Back to Journals » International Journal of Nanomedicine » Volume 20
Emerging Trends in the Application of Nanosuspension-Based Biomaterials for Anticancer Drug Delivery
Authors Aldeeb MME , Wilar G , Suhandi C , Mohammed AFA, Mahmoud SA, Elamin KM , Wathoni N
Received 20 December 2024
Accepted for publication 14 June 2025
Published 1 July 2025 Volume 2025:20 Pages 8587—8607
DOI https://doi.org/10.2147/IJN.S513030
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Jie Huang
Mohamed Mahmud E Aldeeb,1– 3 Gofarana Wilar,4 Cecep Suhandi,1,2 Ahmed Fouad Abdelwahab Mohammed,5 Safwat A Mahmoud,6 Khaled M Elamin,7 Nasrul Wathoni2
1Doctoral Program of Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia; 2Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia; 3Department of Pharmaceutics, Faculty of Pharmacy, Elmergib University, Alkhoms, 40414, Libya; 4Department of Pharmacology and Clinical Pharmacy, Faculty of, Universitas PadjadjaranPharmacy, Jatinangor, 45363, Indonesia; 5Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61517, Egypt; 6Center for Scientific Research and Entrepreneurship, Northern Border University, Arar, 73213, Saudi Arabia; 7Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
Correspondence: Nasrul Wathoni, Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia, Tel +62-22-842-888-888, Email [email protected]
Abstract: The treatment of cancer remains a formidable challenge, largely due to the difficulty in achieving efficient co-delivery of chemotherapeutic and immunotherapeutic agents to specific tumor sites. Nanosuspension-based biomaterial drug delivery systems for anti-cancer (NBDDSC) have emerged as promising platforms for enhancing drug solubility, stability, and targeted delivery. These systems can be categorized into natural polymer-based, synthetic polymer-based, and hybrid nanosuspensions, each offering distinct advantages in biocompatibility, drug loading, and controlled release. However, the majority of existing NBDDSC rely on synthetic materials that function primarily as excipients, offering no intrinsic therapeutic value. These materials often require intricate manufacturing processes, which can result in issues with batch consistency, reduced stability, and diminished therapeutic efficacy. Additionally, the potential side effects associated with synthetic components further underscore the limitations of these systems. This review explores various preparation methods for nanosuspensions, including antisolvent precipitation, high pressure homogenization, and ultrasonication, highlighting their impact on particle size, drug encapsulation, and stability. Furthermore, the targeted applications of these nanosuspensions in treating of cancers such as glioma is discussed to emphasize their potential clinical relevance. By addressing current limitations, this review underscores the critical importance of simpler, safer, and clinically translatable NBDDSC in advancing cancer therapy.
Keywords: nanosuspension, cancer, biomaterials, tumor, NBDDSC
Graphical Abstract:
Introduction
Over the past two decades, the pharmaceutical industry has made significant strides in developing modern technologies to enhance drug discovery and delivery,1 cancer treatment remains a significant challenge due to limitations in drug solubility, bioavailability, and targeted delivery.2–4 Many potent chemotherapeutic and immunotherapeutic agents suffer from poor water solubility, leading to inadequate absorption, suboptimal therapeutic effects, and systemic toxicity. For instance, drugs such as paclitaxel, curcumin, and docetaxel exhibit low aqueous solubility, necessitating innovative delivery systems to enhance their clinical efficacy.
NBDDSC (Figure 1) have emerged as a promising strategy to address these challenges. By reducing drug particle size to the nanometer scale, these systems significantly improve solubility, dissolution rate, and bioavailability. Additionally, nanosuspensions offer enhanced stability and controlled release, making them suitable for both chemotherapeutic and immunotherapeutic applications. Unlike conventional drug carriers, such as liposomes and polymeric nanoparticles, nanosuspensions require fewer excipients and exhibit simpler formulation processes, improving cost-effectiveness and scalability.5
![]() |
Figure 1 The concept of NBDDSs. Created in BioRender. Suliman, K. (2025) https://BioRender.com/ezu4wke. |
The issue of poor solubility not only impedes bioavailability but also affects the therapeutic efficacy of drugs, particularly in cancer treatment. To ensure therapeutic effectiveness, drugs must be bioavailable and easily absorbed, whether administered orally or intravenously.6–8 Consequently, the pharmaceutical industry faces the ongoing challenge of for example, poorly soluble anticancer drugs such as sorafenib and etoposide often require alternative delivery methods to enhance their therapeutic impact. Consequently, the pharmaceutical industry faces the ongoing challenge of developing novel drug delivery systems that enhance the solubility and bioavailability of poorly soluble drugs.9,10
Immunotherapy has revolutionized cancer treatment, particularly in malignancies such as melanoma, lung cancer, and triple-negative breast cancer. However, its clinical success is often hindered by poor drug solubility, rapid degradation, and limited tumor penetration. Nanosuspensions provide a viable solution by enabling the co-delivery of chemotherapeutic agents with immune checkpoint inhibitors (eg, anti-PD-1/PD-L1 antibodies) or cytokines, enhancing immune activation and tumor targeting. Furthermore, nanosuspensions can be engineered to improve tumor accumulation via passive (enhanced permeability and retention effect) or active (ligand-mediated) targeting strategies.11
Among the various strategies developed to address these solubility issues, nanosuspension technology has emerged as one of the most promising. Nanosuspensions are colloidal dispersions of drug nanocrystals in liquid media, typically water.5,12,13 They offer a versatile approach to delivering poorly soluble drugs by reducing particle size to the nanometer scale, thus increasing surface area and dissolution rate.8,14,15 The ability to use nanosuspensions for both oral and intravenous administration further adds to their appeal, making them suitable for various therapeutic applications, including cancer treatment.16–19
Cancer therapy, in particular, presents unique challenges that can be addressed by NBDDSCs for cancer. Conventional chemotherapy and immunotherapy often suffer from suboptimal drug delivery to targeted tumor sites, leading to reduced efficacy and increased systemic toxicity.20–22 Nanosuspensions, due to their nanoscale size and ability to be tailored for controlled and targeted release, offer a promising solution for co-delivering chemotherapeutic and immunotherapeutic agents. Moreover, their relatively simple formulation process and the use of minimal excipients make them cost-effective and easier to manufacture compared to more complex drug delivery systems.20,22–26
The preparation methods of nanosuspensions play a crucial role in optimizing drug encapsulation, stability, and therapeutic performance. Techniques such as high-pressure homogenization, antisolvent precipitation, and ultrasonication facilitate the formulation of stable nanosuspensions with uniform particle size. Additionally, co-delivery strategies, including hybrid nanosuspension systems combining natural and synthetic polymers, further enhance drug loading efficiency and controlled release kinetics.27
Despite these advantages, the clinical translation of NBDDSC faces several challenges, particularly in terms of reproducibility, scalability, and toxicity. These challenges are exacerbated by the complexity of synthetic polymers, which can lead to batch-to-batch variability, instability, and toxicity, necessitating the use of safer, biodegradable alternatives.16,19 Biodegradable biomaterials such as chitosan, alginate, and hyaluronic acid provide safer and more biocompatible options for nanosuspension formulations, reducing adverse effects and enhancing therapeutic outcomes.
Moreover, NBDDSs allow for the co-delivery of multiple therapeutic agents, such as chemotherapeutic drugs and Immunotherapeutics, in a single formulation. This co-delivery capability is essential in cancer therapy, as it enables the simultaneous targeting of multiple pathways involved in tumor growth and immune evasion. By enhancing drug penetration into solid tumors and reducing off-target toxicity, nanosuspension-based systems have shown great potential in improving patient outcomes in cancers such as pancreatic, colorectal, and breast cancer.14
This review aims to provide a detailed exploration of recent trends and challenges in the development of NBDDSC for cancer therapy. By examining the integration of biodegradable biomaterials, novel stabilization techniques, and scalable manufacturing methods, this discussion underscores the critical role of interdisciplinary research in advancing NBDDSC technology. The ultimate goal is to facilitate the clinical translation of these systems, enabling safer and more effective cancer treatments.
Preparation Methods of Nanosuspension
The preparation methods for NBDDSCs play a pivotal role in determining their stability, particle size, and therapeutic efficacy (Figure 2). These methods directly influence the physicochemical properties of the nanosuspensions, including their dissolution rate, bioavailability, and capacity for targeted drug delivery. Achieving consistent and scalable formulations requires precise control over particle size and surface characteristics to ensure batch-to-batch reproducibility and long-term stability.28,29 Recent innovations have focused on refining preparation techniques to address challenges such as aggregation, high-energy input, and scalability.
![]() |
Figure 2 Common preparation methods of NBDDSs. Created in BioRender. Suliman, K. (2025) https://BioRender.com/3i9gma1. |
Nanosuspensions are commonly generated by condensation/aggregation of particles from molecular dispersion to nanosized particles, as in the precipitation method (bottom-up technique), or reduction/dispersion of big particles to nanosized range, as in the milling process (Scale down technology).1,18,29 The surface area of a nanosuspension significantly increases over its initial surface area owing to its smaller particle size. Thus, it can raise the saturation solubility, as determined by the Ostwald–Freundlich equation.15,30 there are several methods to prepare nanosuspension including.
