Next Article in Journal
Development of an LC-MS Method for the Analysis of Birch (Betula sp.) Bark Bioactives Extracted with Biosolvents
Previous Article in Journal
Docosahexaenoic Acid Inhibits Osteoclastogenesis via FFAR4-Mediated Regulation of Inflammatory Cytokines
Previous Article in Special Issue
Effects of Polysaccharides Extracted from Stem Barks on the Spontaneous Contractile Activity of the Ileal Smooth Muscle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nano-Phytomedicine: Harnessing Plant-Derived Phytochemicals in Nanocarriers for Targeted Human Health Applications

1
School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
2
School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
3
Directorate of Research, Nehru Kendra, Mahatama Jyotiba Phule Rohilkhand University, Bareilly 243006, India
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(15), 3177; https://doi.org/10.3390/molecules30153177
Submission received: 30 June 2025 / Revised: 25 July 2025 / Accepted: 27 July 2025 / Published: 29 July 2025
(This article belongs to the Special Issue Phytochemistry, Human Health and Molecular Mechanisms)

Abstract

Phytochemicals from medicinal plants offer significant therapeutic benefits, yet their clinical utility is often limited by poor solubility, instability, and low bioavailability. Nanotechnology presents a transformative approach to overcome these challenges by encapsulating phytochemicals in nanocarriers that enhance stability, targeted delivery, and controlled release. This review highlights major classes of phytochemicals such as polyphenols, flavonoids, and alkaloids and explores various nanocarrier systems including liposomes, polymeric nanoparticles, and hybrid platforms. It also discusses their mechanisms of action, improved pharmacokinetics, and disease-specific targeting. Further, the review examines clinical advancements, regulatory considerations, and emerging innovations such as smart nanocarriers, AI-driven formulation, and sustainable manufacturing. Nano-phytomedicine offers a promising path toward safer, more effective, and personalized therapies, bridging traditional herbal knowledge with modern biomedical technology.

1. Introduction

1.1. Background of Phytochemicals in Traditional and Modern Medicine

Phytochemicals are biologically active secondary metabolites derived from plants, including polyphenols, alkaloids, terpenoids, and flavonoids. These compounds serve crucial roles in plant defense and have long been recognized for their therapeutic properties in traditional medicine systems such as Ayurveda, Traditional Chinese Medicine (TCM), and Indigenous medicinal practices worldwide [1]. Historically, plant-derived products like willow bark (source of salicin) and opium poppy (source of morphine) formed the basis for modern drug development [2]. The transition from ethnobotanical usage to evidence-based applications marked the beginning of phytochemistry as a discipline. In modern medicine, several plant-derived drugs have achieved clinical success, such as paclitaxel (from Taxus brevifolia) for cancer, artemisinin (from Artemisia annua) for malaria, and berberine (from Berberis vulgaris) for infections and metabolic syndromes [3]. With technological advancement in analytical chemistry and pharmacology, the molecular targets and mechanisms of these compounds have become more clearly defined. However, despite significant bioactivity, their clinical use remains restricted due to poor pharmacokinetics and variable efficacy in vivo. Nonetheless, rising interest in “natural” therapies and increased awareness of chronic disease management have renewed focus on plant-based compounds for their multi-target actions, low toxicity profiles, and immunomodulatory effects [4]. This renewed scientific interest sets the stage for improving delivery systems that can address the inherent limitations of phytochemicals and maximize their therapeutic potential in modern healthcare.

1.2. Limitations of Free Phytochemicals

Despite their promising biological activity, most free phytochemicals exhibit poor drug-like characteristics that hinder their clinical application. One of the most critical limitations is low aqueous solubility. Over 40% of plant-derived compounds have poor water solubility, which restricts their absorption in the gastrointestinal tract and reduces systemic circulation levels [5]. Additionally, once absorbed, many phytochemicals undergo rapid first-pass metabolism in the liver and intestines, leading to fast degradation and clearance from the body [6]. Another key issue is chemical instability. Many phytochemicals, such as curcumin and quercetin, are sensitive to light, pH, and enzymatic oxidation, resulting in the loss of bioactivity before reaching their target tissues [7]. Furthermore, poor membrane permeability and non-specific distribution limit their accumulation at disease sites while increasing the risk of side effects elsewhere. For instance, flavonoids with potent in vitro anticancer activity often fail in clinical trials due to these delivery challenges [8]. In essence, the therapeutic potential of phytochemicals is constrained by a triad of solubility, stability, and bioavailability issues. These challenges necessitate the development of advanced delivery systems such as nanocarriers that can protect the compounds, enhance absorption, and facilitate site-specific action to realize their full clinical value [9].

1.3. Emergence of Nanotechnology in Phytomedicine

Nanotechnology offers a transformative approach to improving the pharmacological performance of phytochemicals. By encapsulating these compounds in nanoscale carriers (typically 1–100 nm), researchers can significantly enhance their solubility, protect them from degradation, and extend their circulation time [10,11,12,13,14,15,16]. Nanocarriers like liposomes, solid lipid nanoparticles (SLNs), polymeric nanoparticles, dendrimers, and metallic nanostructures have shown promising results in stabilizing phytochemicals and enabling targeted delivery [17,18,19]. For example, encapsulating curcumin in polymeric nanoparticles has been shown to improve its bioavailability by over 2000%, while quercetin-loaded liposomes can deliver controlled release and site-specific targeting [20]. Furthermore, the surface of nanocarriers can be functionalized with ligands such as antibodies, peptides, or folic acid to ensure specific binding to target cells, improving therapeutic efficacy and reducing systemic toxicity [21]. Stimuli-responsive carriers can also trigger drug release in response to pH, temperature, or enzymatic activity at the disease site, enhancing precision medicine strategies. Additionally, green synthesis approaches using plant extracts in nanoparticle preparation provide eco-friendly and cost-effective routes to phytochemical stabilization [22,23]. As such, nanotechnology not only enhances the pharmacokinetics of plant-derived therapeutics but also opens the door to multifunctional treatment platforms capable of combining therapy with diagnostics (theranostics), paving the way for next-generation phytomedicines [24]. Figure 1 illustrates the transformative integration of nanotechnology into phytomedicine, emphasizing how plant-derived bioactive compounds are engineered into multifunctional nanoformulations for advanced therapeutic applications. Phytochemicals—such as polyphenols, alkaloids, and terpenoids—exhibit potent biological activities, but their clinical translation is often hindered by low solubility, instability, and poor bioavailability. Nanotechnology addresses these limitations through encapsulation strategies using various nanocarriers (e.g., liposomes, polymeric nanoparticles, nanoemulsions), which protect the phytochemicals, enhance cellular uptake, and provide controlled release. The schematic shows the central role of these carriers in enabling targeted delivery, reducing systemic toxicity, and facilitating precision medicine approaches. Furthermore, nanocarrier design can be tuned to respond to specific stimuli (pH, enzymes, redox conditions), offering spatiotemporal control over drug release. This convergence of botanical pharmacology and nanotechnology not only improves therapeutic efficacy but also supports the development of eco-friendly and biocompatible treatments with potential applications in oncology, neurodegeneration, and infectious diseases. The figure encapsulates this paradigm shift in drug delivery science, highlighting a new era of sustainable and personalized phytomedicine.

1.4. Objective and Scope of the Review

This review aims to provide a comprehensive overview of nano-phytomedicine, focusing on how nanotechnology can address the delivery challenges of phytochemicals and enable targeted therapy for various health conditions. The objectives are:
  • To categorize major nanocarrier systems including lipid-based, polymeric, and inorganic nanomaterials used in phytochemical encapsulation and delivery, with an emphasis on their physicochemical properties and compatibility with bioactives.
  • To elucidate molecular mechanisms of nano-phytochemical interactions with human cells, including uptake, intracellular trafficking, release kinetics, and downstream signaling modulation.
  • To discuss therapeutic applications across key areas such as oncology, neurodegeneration, cardiovascular disease, metabolic disorders, and infectious diseases, highlighting both preclinical and clinical outcomes.
  • To evaluate the pharmacokinetic and toxicological profiles of nanoformulated phytochemicals, considering parameters such as bioavailability, biodistribution, toxicity, and immunogenicity.
  • To assess the current translational landscape, including clinical trials, regulatory barriers, commercialized products, and future scalability for global healthcare implementation.
  • To explore future directions such as AI-guided formulation design, personalized nano-phytotherapy, smart delivery systems, and sustainable manufacturing.
By synthesizing recent advances and challenges, this review aims to offer insight into the integration of phytochemistry with nanotechnology, and how this convergence can revolutionize drug delivery and human health outcomes [25].

2. Overview of Plant-Derived Phytochemicals Relevant to Human Health

2.1. Major Classes: Polyphenols, Alkaloids, Terpenoids, Flavonoids, and Glycosides

Figure 2a provides an integrated schematic overview of the key classes of phytochemicals relevant to human health, including polyphenols, alkaloids, terpenoids, flavonoids, and glycosides, along with representative chemical structures and therapeutic roles. Figure 2b, representative chemical structures of major phytochemical classes including polyphenols (e.g., quercetin), alkaloids (e.g., berberine), terpenoids (e.g., paclitaxel), flavonoids (e.g., catechin), and glycosides (e.g., digoxin). These structural frameworks underpin diverse therapeutic activities, supporting their integration into nanoformulated delivery platforms for enhanced bioactivity and targeted treatment. These scaffolds form the basis for nanoformulation strategies in modern phytomedicine. Polyphenols are the largest family of plant secondary metabolites, defined by multiple phenolic rings and subdivided into phenolic acids, flavonoids, stilbenes, and lignans [26]. These compounds are abundant in dietary sources such as fruits, vegetables, tea, and wine. Flavonoids such as quercetin, catechins, and anthocyanins are especially prominent and contribute to cardiovascular protection, blood sugar regulation, and neuroprotection, as evidenced in numerous epidemiological and intervention studies [27]. Alkaloids, characterized by nitrogen-containing heterocycles, exhibit diverse pharmacological effects. Classic examples include morphine and codeine (opioid analgesics), quinine (antimalarial), and berberine (antimicrobial and metabolic regulator) [28]. Their mechanisms of action include receptor binding, enzyme modulation, and ion channel regulation at the molecular level. Terpenoids (isoprenoids), derived from five-carbon isoprene units, encompass subclasses such as monoterpenes (e.g., limonene), diterpenes (e.g., paclitaxel), and tetraterpenes (e.g., carotenoids). These exhibit potent anti-inflammatory, antioxidant, and anticancer properties with paclitaxel serving as a well-known chemotherapeutic agent [29]. Flavonoids, a major subclass of polyphenols, are multifunctional molecules capable of modulating oxidative stress, inflammation, mutagenesis, and carcinogenesis. They interact with key cellular pathways including NF-κB, MAPK, and PI3K/Akt, contributing to their observed protective effects in cardiovascular and cognitive health [30]. Glycosides, such as cardiac glycosides (e.g., digoxin) and anthraquinone glycosides (e.g., senna), play critical roles in modern therapeutics. Digoxin, isolated from Digitalis, enhances cardiac contractility via Na+/K+-ATPase inhibition, whereas anthraquinone glycosides stimulate intestinal motility, providing laxative benefits [31]. By depicting their representative molecular frameworks and associated health applications, Figure 2 contextualizes these chemical classes within modern phytomedicine. This visual integration renders a separate general subchapter unnecessary while enhancing reader comprehension.

