethodAdvantagesLimitationsSubdivide Method
Bottom-up
  • simple operation
  • most cost-effective
  • more suitable for intravenous administration of anticancer compounds
  • residual organic solvent may cause toxicity
  • not suitable for drugs which are difficult to dissolve in organic solvents
  • the nucleation and crystal growth processes are difficult to control
  • anti-solvent precipitation
  • supercritical fluid technology
  • evaporative precipitation into aqueous solution
Top-down
  • no need to use an organic solvent
  • easy to scale up production
  • high cost
  • residues of milling media
  • wet media milling
  • high-pressure homogenization
Combinative methods
  • smaller particle sizes
  • relatively short times
  • complicated process
  • residual organic solvent is hard to remove
  • NanoEdge ™
  • H69 technology
  • H42 technology
  • H96 technology
Table 4. Summary of nanocrystal formulations used in herbal medicines for cardiovascular diseases.
Table 4. Summary of nanocrystal formulations used in herbal medicines for cardiovascular diseases.
Extract/CompoundsStabilizersPreparation MethodsParticle SizeBioavailabilityAdvantagesApplicationsRef.
BerberineTPGSHigh-pressure homogenization73.1 ± 3.7 nm/Exhibited superior hypoglycemic, total cholesterol (TC), and body weight reduction effects compared to bulk Ber and metformin (Met, 300 mg/kg).Antidiabetic effect[98]
Ginkgo Lactone/ginkgolide5% P 188 and 5% HPMCHigh-pressure homogenization254 ± 2.8 nmTwofold higher in Cmax and AUC0–t for three ginkgolidesExhibited a significantly higher antiplatelet aggregation effectAntiplatelet aggregation[99]
Honokiol Bovine serum albumin (BSA) and PVPAntisolvent sonoprecipitation116.2 ± 2 nmHonokiol nanocrystals improved the oral bioavailability in rats by 3.94-fold in Cmax and 2.2-fold in AUC0–tHonokiol was released more quickly in vitro from nanocrystals, with no burst release. Honokiol nanocrystals improved the oral bioavailability.
After intraperitoneal administration, Honokiol nanocrystals could dramatically alter the biodistribution, resulting in much higher drug levels and tissue bioavailability in the blood, heart, and brain.
Cardio-cerebro-vascular system[97]
Betulin 0.5% Tween 80Antisolvent precipitation~110 nmThe oral bioavailability of the betulin nanocrystal was 2.21 times that of raw betulin.Higher dissolution rate, solubility, and bioavailability of botulin nanocrystal compared with raw botulin.
Excellent hypoglycemic effect compared with raw betulin
Diabetes mellitus[100]
Ursolic acid 2% of PVANanoprecipitation246.4 ± 4.21 nm/Ursolic acid nanocrystal showed a significant reduction in elevated blood glucose level in a dose-dependent manner with prominent lipid-lowering and antioxidant effects. Type II diabetes[101]
BreviscapineSoybean phospholipidAntisolvent sonoprecipitation303.7 ± 7.3 nm/Breviscapine nanocrystal displayed good stability, increased solubility, and better in vitro releaseCardiovascular and cerebrovascular diseases[102]
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

Guo, M.; Qin, S.; Wang, S.; Sun, M.; Yang, H.; Wang, X.; Fan, P.; Jin, Z. Herbal Medicine Nanocrystals: A Potential Novel Therapeutic Strategy. Molecules 2023, 28, 6370. https://doi.org/10.3390/molecules28176370

AMA Style

Guo M, Qin S, Wang S, Sun M, Yang H, Wang X, Fan P, Jin Z. Herbal Medicine Nanocrystals: A Potential Novel Therapeutic Strategy. Molecules. 2023; 28(17):6370. https://doi.org/10.3390/molecules28176370

Chicago/Turabian Style

Guo, Mengran, Shugang Qin, Shiyan Wang, Min Sun, Huiling Yang, Xinchun Wang, Ping Fan, and Zhaohui Jin. 2023. "Herbal Medicine Nanocrystals: A Potential Novel Therapeutic Strategy" Molecules 28, no. 17: 6370. https://doi.org/10.3390/molecules28176370

Article Metrics

Back to TopTop