Precipitation Technology
Sub-colloidal materials combine to form particles within the colloidal size range. By creating a supersaturated drug solution in a water-miscible organic solvent at an optimal temperature, and then rapidly agitating it, a small amount of the drug is dispersed in water (a non-solvent), to quickly produce nuclei. This method is highly cost-effective and suitable for producing nanosuspensions with high purity.31 The high level of supersaturation caused by a change in the solvent leads to quick nucleation while preventing supersaturation near the nucleating crystals. According to Ostwald law of nucleation, rapid nucleation and moderate growth rate are critical factors for successful thermodynamically stable crystal formation. The primary benefits of the precipitation method include the production of uniformly sized, finely distributed drugs, ease of scale-up, a straightforward and affordable procedure.32
Elevated levels of supersaturation can produce a needle- or acicular-like crystals, that are easily broken down to produce more nuclei at the expense of crystal development.33 Furthermore, the absence of impurities may result in lattice flaws or faults that can be homogenized to reduce the particle size to the nanoscale. Challenges include controlling the rate of nucleation and growth to prevent aggregation, which can be addressed using surfactants and crystallization inhibitors. Innovations like Nanomorph™ technology have been developed to create stable amorphous nanoparticles, enhancing solubility.34
Homogenization Technology
Homogenization reduces the particle size by forcing the suspension through a valve with a tiny opening at high pressure (100–1000 pressures). The abrupt drop in the fluid velocity results in a loss of static pressure, which in turn creates implosion forces generated by cavitation. Shock waves within the liquid medium then break down the microparticles (less than 25 μm) into nano-sized particles.35 Furthermore, particles with intrinsic crystal flaws can be fractured by the shear forces created by particle collisions at high velocities. Viscosity enhancers can aid in the nanosizing process by increasing the particle density inside the dispersion area and inhibiting crystal formation. The metastable amorphous particles generated by the precipitation process can be transformed into stable crystal forms through homogenization.36 High pressure homogenization is scalable and can be used for producing formulations suitable for parenteral applications.37 Typically, several cycles are required to generate particles within the intended size range. The main advantages of this approach are its simplicity in scaling up, versatility in diluting or concentrating suspensions, low risk of contamination, and viability for aseptic manufacturing38 Despite its benefits, high-pressure homogenization requires multiple cycles to achieve the desired particle size, which can increase production costs and processing time. Additionally, the high shear forces can compromise the stability of certain biomaterials. Recent innovations, such as combining homogenization with temperature-sensitive stabilizers like chitosan or vitamin E TPGS, have mitigated these issues, enabling more efficient and stable production of nanosuspensions.7,22
Wet Milling or Media Milling
This approach uses a media mill or high-shear ball mill to prepare nanosuspensions. Particle attrition and impact both contribute to the size reduction of the medication, stabilizers (s), and water in the chamber. This technique is more appealing because of its low energy consumption, ease of scaling up, minimum batch-to-batch fluctuation, capacity to handle large quantities of materials, and the fact that it already led to four FDA-approved medications.27 The amount of material in the mill is significant because too little feed results in inefficiency and abrasive wear on the mill parts, whereas too much feed has a cushioning effect. The remaining particles in the final product can occasionally be contaminated by deterioration of the milling39 Nevertheless, this issue was reduced by employing a strongly crosslinked polystyrene resin milling medium. The increased amorphous fraction introduced into the materials by a longer milling process can cause instability.40 Recent advancements in bead milling have improved the efficiency of the process, allowing for finer particle sizes and enhanced stability.41 However, wet milling has limitations, including the potential for contamination from the milling media and the generation of heat, which can degrade thermolabile drugs. To address these challenges, researchers have developed modified milling systems that incorporate temperature control mechanisms and inert milling materials to minimize contamination. These advancements have made wet milling a robust option for scaling up nanosuspension production in industrial settings.39
Dry Co-Grinding
A stable nanosuspension was produced by dry co-grinding process using a variety of polymers and copolymers, including polyvinyl pyrrolidone, hydroxypropyl methylcellulose (HPMC), polyethylene glycol, sodium dodecyl sulfate, and cyclodextrin derivatives.15,23 Dry grinding techniques are more cost-effective than wet grinding and do not require the use of hazardous solvents. The enhancement of surface polarity and conversion of a large amount of the drug’s crystalline state to an amorphous form are the most notable effects of the method. For weakly soluble drug nanosuspensions, controlled stability of the amorphous phase can greatly increase the saturation solubility and, consequently, the rate of dissolution.6,22,35
Emulsion-Solvent Evaporation Technique
Drugs are first dissolved in organic solvents or cosolvents to generate an emulsion, which is then dispersed in an aqueous phase containing a surfactant that serves as a stabilizer.25,42 Nanosuspensions are produced by the rapid evaporation of a solvent under low pressure. The size of the globules and stabilizer concentration are important variables to consider when using the emulsification process.28
Ultrasound Assisted Sonocrystallization Method
This is a cutting-edge method for creating stable nanosuspensions. Ultrasound between 20 and 100 kHz improves particle size reduction and regulates the size distribution of the active ingredient in pharmaceuticals. Additionally, This technique is particularly useful for thermosensitive drugs, as it reduces the need for high temperatures.43
Miscellaneous Methods
Another method for producing drug nanosuspensions is to dilute the emulsion, which will allow the dispersed phase to fully diffuse into the continuous phase and produce a nanosuspension. Similar procedures can be applied to produce nanosuspensions in microemulsions. To get the best possible drug loading, it is important to investigate the impact of globule size and surfactant quantity on internal phase drug uptake.13,42 To make the nanosuspension produced by these techniques suitable for administration, clinging solvents and other components must be removed using ultrafiltration. To eliminate the incompatibilities between the different formulation components and increase the physical and chemical stability, lyophilization of the nanosuspensions is required.44 Nanosuspensions can be sterilized by gamma irradiation, steam heat sterilization, or membrane filtration (<0.22 μm). According to published research, optimizing the bottom-up nanosuspension technique necessitates careful selection and calibration of the concentration of excipients, such as polymers and surfactants.23,41
Formulation Consideration of Nanosuspensions
The formulation of nanosuspensions for cancer treatments entails the preparation of colloidal dispersions consisting of drug nanocrystals in a liquid medium, typically water, aimed at improving the solubility and bioavailability of poorly soluble pharmaceuticals (Figure 3). Within this framework, biomaterials defined as substances used in medical applications to support, enhance, or replace damaged tissues or biological functions are of paramount importance. These biomaterials can be classified as natural or synthetic and can be derived from diverse sources, including metals, ceramics, polymers, glass, and even living cells and tissues. Frequently utilized biomaterials in the formulation of nanosuspensions encompass stabilizers, polymers (natural and synthetic), surfactants, buffers, and complexing agents.28,44 Table 1 presents a comprehensive overview of the different biomaterials utilized in this context, detailing their specific functions and contributions to improving the performance of nanosuspensions in cancer therapy.
![]() |
Table 1 Nanosuspension-Based Biomaterials for Cancer Therapy |
![]() |
Figure 3 Formulation consideration of nanosuspensions. Created in BioRender. Suliman, K. (2025) https://BioRender.com/e55hkfu. |
According to published research, active pharmaceutical ingredients (APIs) with high log P and enthalpy values are more likely to stabilize nanosuspensions via both electrostatic and steric stabilization. Furthermore, physical characteristics, such as molecular weight, have not been shown to directly affect the stabilization stage or particle size.22,29 However, because of the high surface energy of nanosuspensions, the stabilizer plays a crucial role in preventing agglomeration or aggregation. Various particle size analysis techniques can be used to investigate variations in the polydispersity index and particle size distribution at different stages of nanosuspension, including during manufacturing, storage, and stability testing.28,44 Stabilizers also play an important role in preventing Ostwald ripening, wetting hydrophobic drug particles, and providing steric or ionic repulsion to create physically stable products. The in vivo stability of nanosuspensions and their physical stability are strongly influenced by the concentration of the stabilizer. Natural polymers like chitosan and alginate are favoured due to their biocompatibility, biodegradability, and controlled-release properties. These polymers have been used in formulations of paclitaxel and curcumin to enhance stability and bioavailability.21,23,32
The Table 2 summarizes the key differences between natural, synthetic, and hybrid nanosuspensions based on various properties. These properties highlight the advantages and limitations of each polymer type for drug delivery applications.
![]() |
Table 2 Comparison of Key Properties Across Natural, Synthetic, and Hybrid Nanosuspensions for Drug Delivery Applications |
Natural Polymer-Stabilized Nanosuspensions
Chitosan Nanosuspensions
Chitosan is a biopolymer product produced from chitin, and it has drawn a considerable interest for its superior film-forming properties and its capacity to stabilize nanosuspensions. This multifunctional organic molecule is essential for protecting the stability of nanosuspensions and guaranteeing their homogeneous dispersion. Likewise, alginate, a polysaccharide derived from brown algae, enhances the stability of nanosuspensions by creating a protective layer around them. The encapsulation of nanosuspensions serves the dual purpose of protecting them and improving their thermal stability, hence establishing alginate as a highly useful constituent in nanosuspension formulations. Furthermore, the efficacy of gelatin, derived from collagen, in stabilizing nanosuspensions has been thoroughly investigated. Its unique properties enable it to specifically interact with nanosuspensions, thereby providing a critical stabilizing effect that is essential for maintaining the integrity and functionality of the nanosuspension system.1,6
Several authors have successfully created chitosan-based porous nanofibers by combining chitosan with polyethylene oxide (PEO) followed by electrospinning and dehydration. They were observed using scanning electron microscopy to obtain porous morphological, Chitosan-based porous nanofibers in nanosuspension systems hold significant promise for cancer treatment, providing a targeted and controlled drug delivery platform. These systems are characterized by excellent biocompatibility and stability, with the added potential for intrinsic anticancer effects.71–77 The nanofibers were immersed in a 0.1% w/w solution of paclitaxel to load the medication. For encapsulation, porous chitosan nanofibers were submerged in a 4% w/w solution of polyanion-type hyaluronic acid. Differential scanning calorimetry, or DSC, and Fourier transform infrared spectroscopy were used. The analysis of paclitaxel release from the encapsulated fibers in PBS was conducted using UV-visible spectroscopy.76,78–80