2.2. Therapeutic Properties: Antioxidant, Anticancer, Anti-Inflammatory, Antimicrobial

Phytochemicals exhibit multiple overlapping therapeutic effects:
  • Antioxidants: Polyphenols neutralize free radicals and reactive oxygen species (ROS) in vitro, though their direct antioxidant activity in vivo is less certain due to poor bioavailability [32]. Still, their metabolites may indirectly upregulate endogenous antioxidant pathways via Nrf2 signaling.
  • Anticancer activity: Numerous phytochemicals modulate apoptosis, cell proliferation, angiogenesis, and metastasis. For example, catechin-3-gallate has been shown to enhance therapeutic efficacy through ROS induction and mitochondrial pathway activation in tumor cells [33]. Additionally, carrier-free self-assembled phytochemical nanoparticles (e.g., curcumin conjugates) demonstrate synergistic targeting and cytotoxicity in cancer models [34].
  • Anti-inflammatory effects: Myricetin, quercetin, and resveratrol inhibit pro-inflammatory cytokines such as IL-1β and TNF-α through downregulating signaling kinases and transcription factors (NF-κB, MAPK), showing promise in inflammatory and autoimmune disorders [35,36].
  • Antimicrobial activity: Polyphenol-rich plant extracts and alkaloids (like berberine) show broad-spectrum antimicrobial effects. For instance, berberine disrupts bacterial cell membrane integrity, while flavonoids inhibit viral enzymes and microbial adhesion [37].
These bioactivities highlight the multifaceted therapeutic potential of these phytochemicals—when adequately delivered and bioavailable.

2.3. Bioavailability Challenges and Need for Delivery Enhancement

Phytochemicals face significant barriers in clinical translation due to poor pharmacokinetic properties:
  • Poor solubility and stability: Many are hydrophobic and insoluble in aqueous media, leading to limited absorption across the gastrointestinal tract [38]. Moreover, they are prone to degradation through oxidation, heat, light, or pH changes [39].
  • Rapid metabolism and clearance: After absorption, phytochemicals are quickly subjected to phase I/II metabolism (e.g., glucuronidation, sulfation), yielding low plasma concentrations and short half-lives that necessitate frequent dosing [40].
  • Low membrane permeability: Their molecular size, polarity, or lipophilicity often limits cellular uptake and penetration through biological barriers like the blood–brain barrier [41].
  • Variability in bioavailability: Individual differences in gut microbiota, genetic polymorphisms in metabolic enzymes, and interactions with food can result in erratic absorption and therapeutic response [42].
As conventional dosage forms fail to resolve these issues, nanotechnology has emerged as a leading strategy. Nanocarriers including liposomes, polymeric nanoparticles, solid-lipid structures, chitosan-based carriers, nanoemulsions, and mesoporous silica provide tailored solutions: they enhance solubility, protect against degradation, facilitate controlled release, improve permeability, and enable target-specific delivery [43,44,45]. For example, chitosan nanoparticles administered orally increase mucoadhesion, protect from gastric enzymes, and facilitate transport across the intestinal epithelium [46,47]. In a comparative study, curcumin-loaded chitosan nanoparticles demonstrated a 7.4-fold increase in oral bioavailability compared to free curcumin. Mesoporous silica supports the stabilization of poorly soluble phytochemicals in amorphous form, boosting dissolution rates [48]. A study showed that mesoporous silica nanocarriers encapsulating resveratrol increased its aqueous solubility by over 10-fold and enhanced its antioxidant activity in vitro. Hybrid delivery systems combining multiple materials or nanocarrier types offer synergistic advantages. For instance, lipid-polymer hybrid nanoparticles encapsulating quercetin achieved enhanced cellular uptake and prolonged systemic circulation compared to either liposomes or polymeric carriers alone. Stimuli-responsive nanocarriers that release phytochemicals in response to environmental triggers (pH, temperature, redox state, or enzymes) are particularly promising. For example, pH-responsive nanoparticles carrying berberine showed selective release in acidic tumor microenvironments, improving cytotoxicity against cancer cells while sparing healthy tissue. In comparing delivery systems, studies have shown that nanoemulsions, while improving solubility, are less stable than solid lipid nanoparticles under physiological conditions. Conversely, polymeric nanoparticles provide controlled release but may show slower release kinetics compared to liposomal systems. These findings underscore the importance of tailoring nanocarriers to the specific physicochemical and therapeutic profile of each phytochemical. So the integrating nanocarrier technologies with in vitro and in vivo pharmacokinetic evaluations enables rational design for optimized therapeutic performance. Next Figure 3 provides a visual summary of these nanocarriers systems and their comparative performance metrics in enhancing phytochemical bioavailability. Table 1 presents a comparative overview of the major classes of plant-derived phytochemicals, highlighting representative compounds, their primary therapeutic effects, and key delivery challenges. Polyphenols such as curcumin and resveratrol are widely studied for their potent antioxidant and anticancer properties, yet their clinical efficacy is often limited by poor water solubility and rapid metabolism. Alkaloids like berberine and vincristine exhibit strong pharmacological activity, but their therapeutic use can be constrained by toxicity concerns at high doses. Terpenoids, including paclitaxel and carotenoids, offer antimicrobial and anticancer benefits but suffer from oxidative instability. Flavonoids, commonly found in fruits and vegetables, demonstrate neuroprotective and anti-inflammatory effects, though their bioavailability is hindered by rapid metabolism and poor membrane permeability. Lastly, glycosides such as digoxin are known for their cardiotonic effects but face challenges related to gastrointestinal degradation. Overall, the table underscores the need for innovative delivery strategies such as nanocarriers to overcome these limitations and harness the full therapeutic potential of phytochemicals.

3. Nanocarrier Systems for Phytochemical Delivery

3.1. Lipid-Based Carriers: Liposomes, Solid Lipid Nanoparticles (SLNs), and Nanostructured Lipid Carriers (NLCs)

Liposomes are spherical vesicles composed of phospholipid bilayers encapsulating an aqueous core. Typically ranging from 50 to several hundred nanometers, these biocompatible carriers are ideal for delivering both hydrophilic and hydrophobic phytochemicals [49]. Their structural flexibility allows oral, topical, and parenteral delivery, with the lipid bilayer protecting compounds like polyphenols and carotenoids from degradation. Liposomal encapsulation can substantially enhance bioavailability: for instance, liposomal curcumin formulations have shown significantly better plasma levels than free curcumin [50]. Active targeting is achievable through surface modification with ligands such as polyethylene glycol (PEG), antibodies, or specific peptides, enhancing selective uptake in target tissues [51].
Solid Lipid Nanoparticles (SLNs) consist of solid lipids stabilized by surfactants, offering stability at room and body temperatures. SLNs combine the benefits of lipid emulsions and polymeric carriers, such as controlled release, protection against chemical degradation, and scalable manufacturing, while avoiding organic solvents [52]. They can incorporate both lipophilic and hydrophilic phytochemicals, resisting filtration by the mononuclear phagocyte system when optimized at 120–200 nm [53]. SLNs have been shown to improve oral bioavailability of compounds like ferrous sulfate and lipophilic phytochemicals via enhanced gastrointestinal stability and absorption [54].
Figure 3 provides a comprehensive illustration of the diverse nanocarrier systems employed for phytochemical delivery, categorized into lipid-based, polymeric, inorganic, hybrid, and stimuli-responsive platforms. Lipid-based systems such as liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs) are shown to enhance solubility, protect against degradation, and facilitate passive targeting via the enhanced permeability and retention (EPR) effect. Polymeric nanoparticles, crafted from both natural (e.g., chitosan, alginate) and synthetic (e.g., PLGA, PEG) polymers, offer tunable release kinetics and surface modifications for site-specific delivery. Inorganic carriers including metal and metal oxide nanoparticles (like gold, silver, and ZnO) and mesoporous silica are highlighted for their intrinsic bioactivity, stability, and potential for imaging-guided therapy. Hybrid systems integrate lipid, polymer, or inorganic components to optimize payload encapsulation and dual functionality. Stimuli-responsive nanocarriers, which release their phytochemical load upon exposure to triggers such as pH, redox potential, or enzymes, provide controlled and precision-targeted therapeutic action, marking a pivotal innovation in nano-phytomedicine.
Nanostructured Lipid Carriers (NLCs) are advanced lipid-based systems containing mixed solid and liquid lipids to increase payload capacity and reduce crystallinity. This design enhances drug loading flexibility and release control [55]. NLCs demonstrate improved encapsulation efficiency and sustained release profiles over SLNs, making them well-suited for phytochemicals prone to rapid degradation. Preclinical studies have shown that NLCs loaded with botanical extracts achieve higher stability and biodistribution compared to free compounds, underscoring their potential for targeted delivery [56]. Lipid-based carriers thus offer protective encapsulation, improved dissolution, controlled release, and opportunities for functionalization. These capabilities position them as leading platforms to address solubility, stability, and targeted delivery challenges intrinsic to phytochemical therapies [57,58,59].

3.2. Polymeric Nanoparticles: Natural and Synthetic Polymers

Polymeric Nanoparticles (PNPs) are colloidal systems prepared from biodegradable polymers, offering robust strategies for encapsulating, protecting, and delivering phytochemicals [60]. These systems can be engineered as nanospheres, with bioactives dispersed within the polymer matrix, or nanocapsules, with a distinct core–shell architecture encapsulating the compound [61]. Additional morphological variants include micelles, dendrimers, and polyplexes, each customizable for specific payloads and release mechanisms [62]. Natural polymers such as polysaccharides (e.g., chitosan, alginate) and proteins (e.g., albumin, gelatin) offer high biocompatibility, biodegradability, and lower immune reactivity. Chitosan, in particular, enhances mucoadhesion and opens tight junctions in epithelial tissues, improving oral delivery of nutrients and phytochemicals [63,64]. Albumin and gelatin are widely used for their high binding capacity and ease of functionalization. For example, albumin nanoparticles loaded with berberine or ginsenosides have demonstrated increased circulation time, tumor targeting, and anticancer activity compared to free compounds [65,66]. Synthetic polymers like PLGA, PLA, PCL, and PEGylated derivatives are prized for their tunable degradation rates, mechanical properties, and reproducibility [67,68]. PLGA nanoparticles have been employed for curcumin, EGCG, and lycopene delivery, employing techniques such as emulsion diffusion and nanoprecipitation. In a recent study, lycopene-loaded PLGA NPs achieved encapsulation efficiencies of about 97%, with controlled release over 12 days [69]. PLGA nanoparticles also enable spray-dry formulations and scale-up processes essential for industrial application [70,71]. Hybrid systems, such as polymer–lipid hybrids (PLHNPs), combine the stability and payload capacity of polymeric cores with the encapsulation efficiency and biocompatibility of lipid shells [72]. PLHNPs have shown improved encapsulation rates, protection against enzymatic breakdown, and enhanced in vivo stability for phytochemicals [73]. Overall, polymeric nanocarriers provide highly tunable platforms capable of high drug loading, controlled and stimuli-responsive release, surface functionality for targeting, and favorable pharmacokinetic profiles. Such versatility makes them core components in the toolbox of nano-phytomedicine [74,75].

3.3. Inorganic Nanoparticles: Metal, Metal Oxide, and Silica Nanoparticles

Inorganic nanoparticles offer robust platforms for phytochemical delivery, characterized by unique physicochemical properties such as controlled size, surface charge, and imaging compatibility. Metal nanoparticles like gold (AuNPs) and silver (AgNPs) can be synthesized via green methods using phytochemicals both as reducing agents and capping ligands (e.g., flavonoids or polyphenols), allowing dual functionality: structural stabilization and therapeutic activity [76,77]. AuNPs functionalized with catechin or quercetin enhance targeted delivery and provide photothermal capabilities for cancer therapy [78]. AgNPs exhibit strong antimicrobial activity due to reactive oxygen species (ROS) generation, and when combined with phytoconstituents, they achieve a synergistic effect against resistant pathogens [79,80]. Metal oxide nanoparticles such as zinc oxide (ZnO) and iron oxide (Fe3O4) offer specialized functionalities. ZnO-NPs have intrinsic antimicrobial properties and can be loaded with curcumin or other bioactives to enhance wound healing, leveraging the combined effect of controlled drug release and inherent surface reactivity [81]. Fe3O4-based particles serve as drug carriers with magnetic targeting abilities, enabling external guidance and enhanced delivery efficiency for phytochemicals like resveratrol in tumor tissues [82]. Silica nanoparticles, particularly mesoporous silica nanoparticles (MSNs), are favored for their high surface area, tunable pore sizes, and ease of functionalization. MSNs can protect encapsulated phytochemicals from harsh gastrointestinal conditions, promoting controlled release profiles for hydrophobic compounds such as green tea catechins [83]. Functional moieties like pH-sensitive groups can be grafted onto the surface to achieve stimuli-responsive delivery ideal for tumor microenvironments or inflamed tissues [84]. Inorganic carriers thus enable multifunctional delivery and theranostic capabilities when combined with imaging agents or responsive triggers [85,86].
Table 2 illustrates the expanding versatility of nanocarrier platforms in phytochemical delivery, highlighting a strategic shift in formulation science from passive encapsulation to functionally adaptive systems. Beyond their individual characteristics, what emerges is a trend toward modularity and hybridization designing systems that merge structural integrity with responsiveness to biological cues. This adaptability allows for greater control over release kinetics and biodistribution, critical for maximizing therapeutic efficacy while minimizing systemic exposure. Notably, the incorporation of targeting ligands or stimuli-responsive elements signifies an evolution from conventional carriers to “smart” delivery vehicles. These advancements align with personalized medicine objectives, enabling site-specific delivery in heterogenous pathological conditions like tumors or inflamed tissues. Moreover, as nanocarrier designs increasingly integrate diagnostic and therapeutic functions, a new class of theranostic phytomedicines is emerging, capable of simultaneous treatment and monitoring. These trends underscore the importance of interdisciplinary innovation in driving the next generation of natural product-based therapeutics.

3.4. Hybrid and Stimuli-Responsive Nanocarriers

Hybrid nanocarriers combine organic and inorganic materials to achieve synergistic effects: improved payload stability, controlled release, and multifunctionality. One outstanding example is lipid–polymer hybrids, featuring a polymeric core for structural integrity and a lipid shell to improve biocompatibility and cellular uptake [87]. For instance, curcumin–PLGA core with a lecithin–PEG shell exhibits greater circulation time, tumor selectivity, and controlled release compared to single-material systems [92]. Gold-coated liposomes or polymeric nanocapsules can integrate photothermal functionality with drug delivery: upon laser irradiation at specific wavelengths, these constructs trigger localized heating that releases phytochemicals in target tissues a strategy beneficial in combined cancer therapy [93,94]. Similarly, magnetic-lipid hybrids incorporate iron oxide cores with lipidic envelopes to allow magnetic guidance and enhanced permeability in tumor regions [95]. Stimuli-responsive nanocarriers release their payload in response to internal (pH, enzymes, redox state) or external triggers (light, magnetic fields, ultrasound), thereby improving precision and therapeutic index. For example, pH-sensitive polymeric micelles release curcumin preferentially within the acidic tumor microenvironment, and enzyme-responsive systems such as matrix metalloproteinase (MMP)-cleavable linkers enable drug liberation only in inflamed tissues [88,89]. Redox-responsive carriers exploit high intracellular glutathione levels to selectively release encapsulated alkaloids like berberine within cancer cells [96]. These hybrid and stimuli-responsive systems are at the forefront of targeted nano-phytomedicine, with the potential to significantly elevate therapeutic outcomes.

3.5. Surface Functionalization and Targeting Strategies

Targeted nanocarrier delivery hinges on surface engineering. Passive targeting, driven by the enhanced permeability and retention (EPR) effect in tumor vasculature, is enhanced by PEGylation, which prolongs systemic circulation and reduces opsonization [90]. Active targeting uses ligands like folic acid, peptides (e.g., RGD), antibodies, or aptamers to direct particles towards cells expressing specific receptors. Folic acid-conjugated nano-structured lipid carriers loaded with resveratrol preferentially accumulate in folate receptor-overexpressing ovarian cancer cells, increasing uptake by over twofold and enhancing cytotoxicity [91]. Mannose or lectin surface decoration targets macrophages for anti-inflammatory phytochemicals like curcumin or quercetin in diseases such as arthritis and inflammatory bowel disease [97]. Antibody-conjugated carriers such as HER2-targeted polymeric nanoparticles loaded with paclitaxel precursor phytochemicals have shown improved selectivity and cytotoxicity in breast cancer models [98]. Stealth coatings using zwitterionic polymers or cell-membrane camouflaging (e.g., leukocyte or platelet membranes) further reduce immune recognition and improve circulation persistence [99]. Multifunctional or dual-targeting constructs allow for sequential targeting of vasculature and cancer cells by co-presenting antibodies and peptides, thereby enhancing tumor penetration [100]. Through judicious surface functionalization, nano-phytomedicines achieve a high degree of specificity and cellular control compared to conventional phytochemical administration methods.

4. Molecular Mechanisms of Action of Nano-Phytomedicine

4.1. Cellular Uptake and Intracellular Trafficking of Nanoformulations

Cellular uptake of nano-phytomedicines is governed by both the intrinsic properties of the nanocarriers such as size, surface charge, and ligand decoration and the specific endocytic pathways prevalent in target cells. Nanoparticles sized between ~50–200 nm are preferentially internalized via clathrin- and caveolin-mediated endocytosis, as well as macropinocytosis, allowing efficient intracellular delivery of phytochemicals [101]. Charge also plays a key role: positively charged nanocarriers interact more readily with negatively charged phospholipids, facilitating adsorption-mediated endocytosis. However, excessive surface cationicity must be balanced to avoid cytotoxicity and rapid clearance [102]. Surface functionalization with targeting moieties such as antibodies, peptides, aptamers, or small molecules (e.g., folic acid) directs nanoparticles to specific cell types. Ligand-receptor binding triggers receptor-mediated endocytosis, conferring selectivity and minimizing off-target distribution. Once internalized, trafficking routes diverge depending on nanosystem design. Lipid nanocarriers and polymeric vesicles may fuse with early endosomes, traverse the endosomal–lysosomal pathway, and eventually release their cargo via pH-responsive mechanisms. In contrast, certain inorganic or polymeric systems incorporate endosomal escape groups—such as proton-sponge domains or fusogenic peptides to release phytochemicals directly into the cytosol, avoiding degradation [103].
Intracellular trafficking also intersects with trafficking to subcellular organelles. Targeting mitochondria, endoplasmic reticulum, or even the nucleus often requires nanosystems equipped with organelle-specific peptides (e.g., TAT peptide, mitochondrial signal sequences). Mitochondria-targeted nano-phytomedicines can amplify ROS-mediated apoptosis specifically in cancer cells, while ER-targeted delivery can disrupt calcium homeostasis and protein synthesis. Moreover, nanoparticle core degradation kinetics determine whether phytochemicals are released early within lysosomes or later during cytosolic diffusion. Nanocarrier design also accounts for efflux mechanisms particularly P-glycoprotein pumps that expel small-molecule phytochemicals. Encapsulation protects the payload during uptake and retention, inhibiting efflux and prolonging intracellular exposure. For example, curcumin-loaded polymeric micelles show sustained cytosolic presence despite P-gp expression [104]. Furthermore, nanoparticle-mediated phytochemical delivery often modulates nutrient-sensing pathways (like mTOR and AMPK) post-uptake, enhancing uptake rates in proliferative tissues such as tumors. Overall, successful nano-phytomedicine design hinges on understanding and controlling cellular uptake and trafficking dynamics. Pairing size and surface chemistry with targeting ligands and endosomal escape strategies optimizes intracellular delivery, subcellular targeting, and payload release—foundations critical to maximizing therapeutic potential across diverse phytochemicals [105].

4.2. Modulation of Redox Balance and Antioxidant Pathways

Redox homeostasis plays a central role in disease progression, with an imbalance between reactive oxygen species (ROS) and cellular antioxidant defenses contributing to conditions ranging from cancer and neurodegeneration to chronic inflammation. Nano-phytomedicines leverage the inherent antioxidant properties of phytochemicals such as polyphenols and flavonoids while overcoming limitations in solubility and stability [106]. Encapsulation enhances intracellular accumulation of antioxidants, enabling robust modulation of redox-sensitive signaling pathways. A canonical pathway modulated by antioxidant phytochemicals is the Nrf2-Keap1 axis. Under oxidative stress, stabilized Nrf2 translocates to the nucleus and induces expression of phase II enzymes like glutathione-S-transferases and NAD(P)H:quinone oxidoreductase 1. Nanoformulations of resveratrol, quercetin, and sulforaphane have demonstrated enhanced Nrf2 activation compared to free forms, leading to improved cellular resilience and reduced oxidative damage in vitro and in vivo [107]. Mechanistically, sustained intracellular release from nanocarriers ensures prolonged antioxidant stimulation. In addition, nanocarrier encapsulation enables phytochemicals to neutralize ROS directly within subcellular compartments. For example, mitochondria-targeted nanocurcumin interrupts ROS generation in situ, preserving membrane integrity and preventing cytochrome-c release. Similarly, cerium oxide or iron oxide-based hybrid carriers can be functionalized or conjugated with phytochemicals to create synergistic antioxidant platforms. In inflammatory pathologies, redox modulation by these nanoconjugates intersects with pro-inflammatory signaling. Nano-phytomedicines that scavenge ROS also suppress NF-κB and MAPK pathways central regulators of cytokine production. For instance, epigallocatechin gallate (EGCG)-loaded polymeric nanoparticles reduce TNF-α and IL-6 release under oxidative stress by both direct radical scavenging and indirect gene modulation [108]. Despite the strength of these effects, accurate modulation demands balance. Over-suppression of ROS can impair immune functions, as controlled ROS levels are required for phagocytosis and pathogen defense. Advanced nanocarriers with redox-feedback sensitivity—releasing payloads only under high ROS—help mitigate this risk. For example, disulfide-containing polymeric carriers release phytochemicals specifically in oxidized environments, sparing normal cells from unnecessary antioxidant exposure [109]. So the nano-phytomedicine systems offer enhanced redox control by delivering antioxidants in targeted and controlled fashions. By focusing antioxidant activity where and when needed guided by biological cues these systems promise therapeutic benefits with reduced side effects, making redox modulation a cornerstone of phytochemical nanotherapy [110].