The in vitro activities against DU145 cancer cells treated with the loaded nanofibers were assessed using the MTT assay. This information provides compelling evidence that the chitosan/hyaluronic acid fibers influenced the pace at which paclitaxel was released and may have applications in postoperative treatment. For the intracellular delivery of anticancer drugs, chitosan-Tripolyphosphate (TPP) nanosuspensions have shown strong potential.71,72,76,77,81 These nanosuspensions have garnered significant attention in the field of drug delivery due to their unique combination of biocompatibility, biodegradability, and mucoadhesive properties. The nanoparticles are commonly prepared via ionic gelation, wherein positively charged chitosan molecules interact with negatively charged sodium tripolyphosphate (TPP). This interaction leads to the spontaneous formation of stable nanoparticles that are well-suited for encapsulating a wide range of therapeutic agents.82–85 One of the most valuable attributes of chitosan-TPP nanoparticles is their strong mucoadhesive property, which markedly enhances drug absorption across mucosal tissues. This adhesive interaction is most effective when the chitosan-to-TPP ratio is optimized. Studies have indicated that a ratio of 4:1 (chitosan:TPP) produces nanoparticles with favorable size and surface charge characteristics, facilitating prolonged retention at the absorption site and thereby improving therapeutic efficacy.71,72,77,81,86
Alginate-Based Nanosuspensions
Alginate-based nanosuspensions are colloidal systems formed from the biopolymer sodium alginate, widely used for stabilizing hydrophobic drugs like Paclitaxel and Docetaxel, which have poor water solubility and bioavailability. By encapsulating these drugs in alginate nanoparticles, their solubility is enhanced, and their stability is improved, preventing degradation and crystallization. These nanosuspensions are especially valuable in drug delivery systems, where they not only enhance solubility but also enable controlled or sustained drug release. Recent advancements have extended their applications to multi-drug delivery systems, where they can co-deliver synergistic drug combinations, improving therapeutic outcomes by offering enhanced efficacy, reduced side effects, and more precise targeting of diseased tissues, such as cancer cells. The ability to combine multiple drugs in a single nanosuspension offers potential benefits like reduced dosage and toxicity. However, challenges remain, including scalability, high drug loading, and regulatory hurdles, which need to be addressed for broader clinical application. Despite these challenges, ongoing research continues to improve alginate-based nanosuspensions, making them a promising strategy for the future of drug delivery, particularly in the treatment of complex diseases like cancer.46
Gelatin-Based Nanosuspensions
Gelatin nanoparticles facilitate tumor-specific targeting of drugs such as Cisplatin and Paclitaxel. Current research has focused on the creation of gelatin-based nanosuspensions that enhance drug delivery efficacy and reduce toxicity in tumor tissues.47
Hyaluronic Acid-Based Nanosuspensions
Hyaluronic acid-based nanosuspensions are effective for targeting tumors with high CD44 expression. This polymer enhances the specificity of nanoparticles to CD44+ cancer cells, as seen with Doxorubicin-loaded systems, ensuring higher therapeutic efficiency and lower off-target effects.48
Dextran-Based Nanosuspensions
Dextran- based nanosuspensions are pivotal in cancer immunotherapy and targeted treatments. Methotrexate-loaded dextran systems have been refined for superior tumor targeting, with recent studies showing enhanced delivery precision and reduced systemic toxicity.49
Collagen - Based Nanosuspensions
Collagen nanosuspensions are increasingly used for carrying anticancer agents like Curcumin. Research has shown these carriers improve the therapeutic impact of drugs while providing sustained-release properties, making them useful in cancer treatment and tissue engineering.50
Starch Nanosuspensions
Starch nanosuspensions are employed for the controlled release of hydrophobic drugs like 5-Fluorouracil. Current advancements emphasize localized drug delivery systems, which provide sustained release at the tumor site, reducing systemic side effects.51
Silk Fibroin-Based Nanosuspensions
Silk fibroin-based nanosuspensions are being developed for sustained and targeted drug delivery, as demonstrated with Paclitaxel. These systems show improved therapeutic efficacy in tumor models, making them a valuable tool in modern oncological applications.52
Synthetic Polymer-Stabilized Nanosuspensions
Poloxamers, such as Poloxamer 188 and Poloxamer 407, are synthetic macromolecules commonly employed for the purpose of stabilizing nanosuspensions. The FDA has classified them as generally regarded as safe (GRAS) for pharmaceutical purposes, encompassing oral, parenteral, and topical usage.6
Poly (lactic-co-glycolic acid) (PLGA) is a novel substance employed in drug delivery systems based on nanoparticle engineering. Pharmacological bioavailability is improved and regulated release is facilitated by PLGA-based nanosuspensions.28,42–44,87–91
Poloxamers
The FDA has deemed these synthetic polymers to be generally recognized as safe (GRAS) for topical, parenteral, and oral pharmaceutical use. Poloxamers 188 and 407 are the most commonly used polymers with stabilizing effects in nanosuspensions. The primary determinants of the stability and formation of nanosuspension crystals include the hydrophilic-lipophilic ratio, shape, functional groups, and molecular weight. In cancer therapy, Poloxamers are frequently employed to improve the delivery of poorly water-soluble drugs, enhance drug stability, and extend circulation duration. Nanosuspensions stabilized with Poloxamers facilitate targeted drug delivery, minimize adverse effects, and optimize therapeutic efficacy by regulating the release of anticancer compounds.92
Additionally, Poloxamers possess the ability to modify cell membrane permeability, which can further enhance drug absorption and promote cellular uptake, making them advantageous in the formulation of cancer treatments.2,13,27,44
PLGA Nanosuspensions
PLGA is a biodegradable copolymer frequently used in the formulation of nanosuspensions. It is hydrolyzed inside the body to produce water and CO2, as well as the safe and non-toxic metabolites lactic acid and glycolic acid. These two primary metabolites have very little systemic toxicity because they can easily be eliminated.36,54–56,78,93,94 Several techniques, including oil-in-water emulsion solvent evaporation, nanoprecipitation, and interfacial deposition, have been used to create PLGA nanosuspensions loaded with paclitaxel. Generally, the release of paclitaxel from the nanosuspension follows a biphasic pattern, with a rapid initial release rate occurring over a period of one–three days, followed by steady, persistent.73–76,79,95,96 It has been demonstrated that these nanosuspensions have higher in vitro cytotoxicity than free paclitaxel in a variety of tumor cell lines, including HeLa, glioma C6, and human small-cell lung cancer. Paclitaxel-loaded nanosuspensions demonstrated significantly improved in vivo tumor-inhibitory effects in transplantable liver tumors. The modified surface of the nanosuspensions resulted in enhanced delivery of paclitaxel.37,74,97,98
Poly(Lactide) Nanosuspension (PLA NSs)
PLA is another popular matrix used to synthesize polymeric nanosuspension, because of its safe and biodegradable characteristics. To provide NSs with long-circulating qualities, a methoxy poly (ethylene glycol)-poly(lactide) copolymer (mPEG-PLA) was created and added, PLA nanosuspensions are a promising platform in nanomedicine for cancer therapy due to their biodegradability, biocompatibility, and ability to enhance the therapeutic efficacy of anticancer drugs while minimizing side effects.57,58,99–102
Amino Acid-Based Stabilizers
It has been shown that leucine copolymers can effectively form stable drug nanocrystals in aqueous media. Lecithin is the recommended stabilizing agent for sterile, steam-heat-sterilizable parenteral nanosuspensions. Albumin has been used in nanosuspensions at concentrations ranging from 0.003% to 5% as a surface-stabilizing and drug-targeting agent. Proline, transferrin, and arginine are other pharmaceutically acceptable amino acid copolymers utilized to stabilize the physical state of nanocrystals.3,5,17,22,91
Leucine, Proline, Arginine
These amino acid-based stabilizers enhance drug solubility and personalization of cancer treatments, especially with agents like Curcumin. Advancements in precision medicine have incorporated these stabilizers to improve nanosuspension stability and therapeutic outcomes.59
Albumin
Albumin-based systems, such as Abraxane® (Paclitaxel albumin-bound nanosuspension), have revolutionized controlled drug release and tumor targeting. Recent innovations focus on optimizing these formulations for enhanced drug solubility and tumor penetration.60
Cellulose Based Derivatives
Hydroxypropyl cellulose (HPC), hydroxyethyl cellulose (HEC), and hydroxypropyl methyl cellulose (HPMC) are frequently used as stabilizing agents. The surface-adsorbed hydrophobic groups of these polymers constitute the fundamental mechanism for their steric stabilization.12,14–19
Hybrid Polymer-Stabilized Nanosuspensions
The aim of coupled hybrid nanosuspensions is to improve the stability of dispersion by combining different types of nanoparticle categories. Nanoparticle aggregation is hindered by the chemical bonding that occurs throughout the formulation process. Drug molecules or genetic material can be integrated into graphene oxide (GO) nanoplatelets to get highly specific and regulated administration. Polymeric lipid hybrid nanoparticles are formed by combining lipid vesicles with polymeric core components. The development of hybrid nanosuspensions covered with cell membranes involves the integration of lipid vesicles with polymeric core components, presenting a novel and sophisticated approach for drug delivery.87–91,103–106
Miscellaneous
Developed by BASF Industries, Soluplus® is a new excipient consisting of a copolymer of polyvinyl caprolactam, polyvinyl acetate, and PEG. Several nanosuspensions with improved stability, accelerated dissolution rate, and greater bioavailability have been utilized as stabilizers. It has been observed that the use of water-soluble polymers such as PVA, PEGylated chitosan, and polyvinyl pyrrolidone as stabilizers greatly increases the rate of dissolution and bioavailability of nanosuspensions.88 A protein-based surfactant, hydrophobin, was used to create a functionalized surface coating for the low-solubility medication, beclomethasone dipropionate. Its adaptability to genetically engineered surface modification makes it appropriate for a variety of drug delivery applications.88,90
Nanosuspensions using polymers such as PTX is a unique approach that increases drug bioavailability and reduces the likelihood of side effects. A variety of synthetic and natural polymers, such as chitosan NSs, PLA NSs, and PLGA NSs, have been employed for PTX nanoencapsulation. The general characteristics of these polymers include regulated drug release, biodegradability, biocompatibility, and ease of manipulation of their physicochemical properties.37,107–110