4.3. Regulation of Apoptosis, Cell Proliferation, and Inflammation

Nano-phytomedicines exert their therapeutic influence by orchestrating key cellular signaling pathways involved in apoptosis, proliferation, and inflammation. Unlike monotherapeutic small molecules, phytochemicals often achieve multitarget modulation, and nanoformulations enhance these effects by improving concentration and site-specificity at cellular targets [111]. A hallmark target in oncology is the intrinsic mitochondrial apoptosis pathway. Encapsulated curcumin, resveratrol, and berberine delivered via nanocarriers can accumulate in mitochondria, where they trigger cytochrome c release, upregulate pro-apoptotic proteins (Bax, Bak), and inhibit anti-apoptotic Bcl-2 family proteins. Nanodelivery increases mitochondrial targeting, leading to robust caspase-9 and -3 activation and apoptosis in cancer cells [112]. This selective activation in tumor cells helps reduce systemic toxicity. In parallel, cell proliferation pathways—particularly PI3K/Akt and MAPK/ERK are frequently dysregulated in disease. Nano-phytomedicines loaded with quercetin or apigenin have demonstrated potent inhibition of Akt phosphorylation and downstream mTOR signaling, reducing cell viability and inducing cell cycle arrest at G0/G1 in carcinoma models. Nanocarrier-enhanced retention ensures sustained exposure—critical for pathway inhibition [113]. Inflammation is another crucial target. Phytochemicals like curcumin and luteolin suppress NF-κB signaling, which regulates pro-inflammatory gene expression. Nano-phytomedicine systems amplify this effect by facilitating cytosolic uptake and nuclear translocation of inhibitory phytochemicals. Liposomal curcumin, for instance, reduces p65 phosphorylation more effectively than free curcumin, leading to downregulation of COX-2, iNOS, TNF-α, and IL-1β [114]. Furthermore, nano-phytomedicines can exert epigenetic effects—modulating histone deacetylases (HDACs), miRNAs, and DNA methylation—to sustain therapeutic responses. Resveratrol-loaded polymeric nanoparticles have been shown to upregulate miR-34a in tumor cells, promoting tumor suppressor gene expression and inhibiting proliferation [115]. Inflammasome activation represents another emerging mechanism. Nanocarriers delivering berberine or curcumin inhibit NLRP3 inflammasome assembly, reducing IL-1β secretion—a valuable action in autoimmune and metabolic diseases [116]. However, achieving specificity in apoptosis and proliferation regulation requires fine-tuning nanoparticle release profiles and cell selectivity. Overactivation of apoptosis in normal tissues can cause toxicity, emphasizing the importance of tumor-targeted nanoparticles and stimuli-responsive release systems. Overall, nano-phytomedicines modulate key cellular regulatory nodes apoptosis, proliferation, and inflammation—through optimized delivery of bioactive compounds. By controlling when, where, and how these pathways are targeted, nanocarrier systems amplify therapeutic efficacy while minimizing harm to healthy tissues [117].

4.4. Interaction with Cellular Signaling

Nano-phytomedicine systems exhibit profound capacity to modulate key intracellular signaling pathways that govern critical cellular processes such as growth, survival, stress response, and inflammation. Encapsulation of phytochemicals enhances their bioavailability and enables targeted delivery, ensuring pathway-specific engagement at therapeutically relevant concentrations [118]. The PI3K/Akt signaling axis plays a central role in cell survival, metabolism, and proliferation, and is often hyperactive in cancer and metabolic disorders. Phytochemicals such as curcumin, resveratrol, and quercetin are known inhibitors of this pathway. Nanoformulations further amplify these effects: for instance, quercetin-loaded polymeric nanoparticles achieve sustained Akt inhibition in breast cancer cells, reducing phosphorylated Akt levels significantly more than free quercetin [119]. Moreover, targeted nanocarriers allow co-delivery of PI3K/Akt inhibitors and phytochemicals, enabling synergistic suppression of proliferative signaling. The MAPK cascade including ERK, JNK, and p38 is triggered in response to mitogenic or stress signals. Phytochemicals like apigenin, kaempferol, and luteolin can selectively modulate MAPK sub-pathways. When delivered via nanoparticles, these compounds achieve higher intracellular concentrations, enhancing suppression of aberrant MAPK signaling. For example, liposomal apigenin reduces ERK phosphorylation in melanoma models, leading to growth arrest and apoptosis [120]. Redox-active phytochemicals in stimuli-responsive carriers also activate stress-activated kinases (e.g., p38, JNK), inducing apoptotic pathways in tumor cells. NF-κB is a master regulator of inflammation and cell survival. Persistent activation is implicated in chronic inflammatory diseases and cancer. Encapsulated curcumin, resveratrol, and EGCG delivered via liposomes or polymeric micelles exhibit enhanced NF-κB inhibition compared to free compounds. Nanocarrier-based delivery results in significant reduction in IκBα degradation and nuclear translocation of NF-κB subunits under inflammatory stimuli [121]. In macrophage and endothelial cell models, nano-phytomedicines reduce downstream cytokine secretion (TNF-α, IL-6), demonstrating functional pathway modulation [122]. By employing nanocarriers, researchers can tailor phytochemical dosing to durably modulate signaling without systemic toxicity. Temporal control via sustained release helps maintain signaling inhibition without triggering compensatory feedback loops. Surface functionalization with targeting ligands ensures preferential delivery to cells overexpressing receptors associated with specific signaling pathways. However, signaling pathway modulation is highly context-dependent. Nanocarrier design must account for tissue-specific expression of receptors and transporter proteins, as well as intracellular enzyme kinetics. Combination strategies that include pathway-specific inhibitors enhance potency but raise considerations about off-target effects and drug–drug interactions. So the, nano-phytomedicines offer precisely controlled modulation of PI3K/Akt, MAPK, and NF-κB pathways via improved delivery and targeting, translating phytochemical bioactivity into potent therapeutic interventions [123]. Table 3 illustrates the multifaceted biological actions mediated by nano-phytomedicine systems, emphasizing their capacity to orchestrate intracellular processes beyond conventional pharmacology. A critical observation is the convergence of these mechanisms toward restoring homeostasis in diseased cells be it through oxidative balance, gene expression modulation, or signaling pathway correction. Importantly, nano-enabled delivery allows precise spatial and temporal control over bioactivity, which is vital when modulating interconnected cellular circuits. For example, regulating redox states through Nrf2 not only suppresses oxidative damage but also affects downstream epigenetic and apoptotic events. Likewise, manipulating epigenetic regulators like DNMTs or HDACs via nanoformulations offers the potential for long-term reprogramming of pathological gene networks a feature highly desirable in chronic diseases or cancer prevention. Additionally, the synergy among various pathways such as simultaneous suppression of PI3K/Akt and NF-κB underscores the systems-level impact nano-phytochemicals can exert. Collectively, this table reflects how nanotechnology transforms phytochemicals into multitargeted therapeutic platforms.

4.5. Epigenetic and Genomic Regulation by Nano-Phytochemicals

Emerging evidence suggests that nano-phytomedicines can regulate gene expression and epigenetic markers, adding another layer to their therapeutic impact. Phytochemicals like curcumin, resveratrol, and sulforaphane exhibit epigenetic actions such as DNA methylation inhibition, histone modification, and microRNA regulation which can be augmented when delivered via nanocarriers [124]. DNA methyltransferases (DNMTs) maintain gene-silencing methyl marks in promoters of tumor suppressor genes. Phytochemicals such as EGCG and sulforaphane are natural DNMT inhibitors. Nanoformulations improve nuclear delivery, increasing intracellular concentration and nuclear accumulation. For instance, PLGA nanoparticles loaded with EGCG reduce DNMT activity and promote re-expression of silenced tumor suppressor genes in prostate cancer cell lines [125]. Sustained-release kinetics help maintain anti-DNMT activity with fewer dosing frequency limitations. Histone acetylation status regulates DNA accessibility. Phytochemicals like curcumin act as histone deacetylase (HDAC) inhibitors, increasing histone acetylation and promoting gene transcription. Enhanced delivery via nano-carriers ensures effective nuclear concentration. Curcumin-loaded nanocarriers have shown greater HDAC inhibition and reactivation of silenced genes in leukemia models compared to free drug [126]. MicroRNAs (miRNAs) regulate multiple genes and pathways simultaneously. Nano-phytomedicines can influence miRNA expression by improving cellular uptake. Resveratrol-loaded polymeric nanoparticles can upregulate tumor-suppressive miRNAs such as miR-34a and downregulate oncogenic miR-21 in cancer cells, leading to reduced proliferation and increased apoptosis [127]. Epigenetic reprogramming via nano-phytomedicines has lasting effects on gene expression. For example, early exposure to curcumin-loaded nanoparticles induces chromatin state alterations in stem cell models, influencing differentiation pathways and disease resistance [128]. These findings highlight the potential of nano-phytomedicines in disease prevention, beyond treatment. Epigenetic modulation affects broad gene networks, raising concerns about unintended gene activation. Controlled release systems help achieve therapeutic concentrations while avoiding epigenetic overstimulation. Tissue-specific targeting assists in avoiding off-target gene regulation. Safety and efficacy evaluation must include genome-wide expression profiling. Nano-phytomedicines’ ability to modulate cellular signaling and epigenetic landscapes adds depth to their pharmacological potential. By providing enhanced delivery to intracellular and nuclear targets, these systems drive therapeutic effects beyond conventional mechanisms, enabling precise and durable regulation of disease-relevant pathways [129].
Figure 4 illustrates the multifaceted molecular mechanisms by which nano-phytomedicines exert their therapeutic effects at the cellular level. Upon administration, nanocarriers facilitate the efficient uptake of encapsulated phytochemicals through endocytosis, often via clathrin- or caveolae-mediated pathways, enabling intracellular trafficking toward endosomes and lysosomes. Once internalized, these nanoformulations modulate oxidative stress by scavenging reactive oxygen species (ROS) and activating the Nrf2 pathway, thereby restoring redox homeostasis and preventing cellular damage. Concurrently, nano-phytochemicals influence cell fate by regulating key apoptotic and proliferative markers, including caspases, Bcl-2 family proteins, and pro-inflammatory cytokines, resulting in anti-inflammatory and anticancer effects. Additionally, these formulations interact with critical signaling cascades such as PI3K/Akt, MAPK, and NF-κB, inhibiting pathological cell survival and inflammatory responses. Importantly, nano-phytomedicines also impact epigenetic and genomic processes by altering DNA methylation, histone modifications, and non-coding RNA expression, enabling long-term modulation of gene expression profiles. This comprehensive modulation underscores their potential as next-generation therapeutics for complex, chronic diseases.