Concurrent chemotherapy often fails due to the presence of cancer stem cells (CSCs) and the development of drug resistance, driven in part by altered microRNA expression within tumors One of the key contributors to this resistance is the dysregulation of signalling pathways, such as the hedgehog (Hh). To overcome this, combining paclitaxel with the Hh inhibitor cyclopamine (CYP) has been shown to effectively target drug-resistant cancer cells. Additionally, inhibition is a subset of cells called side populations, which are common in cancer stem cells. Researcher synthesized mPEG-b-PCC-g-PTX-g-DC (P-PTX) and mPEG-b-PCC-g-CYP-g-DC (P-CYP) PTX–polymer conjugates, forming micelles.76,108,111 This combination prevented the growth of tumor colonies and helped fighting drug resistance to paclitaxel. Additionally, this combination inhibited Hh signaling and increased the expression of tumor suppressor miRNAs.86,112–115
Surfactant and Co-Surfactants
The choice of surfactant and co-surfactant is crucial for creating nanosuspensions, with microemulsions serving as the model. This may have an impact on drug loading in the internal phase, drug solubility, and phase behavior. For the stabilization and development of nanosuspensions, a variety of surfactants, including Tweens and sodium dodecyl sulfate, as well as co-surfactants, including bile salts, Transcutol, glycofurol, ethyl alcohol, and isopropyl alcohol, have been effectively employed, surfactants and co-surfactants are integral to nanosuspension systems, including those used for cancer therapy, as they improve stability, solubility, and delivery efficiency.88
D-α-Tocopheryl Polyethylene Glycol 1000 Succinate (Vitamin E TPGS)
An esterified, water-soluble derivative of vitamin E (tocopherol), vitamin E polyethylene glycol succinate, has been utilized as a stabilizing and solubilizing ingredient in several nanosuspension formulations. Because of its high physical stability and low toxicity profile, it is considered the best excipient for parenteral, ophthalmic, and oral use.17,42,87
Hydrophobin
Hydrophobin-modified nanocarriers are being explored for their ability to optimize drug delivery systems. These carriers have shown promise in sustained release applications for drugs such as Doxorubicin, improving cancer therapy outcomes.66
Nanosuspensions in Targeted Anticancer Treatment
Nanosuspension-based Biomaterial Drug Delivery Systems for Cancer (NBDDSC) represent an innovative approach to targeted cancer therapy, specifically designed to improve the delivery of poorly soluble anticancer drugs (Figure 4). By leveraging nanoscale technology, these systems enhance drug solubility, bioavailability, and precision, ensuring more effective delivery to tumor sites while minimizing adverse effects on healthy tissues. Integrating nanotechnology with biocompatible materials, NBDDSC provides a promising, patient-centered solution for developing safer and more effective cancer treatments. In terms of targeting strategies, nanosuspension-based biomaterials utilize the enhanced permeability and retention (EPR) effect for passive targeting, allowing nanoparticles to accumulate in tumor tissues due to leaky vasculature and impaired lymphatic drainage. Active targeting, in contrast, involves functionalizing nanoparticles with specific ligands that can recognize and bind to receptors overexpressed on cancer cells, thereby improving the precision of drug delivery. Combining passive and active targeting strategies can further enhance overall targeting efficiency, increasing therapeutic efficacy while reducing systemic side effects.116
![]() |
Figure 4 Nanosuspensions in targeted anti-cancer treatment. |
Hybrid Membrane Coated Nanosuspensions for Anti-Glioma Therapy
With a median life span of less than 24 months and a 5-year survival rate of approximately 2–4%, glioma is one of the worst tumors that affects humans. In an effort to improve patient outcomes, doctors and scientists working on combinatorial therapies, particularly the combination of chemotherapy and immunotherapy, have expressed great interest in this short survival expectation.117,118 While trials (on both female and male ICR mice) combining immunotherapy and chemotherapy have shown promise in treating other tumors, glioma remains uniformly fatal, with little improvement in overall survival. In contrast to other tumors, gliomas are characterized by their isolation behind the blood-brain tumor barrier (BBTB) and physiological barrier (BBB).119 They also exhibit a higher mutational burden and heterogeneity while impairing the immune function due to both the tumor’s own growth and the unique environment of the brain. Owing to these restrictions, relatively little chemotherapy and immune checkpoint blockade medication actually reach the glioma.120 Therefore, in addition to the development of chemotherapeutic and immunotherapeutic drugs, the treatment of glioma has several challenges resulting from the lack of a potent platform that might deliver these therapeutic agents to the targeted areas.121–123
The co-distribution of these therapeutic agents has been the subject of numerous NBDDSC to date; yet, the US Food and Drug Administration has not approved any of these systems for the treatment of disorders affecting the central nervous system (CNS).124 Currently, practically all NBDDSC rely primarily on the properties of the materials they are composed of target tumors; nevertheless, these compounds have no inherent therapeutic activity and solely act as excipients. Customized materials and co-loading of many therapeutic agents sometimes entail intricate manufacturing procedures, which can lead to fluctuations in the reproducibility of batches and diminish the stability of non-digestible drug delivery systems, frequently culminating in inadequate therapeutic results. Furthermore, these systems are constrained by the inherent drawbacks of synthetic materials, including their possible negative side effects. Thus, the creation of a straightforward, safe, and effective delivery system is crucial for clinical translation.125
The study of biomimetic nanocarriers is being advanced by the new NBDDSC frontiers. Among these, cell membrane-coated nanosuspensions have been extensively studied for their potential to treat cancer. The potential uses range from improving the immunogenicity of cancer vaccines to improving the efficacy of cancer medicine delivery.126 These types of biomimetic NSs for drug delivery display distinct roles from the source cells, offering a different approach to overcome biological barriers and increase the effectiveness of drug administration. Unfortunately, many synthetic materials are needed to create a scaffold for these biomimetic NSs, which frequently leads to low drug-loading rates.125
An enhanced tumor-inhibitory effect can be attained by increasing the drug concentration at the target site through high drug loading. In study, researchers applied a cell membrane coating to nanosuspensions. Like a “pure drug particle”. Nanosuspensions are easily transferred to the clinical setting and have a high drug carrying capacity because they do not contain a carrier, which helps to increase drug concentration at the targeted areas127 Cell membrane-bound tumor antigens are not only employed for drug administration; they are also used to teach the immune system how to identify and combat cancer. Previous studies have produced mimic particle carriers that have been altered to include these surface antigens, which has increased the effectiveness of vaccines.128 Dendritic cells (DCs) are antigen-presenting cells that activate many immune resistance mechanisms. Tumor-associated antigens can stimulate mature DCs to increase their number and activate CD8+ and CD4+ T cells. T cells are selectively recognized by activated CD8+ T lymphocytes (CTLs), which then trigger tumor death.127–134
Additionally, major histocompatibility complex (MHC) molecules are expressed more often on the surface of tumor cells when CD4+ helper T lymphocytes are activated. This helps CTLs identify tumor cells and accelerate the death of cancer cells.129,135 DC-based immunotherapy has recently attracted considerable interest. In patients with glioblastoma, melanoma, renal cell carcinoma, and prostate and ovarian malignancies, DC vaccinations have been shown to be both safe and efficacious.49,136,137 Unfortunately, the effectiveness of these vaccinations still falls short of predictions based on basic science and evidence from clinical trials conducted over the past 20 years. Two primary factors were found responsible for this failure (1) the majority of tumor DC vaccines focus on a single antigen; however, tumor cells can quickly evade immune responses by mutating their antigen. (2) The body’s proteolytic enzymes readily break down the extracted tumor proteins, making it difficult to obtain the optimal treatment outcome and produce immunological effects that are not long-lasting. This complicates the extraction procedure.138–143 Consequently, enhanced stability and effective antigen loading are crucial for optimizing DC vaccines. Generally, a wide range of antigens, including tumor-associated and tumor-specific antigens, are present in cancer cells. High concentrations of tumor antigens in cancer vaccines are necessary to cross the threshold for T-cell recognition and end immunological tolerance.119,123,127
Nanosuspensions have shown promising therapeutic potential not only in glioma but also across various other cancer types. For instance, doxorubicin and methotrexate nanosuspensions have been effectively utilized in the treatment of breast and lung cancers, demonstrating improved drug solubility and extended circulation times. These nanosuspension formulations also contribute to reduced systemic toxicity by facilitating more targeted delivery to tumor sites. Furthermore, nanosuspensions hold promise in overcoming multidrug resistance (MDR) by enhancing drug delivery to cancer cells and increasing the intracellular accumulation of chemotherapeutic agents.144
Paclitaxel Nanosuspensions
Nanosuspensions are submicron colloidal dispersions containing nanosized drug particles stabilized by surfactants. These formulations suspend poorly water-soluble drugs directly in a liquid medium without incorporating a matrix material, enhancing their solubility in both lipid and aqueous environments.6 Improved solubility allows the API to achieve peak plasma levels more rapidly due to an accelerated dissolution rate.145 Nanosuspensions are particularly advantageous for drugs with limited solubility, permeability, or both, and can be administered intravenously without blocking blood vessels because of the small particle size.146
In a recent study, Fan et al developed PTX nanosuspensions ((PTX)NS) coated with glioma C6 cancer cell membrane (CCM) and modified with a DWSW peptide (DS DY DP DG DW DS DW), which is a D-peptide with enhanced proteolytic stability and the ability to penetrate biological barriers. These nanosuspensions were fabricated using an ultrasonic precipitation technique and demonstrated capabilities to penetrate the blood-brain barrier (BBB) and target tumor sites. This occurs through various mechanisms, such as receptor mediated endocytosis, enhanced permeation through tight junctions, and active targeting via surface modification with ligands. Drugs like paclitaxel and curcumin, when delivered in nanosuspension form, have demonstrated enhanced brain bioavailability, which is crucial for treating central nervous system (CNS) diseases, including gliomas. However, there remains a need for more extensive in vivo studies to fully understand the extent of BBB penetration and its clinical significance147 The cancer cell membrane coating camouflaged the nanosuspension, facilitating immune evasion and BBB crossing, leading to a preferential accumulation in glioma tissues. In glioma-bearing mice, DWSW-CCM-(PTX)NS significantly extended survival and inhibited tumor growth. This biomimetic approach conferred biological properties such as homologous adhesion and immune evasion to the nanosuspension, underscoring its potential in targeted tumor therapies and offering a comprehensive strategy to enhance nanosuspension targeting.148
Challenges and Limitations
Despite the promising potential of NBDDSC, several significant challenges hinder their clinical translation. These include toxicity concerns, solubility and bioavailability limitations, reproducibility and batch-to-batch variability, manufacturing scalability, and complex regulatory requirements Table 3.