5. Therapeutic Applications in Human Health

5.1. Cancer Therapy: Tumor Targeting and Chemoprevention

Nanocarrier-enabled delivery of phytochemicals offers a multifaceted approach to cancer therapy exemplifying enhanced targeting, controlled release, and synergistic mechanisms:
  • Improved tumor accumulation and retention: Nanocarriers exploit the Enhanced Permeability and Retention (EPR) effect, resulting in higher tumor uptake compared to free compounds. Lipid- and polymer-based nanoformulations of curcumin, resveratrol, and EGCG have shown significantly increased tumor accumulation and therapeutic efficacy in breast, prostate, pancreatic, and colon cancers compared to their native forms [130,131].
  • Targeted delivery and cellular uptake: Surface functionalization strategies—such as folate receptors, peptides, or antibodies—enhance selective cellular uptake. Folic acid-decorated lipid carriers loaded with resveratrol exhibited over twice the uptake in ovarian cancer cells versus controls [132]. Similarly, HER2-targeted polymeric nanoparticles delivering phytochemical paclitaxel precursors demonstrated potent cytotoxicity in breast cancer models [133].
  • Synergistic mechanisms in tumor suppression: Phyto-nanocarriers modulate multiple anticancer pathways including apoptotic induction, cell cycle arrest, anti-angiogenesis, and ferroptosis. For example, carriers inducing ferroptosis—an iron-dependent form of cell death—have been shown to synergize with photodynamic therapies, leveraging ROS accumulation and GPX4 inhibition [134].
  • Clinical translation moving forward: While numerous phyto-nanodrugs are undergoing preclinical evaluation, a subset has entered clinical testing or is nearing approval [135]. The AuroLase™ gold nanoshells (for head and neck, lung, and prostate tumors) exemplify how phytochemical-functionalized inorganic nanocarriers can be translated into the clinic [136].
These nanomedicine strategies transform natural products into precision oncology agents, with improved pharmacokinetics, targeting, and mechanistic diversity for chemoprevention and tumor treatment.

5.2. Cardiovascular Health: Anti-Atherogenic and Antihypertensive Effects

Phytochemical nanoformulations also show promise in the cardiovascular space, particularly in combating atherosclerosis and hypertension:
  • Anti-atherogenic potential: Nanocarrier-delivered polyphenols (like resveratrol and catechins) and terpenoids mitigate oxidative stress and inflammation in endothelial cells. Enhanced bioavailability leads to reduced LDL oxidation, lower vascular smooth muscle proliferation, and decreased macrophage infiltration in atherosclerotic lesions [137,138]. Some systems leverage magnetic nanoparticles to localize phytochemicals to inflamed plaques under external guidance [139].
  • Blood pressure modulation: Nanoformulations such as naringenin-loaded nanocarriers have demonstrated improved vasodilatory effects, enhanced endothelial nitric oxide synthesis, and reduced vascular resistance in hypertensive animal models. Compared to unencapsulated phytochemicals, these nanoformulations required lower doses for similar antihypertensive outcomes [140].
  • Enhanced therapeutic indices: By increasing solubility and stability, nanocarriers enable sustained-release profiles, maintaining therapeutic concentrations over extended periods—crucial for managing chronic cardiovascular conditions [141].
  • Safety and translational outlook: Lipid-based nanoemulsions and polymeric systems exhibit good biocompatibility and low toxicity in vivo. However, comprehensive long-term safety studies are needed. Clinical translational efforts are underway, though in vivo data and human trials are still limited [142].
Overall, nanoformulated phytochemicals offer a multifactorial therapeutic toolkit—antioxidant, anti-inflammatory, lipid-regulating, and antihypertensive—that may complement or improve upon current cardiovascular therapies.

5.3. Neurodegenerative Disorders: Blood–Brain Barrier Penetration and Neuroprotection

Nanoformulated phytochemicals offer a promising solution for managing neurodegenerative diseases by enhancing delivery to the central nervous system (CNS), overcoming the restrictive blood–brain barrier (BBB):
  • Improved BBB permeability: Traditional phytochemicals like curcumin and resveratrol face poor CNS bioavailability due to the semi-permeable BBB. Encapsulation in liposomes or polymeric nanoparticles coated with surfactants (e.g., polysorbate-80) or targeted ligands (e.g., transferrin) has enhanced brain uptake by several-fold in rodent models [143].
  • Neuroprotective effects: Once inside the CNS, these nano-phytomedicines exert antioxidant activity, suppress neuroinflammation, and support mitochondrial function. For example, curcumin-loaded nanoparticles attenuated microglial activation, decreased levels of neurotoxic cytokines (TNF-α, IL-6), and improved cognitive performance in Alzheimer’s disease (AD) models [144].
  • Synergy with anti-aggregation mechanisms: Nano-encapsulated resveratrol and epigallocatechin gallate (EGCG) interfere with amyloid-β aggregation and tau phosphorylation, key pathological events in neurodegeneration. Targeted delivery increased accumulation near amyloid plaques and enhanced reduction in fibril load [145].
  • Translational potential and safety: While preclinical data are promising, translating these technologies into clinical use requires rigorous assessment of long-term toxicity, dose optimization, scalable production, and regulatory compliance for CNS-targeted nanotherapies [146].

5.4. Metabolic Disorders: Anti-Diabetic and Anti-Obesity Actions

Phytochemical-loaded nanocarriers play an increasingly significant role in managing metabolic diseases by addressing bioavailability issues and enabling targeted delivery:
  • Glycemic control and insulin sensitization: Nanosystems encapsulating berberine, quercetin, and cinnamon polyphenols have shown improved intestinal absorption and bioactivity, leading to reduced fasting glucose, enhanced insulin sensitivity, and decreased HbA1c in diabetic animal models [147]. Silver and chitosan nanoparticles aid in increasing pancreatic β-cell survival and functionality through their controlled release characteristics [148].
  • Anti-obesity effects: Nanoformulations that improve the bioavailability of curcumin or catechins have been shown to inhibit adipogenesis, bolster thermogenesis, and reduce fat accumulation in high-fat-diet rodent models [149]. Lipid polymer hybrids loaded with anti-obesity phytochemicals have achieved sustained release in adipose tissues, improving satiety hormone profiles and suppressing inflammatory cytokines [150].
  • Lipid profile modulation: Nano-delivered silymarin and resveratrol have demonstrated significant reductions in LDL and triglycerides, with concurrent increases in HDL cholesterol [151]. Liposomal formulations appear to amplify the cholesterol-lowering mechanisms of phytochemicals.
  • Safety and therapeutic outcomes: Nanocarrier-based approaches typically show good tolerability in preclinical studies; however, long-term analyses are necessary to verify metabolic safety and assess off-target effects in chronic administration settings [152].
Table 4 underscores the diverse and disease-specific therapeutic roles nano-phytomedicines can play when properly engineered and targeted. What stands out is the ability of these nanosystems not only to enhance solubility and stability of phytochemicals but also to modulate complex biological environments such as tumor microenvironments, atherosclerotic plaques, and inflamed neural tissue through spatially controlled delivery. A crucial advantage is their multifunctionality; for instance, many formulations exert both anti-inflammatory and antioxidant effects, which are central to diseases like neurodegeneration and diabetes, highlighting their system-level therapeutic potential. Moreover, the modular nature of nanocarriers permits combinatorial designs, enabling the co-delivery of multiple bioactives or the integration of imaging agents for theranostic applications. As nano-phytomedicine matures, these platforms are increasingly being tailored with stimuli-responsiveness such as pH-, enzyme-, or ROS-triggered release, offering greater precision and minimizing systemic side effects. This versatility places nano-phytomedicine at the forefront of next-generation, integrative, and personalized healthcare approaches.

5.5. Infectious Diseases: Antibacterial, Antiviral, and Antifungal Applications

The delivery of phytochemicals via nanocarriers is emerging as a powerful tool in combating infectious diseases:
  • Antibacterial strategies: Phytochemical-loaded nanoparticles combine the intrinsic antimicrobial properties of metal (e.g., Ag, ZnO) or polymeric materials with the bioactivity of plant compounds. Silver nanoparticle–curcumin conjugates, for instance, exhibit synergistic bactericidal effects against MRSA and E. coli, disrupting biofilm formation and increasing bacterial membrane disruption [157].
  • Antiviral delivery systems: Nano-encapsulation of quercetin, baicalin, or cynanoside enhances antiviral activity against influenza, herpes simplex, and coronaviruses. Phytochemical–gold nanoparticle conjugates block viral entry and replication via actions like ACE2 receptor inhibition or protease binding [153].
  • Antifungal efficacy: Nanocapsules containing eugenol, thymol, and other essential oils maintain effective drug concentrations in target tissues, adding precision to fungal membrane disruption and inhibiting virulence factors [154].
  • Addressing antibiotic resistance: These systems employ multifunctional mechanisms—ROS production, membrane interaction, efflux pump inhibition—delivering therapeutic synergy at lower, safer doses while reducing the potential for resistance development [93].
  • Stability and administration: Encapsulation ensures controlled release, improved bioavailability, and enhanced topical or inhalable delivery, offering versatility in treating cutaneous, pulmonary, and systemic infections [155].
Figure 5 illustrates the diverse therapeutic roles of nano-phytomedicines across major disease domains. Nanocarrier-based delivery systems enhance tumor targeting and chemoprevention in cancer therapy, exert anti-atherogenic and antihypertensive effects in cardiovascular diseases, enable blood–brain barrier penetration for neurodegenerative disorders, improve glycemic control and lipid metabolism in metabolic disorders, and offer potent antibacterial, antiviral, and antifungal actions in infectious disease management. The schematic highlights site-specific interactions that contribute to improved bioavailability and targeted therapeutic action.