![]() |
Table 3 Summary of Key Challenges and Potential Solutions |
Toxicity Concerns
Remain paramount, especially in oncology, where non-specific drug distribution can exacerbate systemic side effects. Commonly used synthetic excipients such as PLGA and PLA degrade into acidic byproducts, which may lead to localized inflammation and tissue damage.57 Similarly, surfactants like SDS may induce hemolysis and cytotoxicity at higher concentrations. The mononuclear phagocyte system (MPS) uptake of nanosuspensions also leads to off-target accumulation in organs such as the liver and spleen, further increasing toxicity risks. Immunogenicity issues related to synthetic materials can additionally impair treatment efficacy upon repeated administration.60
Solubility and Bioavailability Challenges
Many anticancer agents suffer from poor aqueous solubility and low oral bioavailability, which hinders their therapeutic efficacy when formulated as nanosuspensions. Drugs with high lipophilicity often require solubilizing agents or surface modifiers, which can introduce toxicity or stability issues. Furthermore, limited mucosal permeability and first-pass metabolism in oral delivery systems restrict drug absorption. The physicochemical characteristics of nanosuspensions such as particle size, surface charge, and crystallinity significantly affect dissolution rate and membrane permeability, and hence must be carefully optimized for improved bioavailability.43
Strategies and Potential Solutions
To address toxicity, biodegradable and biocompatible materials like chitosan, alginate, gelatin, and silk fibroin have been explored. These natural polymers degrade into non-toxic byproducts and can exhibit additional therapeutic benefits such as anti-inflammatory and antimicrobial properties.74 Hybrid nanosystems, such as cell membrane-coated nanoparticles, can mimic tumor cells to evade immune clearance and enhance target specificity, reducing off-target accumulation and systemic toxicity. Notably, paclitaxel-loaded cell membrane-coated nanosuspensions have demonstrated superior tumor selectivity in preclinical models.149
To improve reproducibility, technologies like microfluidics, microfluidization, and sonocrystallization offer precise control over particle synthesis and reduce batch variability. These methods enable uniform particle size distribution, stable drug encapsulation, and seamless scale-up31 Integration of real-time quality monitoring systems (eg, in-line spectroscopy, particle size analyzers) also ensures critical attributes are maintained throughout production.43
To ensure regulatory compliance, developers must implement standardized protocols for toxicity testing and conduct robust long-term safety studies. Use of biodegradable materials and consistent production parameters helps meet FDA and EMA expectations for safety, efficacy, and environmental compatibility.152
Scale-Up Challenges
The transition from lab-scale to industrial production introduces unique scale-up challenges. Techniques like wet milling and freeze drying, though efficient in small-scale settings, often suffer from aggregation and poor energy efficiency when scaled. High-pressure homogenization requires multiple processing cycles, raising concerns around cost, yield, and processing time. Furthermore, achieving batch-to-batch uniformity is essential for regulatory approval, requiring the integration of automated systems and process analytical technologies to ensure reproducibility and quality at scale.152
Conclusion
Nanosuspensions represent a transformative approach in drug delivery, offering significant solutions to the longstanding challenges of poor solubility and bioavailability, particularly in cancer therapy. Literature has consistently shown that poorly water-soluble drugs often exhibit low bioavailability, limiting their therapeutic potential. Nanosuspensions effectively enhance solubility by reducing particle size, thus improving dissolution rates and bioavailability. By addressing toxicity concerns through biodegradable polymers and improving reproducibility with advanced manufacturing techniques, nanosuspensions have emerged as a promising tool to minimize systemic toxicity while delivering higher concentrations of drugs to the target site.
Additionally, the use of natural, synthetic, and hybrid polymers in nanosuspensions has shown great promise in tailoring release profiles and targeting specific cancer cells. Among these, hybrid polymer-based nanosuspensions, particularly those combining natural polymers like chitosan with synthetic ones like PLGA, have emerged as the most versatile and efficient for various cancer types. These systems not only enhance drug stability and bioavailability but also allow for improved targeting and controlled release, making them highly promising for personalized cancer therapy.
The best preparation methods for different cancer drug types vary, with techniques like high-pressure homogenization, microprecipitation, and solvent evaporation proving effective for both hydrophobic and hydrophilic drugs. For example, drugs like paclitaxel and doxorubicin, which are commonly used in cancer treatment, benefit from these methods as they enhance their solubility and ensure sustained release. Tailoring the preparation method based on the drug’s physicochemical properties and targeting requirements is crucial for maximizing therapeutic efficacy.
Looking ahead, future research should focus on improving the targeting efficiency of nanosuspensions, particularly in overcoming the blood-brain barrier (BBB) for brain cancer therapies and enhancing drug delivery to multidrug-resistant cancer cells. Furthermore, exploring the combination of nanosuspensions with other novel drug delivery systems, such as immunotherapies and gene therapies, could pave the way for innovative treatments. As for future directions in the field of NBDDSs (Nanostructured Drug Delivery Systems), expanding their applications beyond cancer, including neurological disorders, infectious diseases, and regenerative medicine, holds immense promise. Advancements in scale-up production, long-term stability, and patient-specific formulations will be key to their successful clinical translation.
Acknowledgment
We would like to thank The Rector of Universitas Padjadjaran for the APC . Also, we would like to thank The Libyan Ministry of Higher Education, and Elmergib University for a doctoral scholarship. The authors would also like to extend their appreciation to Northern Border University, Saudi Arabia, for supporting this work through project number (NBU-CRP-2025-2292).
Funding
This work was supported by Penelitian Disertasi Doktor grant from the Indonesian Ministry of Higher Education, Science and Technology (093/C3/DT.05.00/PL/2025).
Disclosure
The authors declare no conflicts of interest in this work.
References
1. Aher SS, Malsane ST, Saudagar RB. Nanosuspension: an overview. Asian J Res Pharm Sci. 2017;7(2):81–86.
2. Yadollahi R, Vasilev K, Simovic S. Nanosuspension technologies for delivery of poorly soluble drugs. J Nanomater. 2015;2015(1):216375. doi:10.1155/2015/216375
3. Pawar SS, Dahifale BR, Nagargoje SP, Shendge RS. Nanosuspension technologies for delivery of drugs. Nanosci Nanotech Res. 2017;4(2):59–66.
4. Kumar Singh S, Vaidya Y, Gulati M, Bhattacharya S, Garg V, Kumar Pandey N. Nanosuspension: principles, perspectives and practices. Curr Drug Deliv. 2016;13(8):1222–1246. doi:10.2174/1567201813666160101120452
5. Kulkarni RR, Phadtare DG, Saudagar RB. A novel approach towards nanosuspension. Asian J Pharm Res. 2015;5(4):186–194. doi:10.5958/2231-5691.2015.00029.5
6. Jacob S, Nair AB, Shah J. Emerging role of nanosuspensions in drug delivery systems. Biomater Res. 2020;24:1–16. doi:10.1186/s40824-020-0184-8
7. Khandbahale SV. A review-Nanosuspension technology in drug delivery system. Asian J Pharm Res. 2019;9(2):130–138. doi:10.5958/2231-5691.2019.00021.2
8. Hitanga J, Sharma N, Chopra H, Kumar S. Nanoprecipitation technique employed for the development of nanosuspension: a review. WJPPS. 2015;4:2127–2136.
9. Wang L, Du J, Zhou Y, Wang Y. Safety of nanosuspensions in drug delivery. Nanomed Nanotechnol Biol Med. 2017;13(2):455–469. doi:10.1016/j.nano.2016.08.007
10. Jayaprakash R, Krishnakumar K, Dineshkumar B, Jose R, Nair SK. Nanosuspension in drug delivery-A review. Sch Acad J Pharm. 2016;5:138–141.
11. Zhang J, Wang S, Zhang D, et al. Nanoparticle-based drug delivery systems to enhance cancer immunotherapy in solid tumors. Front Immunol. 2023;14:1230893.
12. Arora D, Khurana B, Rath G, Nanda S, Goyal AK. Recent advances in nanosuspension technology for drug delivery. Curr Pharm Des. 2018;24(21):2403–2415. doi:10.2174/1381612824666180522100251
13. Chinthaginjala H, Abdul H, Reddy APG, Kodi K, Manchikanti SP, Pasam D. Nanosuspension as promising and potential drug delivery: a review. Int J Life Sci Pharm Res. 2020;11(1):P59–66.
14. Carvalho D de M, Takeuchi KP, Geraldine RM, de MCJ, Torres MCL. Production, solubility and antioxidant activity of curcumin nanosuspension. Food Sci Technol. 2015;35:115–119. doi:10.1590/1678-457X.6515
15. I Jethara S, D Patel A, R Patel M, S Patel M, R Patel K. Recent survey on nanosuspension: a patent overview. Recent Pat Drug Deliv Formul. 2015;9(1):65–78. doi:10.2174/1872211308666141028214003
16. Gajera BY, Shah DA, Dave RH. Development of an amorphous nanosuspension by sonoprecipitation-formulation and process optimization using design of experiment methodology. Int J Pharm. 2019;559:348–359. doi:10.1016/j.ijpharm.2019.01.054
17. Kovalchuk NM, Johnson D, Sobolev V, Hilal N, Starov V. Interactions between nanoparticles in nanosuspension. Adv Colloid Interface Sci. 2019;272:102020. doi:10.1016/j.cis.2019.102020
18. Goel S, Sachdeva M, Agarwal V. Nanosuspension technology: recent patents on drug delivery and their characterizations. Recent Pat Drug Deliv Formul. 2019;13(2):91–104. doi:10.2174/1872211313666190614151615
19. Sattar A, Chen D, Jiang L, et al. Preparation, characterization and pharmacokinetics of cyadox nanosuspension. Sci Rep. 2017;7(1):2289. doi:10.1038/s41598-017-02523-4
20. Kalvakuntla S, Deshpande M, Attari Z, Kunnatur K. Preparation and characterization of nanosuspension of aprepitant by H96 process. Adv Pharm Bull. 2016;6(1):83. doi:10.15171/apb.2016.013
21. Liu T, Müller RH, Möschwitzer JP. Effect of drug physico-chemical properties on the efficiency of top-down process and characterization of nanosuspension. Expert Opin Drug Deliv. 2015;12(11):1741–1754. doi:10.1517/17425247.2015.1057566
22. Jassim ZE, Rajab NA. Review on preparation, characterization, and pharmaceutical application of nanosuspension as an approach of solubility and dissolution enhancement. J Pharm Res. 2018;12:771–774.
23. Afifi SA, Hassan MA, Abdelhameed AS, Elkhodairy KA. Nanosuspension: an emerging trend for bioavailability enhancement of etodolac. Int J Polym Sci. 2015;2015(1):938594. doi:10.1155/2015/938594
24. Thattil JG, Kumar KK, Kumar BD. Nanosuspension technology in pharmaceuticals. J Bio Innov. 2018;7(2):660–677.
25. Pravin P, Adhikrao Y, Varsha G. Different techniques for preparation of nanosuspension with reference to its characterisation and various applications-a review. Asian J Res Pharm Sci. 2018;8(4):210–216.
26. Chen A, Shi Y, Yan Z, et al. Dosage form developments of nanosuspension drug delivery system for oral administration route. Curr Pharm Des. 2015;21(29):4355–4365. doi:10.2174/1381612821666150901105026
27. Pınar SG, Oktay AN, Karaküçük AE, Çelebi N. Formulation strategies of nanosuspensions for various administration routes. Pharmaceutics. 2023;15(5):1520. doi:10.3390/pharmaceutics15051520
28. Guan W, Ma Y, Ding S, et al. The technology for improving stability of nanosuspensions in drug delivery. J Nanopart Res. 2022;24(1):14. doi:10.1007/s11051-022-05403-9
29. Patel HM, Patel UB, Shah C, Akbari B. Formulation and development of nanosuspension as an alternative approach for solubility and dissolution enhancement of aceclofenac. Int J Adv Pharm. 2018;7(5):33–47.
30. Zhang J, Xie Z, Zhang N, Zhong J. Nanosuspension drug delivery system: preparation, characterization, postproduction processing, dosage form, and application. Nanostructures Drug Delivery. 2017;2017:413–443.
31. Tian Y, Wang S, Yu Y, et al. Review of nanosuspension formulation and process analysis in wet media milling using microhydrodynamic model and emerging characterization methods. Int J Pharm. 2022;623:121862. doi:10.1016/j.ijpharm.2022.121862
32. Patel HM, Patel BB, Shah CN, Shah DP. Nanosuspension technologies for delivery of poorly soluble drugs-a review. Res J Pharm Technol. 2016;9(5):625–632. doi:10.5958/0974-360X.2016.00120.7
33. Purkayastha HD, Hossian SKI. Nanosuspension: a modern technology used in drug delivery system. Int J Curr Pharm Res. 2019;11(3):1–3. doi:10.22159/ijcpr.2019v11i3.34098
34. Ferrar JA, Sellers BD, Chan C, Leung DH. Towards an improved understanding of drug excipient interactions to enable rapid optimization of nanosuspension formulations. Int J Pharm. 2020;578:119094. doi:10.1016/j.ijpharm.2020.119094
35. Velmula M, Pavuluri P, Rajashekar S, Rao VUM. Nanosuspension technology for poorly soluble drugs-a review. World J Pharm Pharm Sci. 2015;4(7):1612–1625.
36. Li X, Yuan H, Zhang C, et al. Preparation and in-vitro/in-vivo evaluation of curcumin nanosuspension with solubility enhancement. J Pharm Pharmacol. 2016;68(8):980–988. doi:10.1111/jphp.12575
37. Ahmad J, Ahmad MZ, Akhter H, Akhter H. Surface-engineered cancer nanomedicine: rational design and recent progress. Curr Pharm Des. 2020;26(11):1181–1190. doi:10.2174/1381612826666200214110645
38. Ahmadi Tehrani A, Omranpoor MM, Vatanara A, Seyedabadi M, Ramezani V. Formation of nanosuspensions in bottom-up approach: theories and optimization. DARU J Pharm Sci. 2019;27:451–473. doi:10.1007/s40199-018-00235-2
39. Agarwal V, Bajpai M. Nanosuspension technology for poorly soluble drugs: recent researches, advances and patents. Recent Pat Nanotechnol. 2015;9(3):178–194. doi:10.2174/1872210510999151126112644
40. Sumathi R, Tamizharasi S, Sivakumar T. Formulation and evaluation of polymeric nanosuspension of naringenin. Int J Appl Pharm. 2017;9(6):60–70. doi:10.22159/ijap.2017v9i6.21674
41. Dekate S, Bhairy S, Hirlekar R, Hirlekar R, Hirlekar R. Preparation and characterization of oral nanosuspension loaded with curcumin. Int J Pharm Pharm Sci. 2018;10:90–95. doi:10.22159/ijpps.2018v10i6.22027
42. Goutam SP, Saxena G, Roy D, Yadav AK, Bharagava RN. Green synthesis of nanoparticles and their applications in water and wastewater treatment. Bioremediation Ind waste Environ Saf Vol i Ind waste its Manag. 2020;2020:349–379.
43. Jahan N, Aslam S, ur RK, Fazal T, Anwar F, Saher R. Formulation and characterisation of nanosuspension of herbal extracts for enhanced antiradical potential. J Exp Nanosci. 2016;11(1):72–80. doi:10.1080/17458080.2015.1025303
44. Chakravorty R. Nanosuspension as an emerging Nanotechnology and Techniques for its Development. Res J Pharm Technol. 2022;15(1):489–493. doi:10.52711/0974-360X.2022.00079
45. Grewal AK, Salar RK. Chitosan nanoparticle delivery systems: an effective approach to enhancing efficacy and safety of anticancer drugs. Nano TransMed. 2024;3:100040. doi:10.1016/j.ntm.2024.100040
46. Markeb AA, El-Maali NA, Sayed DM, et al. Synthesis, structural characterization, and preclinical efficacy of a novel paclitaxel‐loaded alginate nanoparticle for breast cancer treatment. Int J Breast Cancer. 2016;2016(1):7549372. doi:10.1155/2016/7549372
47. Joukhan A, Kononenko V, Sollner Dolenc M, Hočevar M, Turk T, Drobne D. Modulation of the effect of cisplatin on nicotine-stimulated A549 lung cancer cells using analog of marine sponge toxin loaded in gelatin nanoparticles. Nanomaterials. 2024;14(9):777. doi:10.3390/nano14090777
48. Ashrafizadeh M, Mirzaei S, Gholami MH, et al. Hyaluronic acid-based nanoplatforms for Doxorubicin: a review of stimuli-responsive carriers, co-delivery and resistance suppression. Carbohydr Polym. 2021;272:118491. doi:10.1016/j.carbpol.2021.118491
49. Khan MS, Gowda BHJ, Nasir N, et al. Advancements in dextran-based nanocarriers for treatment and imaging of breast cancer. Int J Pharm. 2023;643:123276. doi:10.1016/j.ijpharm.2023.123276
50. de Sousa Victor R, Marcelo da Cunha Santos A, Viana de Sousa B, de Araújo Neves G, Navarro de Lima Santana L, Rodrigues Menezes R. A review on Chitosan’s uses as biomaterial: tissue engineering, drug delivery systems and cancer treatment. Materials. 2020;13(21):4995. doi:10.3390/ma13214995
51. Zhao B, Li L, Lv X, et al. Progress and prospects of modified starch-based carriers in anticancer drug delivery. J Control Release. 2022;349:662–678. doi:10.1016/j.jconrel.2022.07.024
52. Wani SUD, Zargar MI, Masoodi MH, et al. Silk fibroin as an efficient biomaterial for drug delivery, gene therapy, and wound healing. Int J Mol Sci. 2022;23(22):14421. doi:10.3390/ijms232214421
53. Mod Razif MRF, Chan SY, Widodo RT, et al. Optimization of a luteolin-loaded TPGS/Poloxamer 407 nanomicelle: the effects of copolymers, hydration temperature and duration, and freezing temperature on encapsulation efficiency, particle size, and solubility. Cancers. 2023;15(14):3741. doi:10.3390/cancers15143741
54. Gonzalez-Valdivieso J, Girotti A, Schneider J, Arias FJ. Advanced nanomedicine and cancer: challenges and opportunities in clinical translation. Int J Pharm. 2021;599:120438. doi:10.1016/j.ijpharm.2021.120438
55. Venkatasubbu GD, Ramasamy S, Gaddam PR, Kumar J. Acute and subchronic toxicity analysis of surface modified paclitaxel attached hydroxyapatite and titanium dioxide nanoparticles. Int J Nanomed. 2015;10(sup2):137–148. doi:10.2147/IJN.S79991
56. Ye YJ, Wang Y, Lou KY, Chen YZ, Chen R, Gao F. The preparation, characterization, and pharmacokinetic studies of chitosan nanoparticles loaded with paclitaxel/dimethyl-β-cyclodextrin inclusion complexes. Int J Nanomed. 2015;10:4309–4319. doi:10.2147/IJN.S83508
57. Bhatt H, Kiran Rompicharla SV, Ghosh B, Torchilin V, Biswas S. Transferrin/α-tocopherol modified poly (amidoamine) dendrimers for improved tumor targeting and anticancer activity of paclitaxel. Nanomedicine. 2019;14(24):3159–3176. doi:10.2217/nnm-2019-0128
58. Feng B, Niu Z, Hou B, Zhou L, Li Y, Yu H. Enhancing triple negative breast cancer immunotherapy by ICG‐templated self‐assembly of paclitaxel nanoparticles. Adv Funct Mater. 2020;30(6):1906605. doi:10.1002/adfm.201906605
59. Zaher S, Soliman ME, Elsabahy M, Hathout RM. Protein nanoparticles as natural drugs carriers for cancer therapy. Adv Tradit Med. 2023;23(4):1035–1064. doi:10.1007/s13596-022-00668-w
60. Liu Y, Li Y, Shen W, Li M, Wang W, Jin X. Trend of albumin nanoparticles in oncology: a bibliometric analysis of research progress and prospects. Front Pharmacol. 2024;15:1409163. doi:10.3389/fphar.2024.1409163
61. Elsebay MT, Eissa NG, Balata GF, Kamal MA, Elnahas HM. Nanosuspension: a formulation technology for tackling the poor aqueous solubility and bioavailability of poorly soluble drugs. Curr Pharm Des. 2023;29(29):2297–2312. doi:10.2174/1381612829666230911105922
62. Tarighatnia A, Foroughi-Nia B, Nader ND, Aghanejad A. Recent trends and advances in nanosystems with tyrosine kinase inhibitors for image-guided cancer treatments. J Drug Deliv Sci Technol. 2023;104938. doi:10.1016/j.jddst.2023.104938
63. Kumar G, Virmani T, Sharma A, Pathak K. Codelivery of phytochemicals with conventional anticancer drugs in form of nanocarriers. Pharmaceutics. 2023;15(3):889. doi:10.3390/pharmaceutics15030889
64. Kontogiannis O, Selianitis D, Lagopati N, Pippa N, Pispas S, Gazouli M. Surfactant and block copolymer nanostructures: from design and development to nanomedicine preclinical studies. Pharmaceutics. 2023;15(2):501. doi:10.3390/pharmaceutics15020501
65. Mehata AK, Setia A, Malik AK, et al. Vitamin E TPGS-based nanomedicine, nanotheranostics, and targeted drug delivery: past, present, and future. Pharmaceutics. 2023;15(3):722. doi:10.3390/pharmaceutics15030722
66. Osmanagaoglu FH, Ekmekcioglu A, Ozcan B, Bayram Akcapinar G, Muftuoglu M. Preparation and characterization of hydrophobin 4-coated liposomes for doxorubicin delivery to cancer cells. Pharmaceuticals. 2024;17(11):1422. doi:10.3390/ph17111422
67. Tuomela A, Hirvonen J, Peltonen L. Stabilizing agents for drug nanocrystals: effect on bioavailability. Pharmaceutics. 2016;8(2):16. doi:10.3390/pharmaceutics8020016
68. Vandelli MA, Romagnoli M, Monti A, et al. Microwave-treated gelatin microspheres as drug delivery system. J Control Release. 2004;96(1):67–84. doi:10.1016/j.jconrel.2004.01.009
69. Woodruff MA, Hutmacher DW. The return of a forgotten polymer—Polycaprolactone in the 21st century. Prog Polym Sci. 2010;35(10):1217–1256. doi:10.1016/j.progpolymsci.2010.04.002
70. Zhang K, Tang X, Zhang J, et al. PEG–PLGA copolymers: their structure and structure-influenced drug delivery applications. J Control Release. 2014;183:77–86. doi:10.1016/j.jconrel.2014.03.026
71. Zhang B, Shi W, Jiang T, et al. Optimization of the tumor microenvironment and nanomedicine properties simultaneously to improve tumor therapy. Oncotarget. 2016;7(38):62607. doi:10.18632/oncotarget.11546
72. Deng W, Qiu J, Wang S, et al. Development of biocompatible and VEGF-targeted paclitaxel nanodrugs on albumin and graphene oxide dual-carrier for photothermal-triggered drug delivery in vitro and in vivo. Int J Nanomed;2018. 439–453. 10.2147/IJN.S150977
73. Huo Q, Zhu J, Niu Y, et al. pH-triggered surface charge-switchable polymer micelles for the co-delivery of paclitaxel/disulfiram and overcoming multidrug resistance in cancer. Int J Nanomed. 2017;Volume 12:8631–8647. doi:10.2147/IJN.S144452
74. Kosaka Y, Saeki T, Takano T, et al. Multicenter randomized open-label Phase II clinical study comparing outcomes of NK105 and paclitaxel in advanced or recurrent breast cancer. Int J Nanomed. 2022;17:4567. doi:10.2147/IJN.S372477
75. Nair PR, Karthick SA, Spinler KR, Vakili MR, Lavasanifar A, Discher DE. Filomicelles from aromatic diblock copolymers increase paclitaxel-induced tumor cell death and aneuploidy compared with aliphatic copolymers. Nanomedicine. 2016;11(12):1551–1569. doi:10.2217/nnm-2016-0007
76. Pearce AK, O’Reilly RK. Insights into active targeting of nanoparticles in drug delivery: advances in clinical studies and design considerations for cancer nanomedicine. Bioconjug Chem. 2019;30(9):2300–2311. doi:10.1021/acs.bioconjchem.9b00456
77. Zhao MD, Li JQ, Chen FY, et al. Co-delivery of curcumin and paclitaxel by “core-shell” targeting amphiphilic copolymer to reverse resistance in the treatment of ovarian cancer. Int J Nanomed. 2019;Volume 14:9453–9467. doi:10.2147/IJN.S224579