6. Pharmacokinetics and Toxicological Considerations

6.1. Bioavailability and Controlled Release Kinetics

Phytochemicals often suffer from poor oral bioavailability due to intrinsic physicochemical limitations such as low aqueous solubility, poor permeability, and extensive first-pass metabolism. Nanocarrier systems have been extensively explored to overcome these barriers and enhance the pharmacokinetic profile of plant-derived bioactives [156]. The encapsulation of phytochemicals into nanoscale vehicles such as liposomes, polymeric nanoparticles, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs) has been demonstrated to significantly improve their solubility, protect against enzymatic degradation, and facilitate controlled and sustained release [158]. Controlled release kinetics is a critical parameter that dictates the therapeutic efficacy and safety profile of nano-phytomedicines. Conventional phytochemical formulations often show rapid clearance and short plasma half-life, limiting their clinical utility [159]. Nanocarriers can be engineered to provide zero-order or sustained release kinetics by manipulating polymer composition, particle size, surface chemistry, and encapsulation efficiency [160]. For instance, PLGA-based nanoparticles degrade hydrolytically, allowing predictable drug release profiles, which can be fine-tuned by adjusting polymer molecular weight and lactide/glycolide ratio [161]. Similarly, SLNs and NLCs leverage lipid matrix crystallinity and composition to control the diffusion of phytochemicals, ensuring prolonged systemic circulation and sustained therapeutic levels [162]. The improved bioavailability conferred by nanocarriers is often attributed to multiple synergistic factors: enhanced dissolution rates due to nanometric size and increased surface area; protection from acidic and enzymatic degradation in the gastrointestinal tract; and facilitated transport across epithelial barriers via endocytosis or paracellular routes [163]. In particular, mucoadhesive polymers like chitosan can transiently open tight junctions, improving paracellular permeability of phytochemicals such as curcumin and quercetin [164]. Pharmacokinetic studies have repeatedly demonstrated that nano-encapsulated phytochemicals achieve higher maximum plasma concentrations (C-max), longer time to reach peak concentration (T-max), and increased area under the curve (AUC) compared to free compounds [165]. For example, liposomal curcumin exhibited a 4- to 5-fold increase in oral bioavailability relative to free curcumin in rodent models [166]. Controlled release formulations also reduce dosing frequency and minimize plasma level fluctuations, enhancing patient compliance and therapeutic outcomes [167]. However, alongside pharmacokinetic enhancements, regulatory scrutiny is increasing regarding excipient safety, nanocarrier degradation products, and release kinetics predictability. Lack of harmonized regulatory guidelines for herbal nanomedicine adds complexity to clinical translation. Formulators must address issues such as reproducibility of release profiles across batches and scale and provide robust in vitro–in vivo correlation (IVIVC) models to gain regulatory acceptance. Controlled release kinetics also pose challenges. Burst release rapid initial release of a significant fraction of the payload may cause toxicity or reduced therapeutic duration [168]. Formulation optimization requires balancing drug loading, carrier stability, and release mechanisms to achieve desired pharmacokinetics. Stimuli-responsive nanocarriers, which release phytochemicals in response to pH, enzymes, temperature, or redox conditions, are promising to further refine delivery precision [169]. In conclusion, nanocarrier-mediated improvements in bioavailability and controlled release kinetics represent a cornerstone of modern phytochemical therapeutics. Nonetheless, for regulatory approval, developers must also meet safety standards related to excipient toxicity, batch consistency, and documentation of long-term stability. Rigorous in vitro–in vivo correlation studies and pharmacokinetic modeling remain essential to optimize formulations for clinical translation, ensuring maximized efficacy with minimized adverse effects [170].

6.2. In Vivo Biodistribution and Organ Targeting

Understanding the in vivo biodistribution of phytochemical-loaded nanocarriers is fundamental to optimizing therapeutic efficacy and minimizing off-target toxicity. Biodistribution studies elucidate how nanoparticles distribute across organs and tissues after administration, shedding light on their pharmacokinetic profiles, clearance mechanisms, and accumulation at target sites. Effective organ targeting enhances drug concentration at disease sites while sparing healthy tissues, thus improving the therapeutic index of nano-phytomedicines [171]. Nanocarriers administered via different routes (oral, intravenous, topical, or pulmonary) exhibit distinct biodistribution patterns governed by size, shape, surface charge, and chemical composition. Generally, nanoparticles sized between 10 and 200 nm achieve optimal circulation time and reduced renal clearance, permitting enhanced extravasation into tissues with compromised vasculature, such as tumors and inflamed sites, through the enhanced permeability and retention (EPR) effect [172]. However, passive targeting via EPR alone is often insufficient for selective delivery, especially in diseases where pathological vasculature heterogeneity limits nanoparticle access [173]. Surface modification with polyethylene glycol (PEG) and other hydrophilic polymers (PEGylation) is widely employed to confer “stealth” properties, reducing protein adsorption (opsonization) and subsequent clearance by the mononuclear phagocyte system (MPS), predominantly in liver and spleen [174]. This prolongs systemic circulation and enhances accumulation at the target site [175]. PEGylated phytochemical nanocarriers have demonstrated increased plasma half-life and improved therapeutic outcomes in preclinical cancer and inflammatory models [176]. Active targeting strategies further enhance biodistribution specificity by decorating nanocarrier surfaces with ligands that bind to receptors overexpressed on diseased cells or tissues. Common targeting moieties include antibodies, peptides, aptamers, and small molecules such as folic acid [177]. For example, folate receptor-targeted nanocarriers delivering resveratrol have shown significantly greater uptake in ovarian cancer tissues compared to non-targeted systems, resulting in improved tumor regression [178]. Similarly, transferrin-modified nanoparticles facilitate efficient blood–brain barrier penetration for neuroprotective phytochemicals [179]. Nanocarrier physicochemical properties also influence biodistribution. Positively charged nanoparticles tend to interact more readily with negatively charged cell membranes but often exhibit rapid clearance and increased toxicity, while neutral or slightly negative surface charges balance circulation time and cellular uptake [180]. Shape and rigidity also affect biodistribution: rod-shaped or flexible particles show enhanced margination and tumor penetration compared to rigid spheres [181]. Organ-specific delivery approaches have been explored to concentrate phytochemicals in specific tissues. For instance, magnetic nanoparticles conjugated with phytochemicals enable magnetic field-guided targeting to tumor or inflamed sites, reducing systemic exposure and toxicity [182]. Similarly, pulmonary delivery of phytochemical-loaded nanoparticles achieves high local concentrations in the lungs for respiratory diseases, bypassing systemic distribution [183]. From a regulatory perspective, demonstrating consistent and reproducible biodistribution profiles across preclinical models is critical for approval. Safety concerns include unintended accumulation in off-target organs such as the liver, kidneys, and spleen, and potential for chronic toxicity or immune activation. Standardized imaging and biodistribution protocols are needed to meet agency expectations for pharmacokinetic validation. Imaging modalities such as fluorescence, MRI, PET, and CT coupled with labeled nanoparticles provide quantitative biodistribution data, enabling real-time monitoring and optimization of delivery systems [184]. These studies have revealed organ accumulation patterns, clearance routes (renal vs. hepatobiliary), and off-target accumulation risks, informing rational nanocarrier design [185]. However, challenges remain in achieving precise targeting, avoiding uptake by off-target organs (e.g., liver, spleen), and translating biodistribution data from animal models to humans due to interspecies physiological differences. Understanding the protein corona formation on nanoparticles in biological fluids is also critical, as it influences biodistribution and cellular interactions [186]. Furthermore, lack of international standardization in labeling methods, imaging endpoints, and biodistribution metrics complicate regulatory harmonization. In summary, in vivo biodistribution and organ targeting govern the success of phytochemical nanocarriers. Integration of passive and active targeting strategies, surface engineering, and advanced imaging techniques enable enhanced therapeutic outcomes with reduced side effects, paving the way for precision nanomedicine applications [187]. Figure 6 encapsulates the integrated pharmacokinetic and toxicological determinants that critically influence the therapeutic efficacy and clinical viability of nano-phytomedicines. Nanocarriers significantly enhance bioavailability by improving the solubility and stability of poorly soluble phytochemicals and enabling controlled and sustained release profiles that mitigate rapid clearance and dosing frequency. The figure emphasizes how strategic nanocarrier design facilitates targeted biodistribution, allowing selective accumulation in diseased tissues such as tumors or inflamed sites while minimizing systemic toxicity. Surface functionalization with ligands or stealth polymers like PEG extends circulation time and improves site-specific uptake, a pivotal factor in achieving therapeutic precision. Moreover, the biocompatibility and immunogenicity assessment segment underlines the need for comprehensive safety profiling, including cytotoxicity, hemocompatibility, and immune system interactions, which are vital to prevent adverse responses. Finally, the figure integrates regulatory considerations, acknowledging the necessity of compliance with international safety guidelines and standardized evaluation protocols to support clinical translation. Together, these elements underscore the multidimensional approach required to develop safe, effective, and regulatory-compliant nano-phytomedicines.

6.3. Biocompatibility and Immunogenicity Assessment

The biocompatibility and immunogenicity of nanocarriers encapsulating phytochemicals are pivotal determinants of their clinical viability and safety. Nanomaterials introduced into the body interact with complex biological systems, potentially triggering immune responses or cytotoxic effects that may limit therapeutic utility or cause adverse reactions [188]. Therefore, rigorous biocompatibility assessment and immunogenicity profiling form integral components of the preclinical evaluation of nano-phytomedicines. Biocompatibility refers to the ability of nanocarriers to perform their desired function without eliciting harmful local or systemic effects. This encompasses cytotoxicity, genotoxicity, hemocompatibility, and effects on normal tissue architecture and function. In vitro cytotoxicity assays using relevant cell lines (e.g., hepatocytes, renal tubular cells, endothelial cells) provide initial screening for nanoparticle safety. Many nanocarriers composed of biodegradable polymers (e.g., PLGA, chitosan) and lipids exhibit low inherent cytotoxicity, attributed to their biocompatible degradation products [189]. For instance, chitosan-based nanoparticles show favorable cellular uptake with minimal cytotoxicity at therapeutic concentrations [190]. Immunogenicity, the ability of nanomaterials to provoke immune activation, is a complex phenomenon involving recognition by the innate immune system, complement activation, cytokine release, and adaptive immune responses. Nanocarriers may be recognized as foreign entities, leading to opsonization, clearance by macrophages, and potential hypersensitivity reactions [191]. The physicochemical characteristics of nanoparticles—size, shape, surface charge, and chemistry critically influence immunogenicity. For example, positively charged nanoparticles often induce stronger immune activation compared to neutral or negatively charged particles [192]. Surface modifications such as PEGylation have been widely adopted to reduce immunogenicity by creating a hydrophilic “stealth” layer that minimizes protein adsorption and macrophage uptake [193]. However, repeated administration of PEGylated nanocarriers can induce anti-PEG antibodies, resulting in accelerated blood clearance and reduced efficacy, a phenomenon known as the “accelerated blood clearance” (ABC) effect [194]. Alternatives such as zwitterionic polymers or biomimetic coatings (e.g., cell membrane camouflage) are under investigation to overcome these limitations [195]. Hemocompatibility assessment evaluates the interaction of nanoparticles with blood components, including erythrocytes, platelets, and coagulation factors. Nanocarriers should not induce hemolysis, platelet aggregation, or coagulation cascade activation to avoid thrombosis or bleeding risks [196]. Several studies report that lipid-based and polymeric phytochemical nanocarriers maintain good hemocompatibility profiles, but careful evaluation is essential for each formulation [197]. In vivo immunotoxicity studies involve monitoring cytokine profiles (e.g., TNF-α, IL-6, IL-1β), complement activation, and histopathological changes in immune organs (spleen, lymph nodes). Animal models reveal that some inorganic nanoparticles (e.g., metal oxides) can induce oxidative stress-mediated immune activation, necessitating dose optimization and surface functionalization [198]. Long-term toxicity studies are indispensable for chronic use phytomedicines, as accumulation or degradation products may cause delayed adverse effects. Nanocarrier biodegradation rates, metabolite toxicity, and potential accumulation in reticuloendothelial system organs require thorough investigation [199]. Regulatory agencies emphasize robust biocompatibility and immunogenicity data in nanomedicine approval pathways. International standards (e.g., ISO 10993) [200] guide preclinical testing frameworks, including in vitro assays, animal studies, and clinical monitoring protocols [201]. Employing standardized methodologies and relevant models ensures reproducibility and safety benchmarking across studies. So the biocompatibility and immunogenicity assessments are crucial to balancing therapeutic benefits and safety risks in nano-phytomedicine development. Rational nanocarrier design, including surface engineering and choice of biocompatible materials, alongside comprehensive preclinical and clinical evaluation, underpins the successful translation of phytochemical nanotherapies to the clinic [202,203].
Table 5 presents a multidimensional perspective on how nanoscale engineering profoundly reshapes the pharmacokinetic behavior and toxicological safety of phytochemical-based therapies. Beyond mere enhancement of solubility or protection from degradation, nanocarriers serve as dynamic modulators of biological interactions governing absorption routes, systemic retention, immune recognition, and metabolic fate. Notably, the strategic manipulation of surface chemistry (e.g., PEGylation, zwitterionic coatings) and morphology (e.g., core-shell vs. matrix) allows for tailored biodistribution and controlled release, aligning therapeutic delivery with pathophysiological needs. However, this sophistication introduces complexity in predicting in vivo responses, as subtle changes in charge, shape, or protein corona formation can shift organotropism or immune compatibility. The integration of nanocarrier adaptability with regulatory-standardized safety assessment, as reflected in ISO-guided protocols, ensures translational viability. In essence, Table 5 underscores that effective nano-phytomedicine design demands not only bioavailability enhancement but a harmonized balance between efficacy, biocompatibility, and immunological invisibility.