78. Hussain S. Nanomedicine for treatment of lung cancer. Lung Cancer Pers Med Nov Ther Clin Manag. 2016;2016:137–147.
79. Chu XY, Huang W, Wang YL, et al. Improving antitumor outcomes for palliative intratumoral injection therapy through lecithin–chitosan nanoparticles loading paclitaxel–cholesterol complex. Int J Nanomed. 2019;14:689–705. doi:10.2147/IJN.S188667
80. Sun D, Zhou S, Gao W. What went wrong with anticancer nanomedicine design and how to make it right. ACS Nano. 2020;14(10):12281–12290. doi:10.1021/acsnano.9b09713
81. Tekade RK, Maheshwari R, Soni N, Tekade M, Chougule MB. Nanotechnology for the development of nanomedicine. Nanotechnol Approaches Targeting Delivery Drugs Genes. 2017;2017:3–61.
82. Zhong T, Yao X, Zhang S, et al. A self-assembling nanomedicine of conjugated linoleic acid-paclitaxel conjugate (CLA-PTX) with higher drug loading and carrier-free characteristic. Sci Rep. 2016;6(1):36614. doi:10.1038/srep36614
83. Shuai WX, Zhang L, Li X, et al. Nanoformulated paclitaxel and AZD9291 synergistically eradicate non-small-cell lung cancers in vivo. Nanomedicine. 2018;13(10):1107–1120. doi:10.2217/nnm-2017-0355
84. Raza F, Zafar H, Khan MW, et al. Recent advances in the targeted delivery of paclitaxel nanomedicine for cancer therapy. Mater Adv. 2022;3(5):2268–2290. doi:10.1039/D1MA00961C
85. Campos J, Varas-Godoy M, Haidar ZS. Physicochemical characterization of chitosan-hyaluronan-coated solid lipid nanoparticles for the targeted delivery of paclitaxel: a proof-of-concept study in breast cancer cells. Nanomedicine. 2017;12(5):473–490. doi:10.2217/nnm-2016-0371
86. Han B, Yang Y, Chen J, et al. Preparation, characterization, and pharmacokinetic study of a novel long-acting targeted paclitaxel liposome with antitumor activity. Int J Nanomed. 2020;15:553–571. doi:10.2147/IJN.S228715
87. Azimullah S, Sudhakar CK, Kumar P, et al. Nanosuspensions as a promising approach to enhance bioavailability of poorly soluble drugs: an update. J Drug Delivery Ther. 2019;9(2):574–582. doi:10.22270/jddt.v9i2.2436
88. Cai Z, Zhao X, Duan J, Zhao D, Dang Z, Lin Z. Remediation of soil and groundwater contaminated with organic chemicals using stabilized nanoparticles: lessons from the past two decades. Front Environ Sci Eng. 2020;14:1–20. doi:10.1007/s11783-020-1263-8
89. Dewangan HK. The emerging role of nanosuspensions for drug delivery and stability. Curr Nanomed. 2021;11(4):213–223. doi:10.2174/2468187312666211222123307
90. Shah DP, Patel B, Shah C. Nanosuspension technology: a innovative slant for drug delivery system and permeability enhancer for poorly water soluble drugs. J Drug Delivery Ther. 2015;5(1):10–23.
91. Vedaga SB, Gondkar SB, Saudagar RB. Nanosuspension: an emerging trend to improve solubility of poorly water soluble drugs. J Drug Delivery Ther. 2019;9(3):549–553. doi:10.22270/jddt.v9i3.2635
92. Bodratti AM, Alexandridis P. Formulation of poloxamers for drug delivery. J Funct Biomater. 2018;9(1):11. doi:10.3390/jfb9010011
93. Sun Q, Bai X, Sofias AM, et al. Cancer nanomedicine meets immunotherapy: opportunities and challenges. Acta Pharmacol Sin. 2020;41(7):954–958. doi:10.1038/s41401-020-0448-9
94. Wei Y, Wei Y, Sheng L, et al. Construction of curcumin and paclitaxel co-loaded lipid nano platform and evaluation of its anti-hepatoma activity in vitro and pharmacokinetics in vivo. Int J Nanomed;2023. 2087–2107. 10.2147/IJN.S399289
95. Jin M, Jin G, Kang L, Chen L, Gao Z, Huang W. Smart polymeric nanoparticles with pH-responsive and PEG-detachable properties for co-delivering paclitaxel and survivin siRNA to enhance antitumor outcomes. Int J Nanomed. 2018;13:2405–2426. doi:10.2147/IJN.S161426
96. Cao X, Tan T, Zhu D, et al. Paclitaxel-loaded macrophage membrane camouflaged albumin nanoparticles for targeted cancer therapy. Int J Nanomed. 2020;15:1915–1928. doi:10.2147/IJN.S244849
97. Tran S, DeGiovanni PJ, Piel B, Rai P. Cancer nanomedicine: a review of recent success in drug delivery. Clin Transl Med. 2017;6:1–21. doi:10.1186/s40169-017-0175-0
98. Gibbens-Bandala B, Morales-Avila E, Ferro-Flores G, et al. 177Lu-Bombesin-PLGA (paclitaxel): a targeted controlled-release nanomedicine for bimodal therapy of breast cancer. Mater Sci Eng C. 2019;105:110043. doi:10.1016/j.msec.2019.110043
99. Zhao J, Koay EJ, Li T, Wen X, Li C. A hindsight reflection on the clinical studies of poly (l‐glutamic acid)‐paclitaxel. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2018;10(3):e1497. doi:10.1002/wnan.1497
100. Harshita, Barkat MA, Beg S, Pottoo FH, Ahmad FJ. Nanopaclitaxel therapy: an evidence based review on the battle for next-generation formulation challenges. Nanomedicine. 2019;14(10):1323–1341.
101. Ahmad S, Lambuk L, Ahmed N, et al. Efficacy and safety of nab-paclitaxel in metastatic gastric cancer: a meta-analysis. Nanomedicine. 2023;18(24):1733–1744.
102. Li X, Chen L, Luan S, et al. The development and progress of nanomedicine for esophageal cancer diagnosis and treatment. In: Seminars in Cancer Biology. Elsevier. Vol. 86;2022:873–885
103. Attari Z, Bhandari A, Jagadish PC, Lewis S. Enhanced ex vivo intestinal absorption of olmesartan medoxomil nanosuspension: preparation by combinative technology. Saudi Pharm J. 2016;24(1):57–63. doi:10.1016/j.jsps.2015.03.008
104. Flach F, Breitung-Faes S, Kwade A. Model based process optimization of nanosuspension preparation via wet stirred media milling. Powder Technol. 2018;331:146–154.
105. Zhang T, Li X, Xu J, Shao J, Ding M, Shi S. Preparation, characterization, and evaluation of breviscapine nanosuspension and its freeze-dried powder. Pharmaceutics. 2022;14(5):923.
106. Ma Y, Cong Z, Gao P, Wang Y. Nanosuspensions technology as a master key for nature products drug delivery and in vivo fate. Eur J Pharm Sci. 2023;2023:106425.
107. Sudha T, Bharali DJ, Yalcin M, et al. Targeted delivery of paclitaxel and doxorubicin to cancer xenografts via the nanoparticle of nano-diamino-tetrac. Int J Nanomed. 2017;12:1305–1315. doi:10.2147/IJN.S123742
108. Liu F, Chen Y, Li Y, et al. Folate-receptor-targeted laser-activable poly (lactide-co-glycolic acid) nanoparticles loaded with paclitaxel/indocyanine green for photoacoustic/ultrasound imaging and chemo/photothermal therapy</em>. Int J Nanomed. 2018;13:5139–5158. doi:10.2147/IJN.S167043
109. Kim MS, Haney MJ, Zhao Y, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomed Nanotechnol Biol Med. 2018;14(1):195–204. doi:10.1016/j.nano.2017.09.011
110. Gener P, Gonzalez Callejo P, Seras-Franzoso J, et al. The potential of nanomedicine to alter cancer stem cell dynamics: the impact of extracellular vesicles. Nanomedicine. 2020;15(28):2785–2800. doi:10.2217/nnm-2020-0099
111. Groo AC, Bossiere M, Trichard L, Legras P, Benoît JP, Lagarce F. In vivo evaluation of paclitaxel-loaded lipid nanocapsules after intravenous and oral administration on resistant tumor. Nanomedicine. 2015;10(4):589–601. doi:10.2217/nnm.14.124
112. Xu X, Ho W, Zhang X, Bertrand N, Farokhzad O. Cancer nanomedicine: from targeted delivery to combination therapy. Trends Mol Med. 2015;21(4):223–232. doi:10.1016/j.molmed.2015.01.001
113. Liu J, Meng T, Yuan M, et al. MicroRNA-200c delivered by solid lipid nanoparticles enhances the effect of paclitaxel on breast cancer stem cell. Int J Nanomed;2016. 6713–6725. 10.2147/IJN.S111647
114. Jiang K, Shen M, Xu W. Arginine, glycine, aspartic acid peptide-modified paclitaxel and curcumin co-loaded liposome for the treatment of lung cancer: in vitro/vivo evaluation. Int J Nanomed. 2018;13:2561–2569. doi:10.2147/IJN.S157746
115. Xu W, Bae EJ, Lee MK. Enhanced anticancer activity and intracellular uptake of paclitaxel-containing solid lipid nanoparticles in multidrug-resistant breast cancer cells. Int J Nanomed. 2018;13:7549–7563. doi:10.2147/IJN.S182621
116. Chen Z, Kankala RK, Long L, Xie S, Chen A, Zou L. Current understanding of passive and active targeting nanomedicines to enhance tumor accumulation. Coord Chem Rev. 2023;481:215051. doi:10.1016/j.ccr.2023.215051
117. Hao W, Cui Y, Fan Y, et al. Hybrid membrane-coated nanosuspensions for multi-modal anti-glioma therapy via drug and antigen delivery. J Nanobiotechnology. 2021;19:1–24. doi:10.1186/s12951-021-01110-0
118. Fan Y, Hao W, Cui Y, et al. Cancer cell membrane-coated nanosuspensions for enhanced chemotherapeutic treatment of glioma. Molecules. 2021;26(16):5103. doi:10.3390/molecules26165103
119. Ma Y, Yi J, Ruan J, et al. Engineered cell membrane‐coated nanoparticles: new strategies in glioma targeted therapy and immune modulation. Adv Healthc Mater. 2024;13:2400514. doi:10.1002/adhm.202400514
120. Vikram, Kumar S, Ali J, Baboota S. Potential of nanocarrier-associated approaches for better therapeutic intervention in the management of glioblastoma. Assay Drug Dev Technol. 2024;22(2):73–85. doi:10.1089/adt.2023.073
121. Mendanha D, De Castro JV, Casanova MR, Gimondi S, Ferreira H, Neves NM. Macrophage cell membrane infused biomimetic liposomes for glioblastoma targeted therapy. Nanomed Nanotechnol Biol Med. 2023;49:102663. doi:10.1016/j.nano.2023.102663
122. Li J, Wei Y, Zhang C, et al. Cell-membrane-coated nanoparticles for targeted drug delivery to the brain for the treatment of neurological diseases. Pharmaceutics. 2023;15(2):621. doi:10.3390/pharmaceutics15020621
123. Li W, Cheng J, He F, et al. Cell membrane-based nanomaterials for theranostics of central nervous system diseases. J Nanobiotechnology. 2023;21(1):276. doi:10.1186/s12951-023-02004-z
124. Yang Y, Cheng N, Luo Q, et al. How nanotherapeutic platforms play a key role in glioma? a comprehensive review of literature. Int J Nanomed. 2023;Volume 18:3663–3694. doi:10.2147/IJN.S414736
125. Zhang S, Zhang X, Gao H, et al. Cell membrane-coated biomimetic nanoparticles in cancer treatment. Pharmaceutics. 2024;16(4):531. doi:10.3390/pharmaceutics16040531
126. Wu Y, Qian Y, Peng W, Qi X. Functionalized nanoparticles crossing the brain–blood barrier to target glioma cells. PeerJ. 2023;11:e15571. doi:10.7717/peerj.15571
127. Jiang Q, Xie M, Chen R, et al. Cancer cell membrane-wrapped nanoparticles for cancer immunotherapy: a review of current developments. Front Immunol. 2022;13:973601. doi:10.3389/fimmu.2022.973601
128. Xia Q, Shen J, Ding H, et al. Intravenous nanocrystals: fabrication, solidification, in vivo fate, and applications for cancer therapy. Expert Opin Drug Deliv. 2023;20(11):1467–1488. doi:10.1080/17425247.2023.2268512
129. Xiang H, Xu S, Li J, et al. Functional drug nanocrystals for cancer-target delivery. J Drug Deliv Sci Technol. 2022;76:103807. doi:10.1016/j.jddst.2022.103807
130. Xu Y, Hsu JC, Xu L, Chen W, Cai W, Wang K. Nanomedicine-based adjuvant therapy: a promising solution for lung cancer. J Nanobiotechnology. 2023;21(1):211. doi:10.1186/s12951-023-01958-4
131. Padmakumar S, Amiji MM. Long-acting therapeutic delivery systems for the treatment of gliomas. Adv Drug Deliv Rev. 2023;197:114853. doi:10.1016/j.addr.2023.114853
132. Xia Z, Mu W, Yuan S, Fu S, Liu Y, Zhang N. Cell membrane biomimetic nano-delivery systems for cancer therapy. Pharmaceutics. 2023;15(12):2770. doi:10.3390/pharmaceutics15122770
133. Hasan I, Roy S, Ehexige E, et al. A state-of-the-art liposome technology for glioblastoma treatment. Nanoscale. 2023;15(45):18108–18138. doi:10.1039/D3NR04241C
134. Meng Y, Chen S, Wang C, Ni X. Advances in composite biofilm biomimetic nanodrug delivery systems for cancer treatment. Technol Cancer Res Treat. 2024;23:15330338241250244. doi:10.1177/15330338241250244
135. Li J, Zhao J, Tan T, et al. Nanoparticle drug delivery system for glioma and its efficacy improvement strategies: a comprehensive review. Int J Nanomed. 2020;15:2563–2582. doi:10.2147/IJN.S243223
136. Nan J, Yang W, Xie Y, Yu M, Chen Y, Zhang J. Emerging nano‐immunotherapeutic approaches to glioma. Small Struct. 2023;4(8):2300016. doi:10.1002/sstr.202300016
137. Peng S, Wang Y, Sun Z, et al. Nanoparticles loaded with pharmacologically active plant-derived natural products: biomedical applications and toxicity. Colloids Surf B Biointerfaces. 2023;225:113214. doi:10.1016/j.colsurfb.2023.113214
138. Zhai BT, Sun J, Shi YJ, et al. Review targeted drug delivery systems for norcantharidin in cancer therapy. J Nanobiotechnology. 2022;20(1):509. doi:10.1186/s12951-022-01703-3
139. Li B, Shao H, Gao L, Li H, Sheng H, Zhu L. Nano-drug co-delivery system of natural active ingredients and chemotherapy drugs for cancer treatment: a review. Drug Deliv. 2022;29(1):2130–2161. doi:10.1080/10717544.2022.2094498
140. Melim C, Magalhães M, Santos AC, Campos EJ, Cabral C. Nanoparticles as phytochemical carriers for cancer treatment: news of the last decade. Expert Opin Drug Deliv. 2022;19(2):179–197. doi:10.1080/17425247.2022.2041599
141. Lenders V, Koutsoumpou X, Phan P, et al. Modulation of engineered nanomaterial interactions with organ barriers for enhanced drug transport. Chem Soc Rev. 2023;52(14):4672–4724. doi:10.1039/d1cs00574j
142. Wang Z, You T, Cai C, et al. Biomimetic gold nanostructure with a virus-like topological surface for enhanced antigen cross-presentation and antitumor immune response. ACS Appl Mater Interfaces. 2023;15(11):13929–13940.
143. Wang X, Liu J, Yn LD, et al. Recent updates on the efficacy of mitocans in photo/radio-therapy for targeting metabolism in chemo/radio-resistant cancers: nanotherapeutics. Curr Med Chem. 2024;2024:1.
144. Lou S, Zhao Z, Dezort M, Lohneis T, Zhang C. Multifunctional nanosystem for targeted and controlled delivery of multiple chemotherapeutic agents for the treatment of drug-resistant breast cancer. ACS omega. 2018;3(8):9210–9219. doi:10.1021/acsomega.8b00949
145. Rabinow BE. Nanosuspensions in drug delivery. Nat Rev Drug Discov. 2004;3(9):785–796. doi:10.1038/nrd1494
146. Patel VR, Agrawal YK. Nanosuspension: an approach to enhance solubility of drugs. J Adv Pharm Technol Res. 2011;2(2):81–87. doi:10.4103/2231-4040.82950
147. Kim M, Shin M, Zhao Y, Ghosh M, Son Y. Transformative impact of nanocarrier‐mediated drug delivery: overcoming biological barriers and expanding therapeutic horizons. Small Sci. 2024;4(11):2400280. doi:10.1002/smsc.202400280
148. Fan Y, Cui Y, Hao W, et al. Carrier-free highly drug-loaded biomimetic nanosuspensions encapsulated by cancer cell membrane based on homology and active targeting for the treatment of glioma. Bioact Mater. 2021;6(12):4402–4414. doi:10.1016/j.bioactmat.2021.04.027
149. Liao J, Fan L, Li Y, et al. Recent advances in biomimetic nanodelivery systems: new brain-targeting strategies. J Control Release. 2023;358:439–464. doi:10.1016/j.jconrel.2023.05.009
150. Satchanska G, Davidova S, Petrov PD. Natural and synthetic polymers for biomedical and environmental applications. Polymers. 2024;16(8):1159. doi:10.3390/polym16081159
151. Drabczyk A, Kudłacik-Kramarczyk S, Jamroży M, Krzan M. Biomaterials in drug delivery: advancements in cancer and diverse therapies. Int J Mol Sci. 2024;25(6):3126. doi:10.3390/ijms25063126
152. Pokrajac L, Abbas A, Chrzanowski W, et al. Nanotechnology for a sustainable future: addressing global challenges with the international network4sustainable nanotechnology. ACS Nano. 2021;15(12):18608–18623. doi:10.1021/acsnano.1c10919
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.