6.4. Regulatory Guidelines for Herbal Nanomedicine Safety

The clinical translation of herbal nanomedicines, including phytochemical-loaded nanocarriers, necessitates strict adherence to regulatory frameworks that ensure safety, efficacy, and quality. Regulatory agencies worldwide, such as the U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), and others, have developed evolving guidelines that address the unique challenges posed by nanoscale drug delivery systems, especially those derived from herbal sources [204]. One primary challenge is the inherent complexity of phytochemical extracts, which contain multiple bioactive constituents with variable compositions influenced by botanical source, extraction methods, and batch variations [205]. When these extracts are integrated into nanocarriers, characterization becomes more intricate due to the interplay of nanomaterial properties and herbal matrix complexity. Regulators require comprehensive physicochemical characterization including particle size distribution, surface charge, morphology, chemical composition, and stability to ensure reproducibility and batch consistency [206]. Toxicological evaluation remains paramount. Herbal nanomedicines must undergo rigorous in vitro and in vivo toxicology assessments, including genotoxicity, cytotoxicity, immunotoxicity, and long-term carcinogenicity studies tailored for both the phytochemical components and the nanocarrier materials [207]. The potential for synergistic toxicity arising from nanoparticle–phytochemical interactions necessitates specific investigation [208]. Regulatory guidance encourages the use of standardized testing protocols such as ISO 10993 and OECD Test Guidelines, adapted for nanoformulations [209]. Pharmacokinetic and pharmacodynamic profiling must demonstrate improved bioavailability and therapeutic benefit without compromising safety. Regulatory agencies stress the importance of detailed ADME (absorption, distribution, metabolism, and excretion) studies and the identification of potential toxic metabolites arising from the phytochemical or nanomaterial degradation [210]. Special attention is given to off-target accumulation and long-term retention in organs such as liver, spleen, kidneys, and brain [211]. Clinical trial design for herbal nanomedicines should include safety endpoints specific to nano-bio interactions, immunogenicity monitoring, and post-market surveillance plans [212]. Given the variable regulatory landscapes for botanical drugs and nanomedicines, hybrid regulatory pathways are emerging. For example, the FDA’s Botanical Drug Development Guidance provides a framework for complex herbal products, which can be combined with the agency’s guidance on nanotechnology applications [213]. Quality control is another critical focus. Current Good Manufacturing Practices (cGMP) must be adapted to accommodate nanoscale materials, including control of nanoparticle size, distribution, surface properties, and loading efficiency. Analytical methods like dynamic light scattering (DLS), electron microscopy, high-performance liquid chromatography (HPLC), and mass spectrometry are essential for routine quality assurance [214]. International harmonization efforts are underway to standardize definitions, characterization methods, and safety assessment protocols for herbal nanomedicines. Organizations such as the International Council for Harmonisation (ICH) and ISO play significant roles in developing guidelines that balance innovation and patient safety [215]. Despite these advances, regulatory approval of herbal nanomedicines remains challenging due to the novelty of the field, complexities in standardization, and limited long-term clinical safety data. Collaboration among researchers, industry, and regulators is vital to establish robust frameworks that facilitate safe and effective commercialization of nano-phytomedicines [216]. In summary, regulatory guidelines for herbal nanomedicine safety are evolving to address the complexities of combining botanical therapeutics with nanotechnology. Comprehensive physicochemical, toxicological, pharmacokinetic, and clinical evaluations aligned with international standards are essential to ensure that these innovative therapies reach patients safely and effectively [217].

7. Clinical Advancements and Translational Challenges

7.1. Preclinical Studies and Clinical Trials of Nano-Phytomedicines

The journey from bench to bedside for nano-phytomedicines is grounded in extensive preclinical evaluations, followed by carefully designed clinical trials. Preclinical studies provide foundational insights into the efficacy, pharmacokinetics, biodistribution, toxicity, and mechanisms of action of phytochemical-loaded nanocarriers. Numerous in vitro and in vivo models have demonstrated enhanced therapeutic outcomes when phytochemicals are delivered via nanocarriers compared to free compounds. For example, curcumin-loaded polymeric nanoparticles have exhibited superior anti-inflammatory and anticancer activities in murine models by improving bioavailability and targeting inflammatory pathways [218]. Rodent and non-rodent animal models play an indispensable role in evaluating the safety profiles and therapeutic indices of nano-phytomedicines. Toxicology studies assess acute, subacute, and chronic effects, including immune responses, organ-specific toxicity, and genotoxicity, informing safe dosage ranges for human trials [219]. Pharmacokinetic studies using techniques such as LC-MS/MS and imaging modalities provide quantitative data on absorption, distribution, metabolism, and excretion [220]. Furthermore, advanced models that better mimic human pathophysiology, such as patient-derived xenografts or organ-on-chip systems, are increasingly employed to bridge translational gaps [221]. Despite promising preclinical data, nano-phytomedicines face hurdles in clinical trial advancement. Clinical trials are limited, largely due to regulatory complexities, scale-up challenges, and the need for robust safety data. Regulatory bodies often lack harmonized frameworks specifically tailored to botanical-based nanomedicines, creating ambiguity in classification—whether as pharmaceuticals, nutraceuticals, or supplements. This ambiguity delays approval pathways, especially when phytochemical compositions vary due to source differences, seasonal variation, and extraction methods. Moreover, the inherently complex, multi-component nature of botanical extracts presents challenges in defining active pharmaceutical ingredients (APIs), which is a fundamental requirement for regulatory clearance [222]. Some formulations have reached Phase I and II trials, primarily focusing on cancer and inflammatory diseases. For instance, a liposomal curcumin formulation has undergone Phase II trials for pancreatic cancer, demonstrating safety and preliminary efficacy [222]. Another example is a solid lipid nanoparticle formulation of resveratrol under investigation for metabolic syndrome management [223]. However, achieving reproducibility of these formulations on a commercial scale remains difficult. Variability in plant sources, differences in phytochemical profiles, and diverse manufacturing techniques contribute to batch-to-batch inconsistency. This not only complicates pharmacokinetic and pharmacodynamic profiling but also undermines regulatory confidence in product stability and uniformity [224]. Standardization challenges further compound translational hurdles. Unlike synthetic drugs with a single active compound, phytochemicals often consist of multiple synergistic components, making it difficult to standardize identity, purity, and potency. Robust and validated analytical methods are essential to ensure consistent phytochemical loading and release profiles, yet such methods are still evolving for complex botanical nanocarriers [225]. Additionally, ensuring appropriate characterization of nanoformulation parameters such as particle size, surface charge, encapsulation efficiency, and release kinetics is critical for regulatory submissions but remains underreported or poorly controlled in some studies. Patient variability, including differences in metabolism, immune status, and comorbidities, further complicates clinical trial design and outcome interpretation [225]. Ultimately, preclinical models have robustly demonstrated the therapeutic promise of nano-phytomedicines, but the transition to human trials requires overcoming substantial scientific, regulatory, and manufacturing barriers. Continued development of predictive preclinical platforms, alongside adaptive clinical trial designs, will be crucial to accelerate the clinical translation of these promising therapeutics [226].

7.2. Commercial Products and Patent Landscape

The commercialization of nano-phytomedicines is still nascent but growing steadily, with several products and patents highlighting the field’s progress and intellectual property (IP) dynamics. Commercial products combining phytochemicals with nanocarriers predominantly target areas such as oncology, dermatology, metabolic disorders, and wound healing [227]. Several liposomal, polymeric, and solid lipid nanoparticle-based phytochemical formulations have entered the market, often marketed as dietary supplements or cosmeceuticals with enhanced bioavailability claims. For example, liposomal curcumin and resveratrol products advertise improved absorption and therapeutic potential for general wellness and anti-inflammatory support [228]. However, few nano-phytomedicines have achieved full pharmaceutical regulatory approval due to stringent safety and efficacy requirements. The patent landscape reflects intense innovation activity, with patents covering novel nanocarrier compositions, surface functionalization methods, targeted delivery systems, and controlled release mechanisms. Patent filings from academia and industry reveal focus areas such as multifunctional hybrid nanocarriers, stimuli-responsive release, and combination therapies integrating phytochemicals with conventional drugs [229]. For instance, patents protecting curcumin-loaded polymeric nanoparticles with tumor-targeting ligands have been granted in multiple jurisdictions [230]. However, the multi-component nature of plant-derived extracts introduces unique challenges for IP protection. Unlike synthetic single-molecule drugs, botanical formulations often consist of complex mixtures of bioactive and non-bioactive components that can vary depending on the source, extraction method, and seasonality. This complexity complicates the standardization of formulations and makes it difficult to claim ownership over the exact composition, leading to reliance on process patents or delivery system patents instead of compound-specific protection [231]. Moreover, existing patent frameworks may not sufficiently accommodate the traditional and empirical knowledge embedded in phytomedicine, requiring innovative legal strategies to establish novelty and inventive steps. This can impact the strength and scope of patent protection, potentially shortening the exclusivity period and reducing incentives for commercial investment. In particular, patent examiners may question the reproducibility and stability of phytochemical compositions if variations between batches are not well characterized, creating hurdles during prosecution. Regulatory definitions also impact market entry. Herbal products are often regulated as supplements or traditional medicines, while nanoformulations with therapeutic claims may be classified as drugs, subjecting them to more rigorous approval pathways [232]. This regulatory dichotomy adds another layer of complexity for manufacturers. A single nano-phytomedicine product may fall under different regulatory frameworks depending on jurisdiction, dosage, route of administration, or labeling claims. Companies must often choose between faster but limited supplement registration and slower, more stringent drug approval routes each with distinct implications for clinical evidence requirements, marketing, and labeling. Furthermore, evolving guidelines for nanomedicines complicate submission strategies, as regulators may demand detailed characterization data on nanocarrier composition, stability, and interaction with phytochemicals. Companies must navigate this dual regulatory landscape to successfully commercialize products. Despite challenges, the growing consumer demand for natural and nanotechnology-enhanced health products, coupled with advances in nanomanufacturing and formulation science, is likely to drive expansion in the nano-phytomedicine market [233]. Partnerships between academia, biotech startups, and pharmaceutical companies facilitate translation from patent innovation to market-ready products.
Figure 7 provides a comprehensive schematic representation of the core pharmacokinetic and toxicological parameters that influence the clinical translation of nano-phytomedicines. The figure illustrates how nanocarrier-based delivery systems significantly enhance the bioavailability of phytochemicals by facilitating controlled and sustained release, improving solubility, and protecting the bioactives from enzymatic degradation. Additionally, it highlights targeted organ biodistribution achieved through passive mechanisms like the enhanced permeability and retention (EPR) effect, as well as active targeting strategies involving ligand-functionalized nanocarriers. The integration of biocompatible materials and surface modifications such as PEGylation contributes to reduced immunogenicity and systemic toxicity, which are critical for long-term therapeutic safety. Furthermore, the figure underscores the importance of preclinical safety assessment frameworks aligned with regulatory guidelines to ensure consistency, reproducibility, and risk mitigation in herbal nanomedicine development. This visual encapsulation reinforces the multifaceted role of nanotechnology in optimizing phytochemical pharmacology for clinical applications.

7.3. Scale-Up and Manufacturing Barriers

Translating nano-phytomedicines from laboratory-scale prototypes to industrial-scale manufacturing presents several formidable challenges. Achieving reproducible, scalable, and cost-effective production of nanocarriers encapsulating complex phytochemical mixtures requires addressing formulation, process engineering, and quality control issues [234]. Batch-to-batch consistency is critical for regulatory approval and therapeutic reliability. Variability in raw botanical material composition can affect nanoparticle loading efficiency, stability, and bioactivity. Standardized sourcing, extraction, and quality assessment of phytochemical inputs are prerequisites for manufacturing consistency [235]. One of the most critical bottlenecks lies in the intrinsic heterogeneity of plant-derived raw materials, which exhibit seasonal, geographical, and species-related variability. This directly impacts the reproducibility of nanoparticle characteristics such as size, charge, drug loading capacity, and release profile—parameters essential for therapeutic efficacy and regulatory compliance. Manufacturing processes such as solvent evaporation, nanoprecipitation, high-pressure homogenization, and microfluidics must be optimized for scale without compromising particle size distribution, surface properties, and encapsulation efficiency [236]. Microfluidic systems show promise for continuous, scalable production with precise control over particle characteristics, but integration into industrial workflows remains limited [237]. Sterility, endotoxin control, and removal of residual solvents or surfactants are additional manufacturing hurdles, especially for parenteral formulations [238]. Robust purification and aseptic processing are mandatory, increasing complexity and cost. High-throughput purification, filtration, and post-synthesis stabilization processes often require specialized infrastructure not available in standard pharmaceutical plants, further slowing industrial adoption. Additionally, the cost of scaling these methods particularly when dealing with sensitive and multicomponent phytochemical payloads remains a formidable economic barrier. Analytical characterization methods need adaptation for high-throughput environments, enabling rapid assessment of nanoparticle size, polydispersity, surface charge, and payload content [239]. Process analytical technologies (PAT) can facilitate real-time monitoring and control, supporting quality by design (QbD) principles [240]. Regulatory expectations emphasize GMP compliance, documentation, and validation of manufacturing protocols. Facilities require significant capital investment and specialized expertise in nanotechnology production [241]. Infrastructural deficiencies, especially in regions lacking established nanomanufacturing ecosystems, impede technology transfer and localization of nano-phytomedicine production. Moreover, interdisciplinary knowledge gaps between natural product chemists and nanofabrication engineers exacerbate process inefficiencies. Moreover, the environmental impact of nanoparticle manufacturing, including solvent use and waste disposal, calls for green chemistry and sustainable manufacturing practices [242]. In sum, overcoming scale-up and manufacturing barriers involves a multidisciplinary approach integrating materials science, process engineering, quality control, and regulatory compliance. Collaborative efforts and technological innovations are essential to bring nano-phytomedicines to large-scale production and market availability [243].

7.4. Cost, Accessibility, and Acceptance in Healthcare Systems

Cost-effectiveness, accessibility, and acceptance within healthcare systems are critical factors influencing the successful adoption of nano-phytomedicines. Although nanotechnology can improve phytochemical therapeutic profiles, the added complexity and manufacturing demands often increase production costs compared to traditional formulations [244]. Economic evaluations must consider not only manufacturing expenses but also potential healthcare savings through improved efficacy, reduced side effects, and decreased dosing frequency [245]. Cost–benefit analyses that incorporate quality-adjusted life years (QALYs) and real-world patient outcomes will guide reimbursement and formulary decisions. Beyond direct costs, hidden economic burdens such as supply chain gaps, import tariffs on nanomaterials, and limited reimbursement policies for natural product-based therapies can significantly hinder clinical integration, particularly in resource-constrained settings. Accessibility in low- and middle-income countries may be hindered by infrastructure limitations, lack of technical expertise, and regulatory disparities. Simplifying formulation processes and leveraging scalable manufacturing technologies could enhance global accessibility [246]. However, widespread clinical deployment is further hampered by sociocultural and systemic challenges, including mistrust in novel technologies, skepticism toward herbal medicines in technologically advanced healthcare settings, and insufficient training of healthcare professionals in nanomedicine. These factors can reduce prescribing confidence and impact adoption rates. Healthcare professional and patient acceptance depend on demonstrated clinical benefits, safety, and clear communication regarding nanotechnology’s advantages and risks. Concerns about nanoparticle toxicity or unfamiliarity with nanomedicine can influence prescribing behavior and patient compliance [247]. Education and awareness programs targeting clinicians, pharmacists, and patients are necessary to build trust and understanding. Equally important is the integration of nano-phytomedicine education into medical and pharmaceutical curricula to equip future practitioners with the knowledge required to evaluate and apply these technologies effectively. Without such systemic educational reform, the innovation-to-application gap may persist. Integration into evidence-based guidelines and endorsement by professional societies will further facilitate acceptance [248]. Lastly, ethical considerations around equitable access and affordability must be addressed to prevent disparities in nano-phytomedicine utilization [249]. Public–private partnerships that support local manufacturing, reduce dependency on imports, and subsidize development costs for essential nano-phytomedicines can improve affordability. Simultaneously, equitable licensing and access frameworks must be promoted to prevent health inequality. Strategic collaborations among stakeholders, including governments, industry, and academia, are essential to navigate cost and acceptance challenges and realize the full potential of nano-phytomedicines in healthcare systems worldwide [250]. Table 6 illustrates the multidimensional progression of nano-phytomedicine from laboratory research to real-world healthcare implementation, highlighting how each stage of development presents both promise and complexity. While preclinical data have validated the potential of nanocarriers to improve phytochemical efficacy and pharmacokinetics, transitioning to clinical success demands more than therapeutic performance—it requires regulatory foresight, formulation standardization, and predictive translational models. The commercialization landscape, though growing, is hampered by the nuanced classification of products and the inherent challenges in protecting phytochemical intellectual property. Furthermore, manufacturing hurdles such as nanoparticle reproducibility, sterility, and regulatory-grade quality assurance create bottlenecks for scalability. At the systemic level, cost, education, and equitable access influence how these innovations are integrated into standard care. Together, these insights underscore that clinical translation of nano-phytomedicines must be navigated through a holistic lens one that merges bioengineering, regulatory science, public health policy, and patient-centric strategies to ensure global therapeutic impact.

8. Future Prospects and Emerging Trends

8.1. Smart and Responsive Nano-Phytomedicine Systems

Smart and responsive nano-phytomedicine systems represent a transformative frontier in targeted and controlled therapeutic delivery. These advanced nanocarriers are engineered to respond to specific endogenous or exogenous stimuli, allowing on-demand release of phytochemicals with improved spatiotemporal precision and reduced off-target effects [251]. Such stimuli-responsive systems can be designed to react to changes in pH, temperature, redox conditions, enzymatic activity, magnetic or electric fields, and light irradiation. In pathological environments, such as tumor tissues or sites of inflammation, microenvironmental triggers like acidic pH or elevated levels of reactive oxygen species (ROS) provide natural stimuli for smart nanocarrier activation [252]. For example, pH-sensitive polymeric nanoparticles encapsulating curcumin have demonstrated enhanced release specifically in acidic tumor microenvironments, minimizing systemic exposure and improving antitumor efficacy [253]. Similarly, redox-responsive systems exploit the higher intracellular glutathione concentration within cancer cells to trigger rapid phytochemical release from disulfide bond-containing nanocarriers [254]. Externally triggered systems offer the advantage of precise temporal control. Magnetic nanoparticles loaded with phytochemicals enable magnetically guided delivery and heat-triggered release (magneto-thermal therapy) [255] Light-responsive nanoformulations use near-infrared (NIR) irradiation to induce localized release, combining photothermal or photodynamic therapy with phytochemical activity [256]. Enzyme-responsive carriers leverage overexpressed enzymes such as matrix metalloproteinases (MMPs) in diseased tissues to initiate drug release, allowing selectivity and minimizing damage to healthy cells [257]. Integration of multiple stimuli-responsive elements within a single nanoplatform is gaining traction, enabling synergistic release mechanisms tailored to complex disease microenvironments [258]. This multi-responsive behavior enhances therapeutic efficacy and circumvents drug resistance mechanisms. Despite promising preclinical results, challenges remain for clinical translation, including ensuring biocompatibility of stimuli-sensitive materials, scalability of complex formulations, and reproducible stimuli responsiveness in heterogeneous human tissues [259]. Advanced characterization techniques and computational modeling aid in optimizing responsiveness and predicting in vivo behavior. In summary, smart and responsive nano-phytomedicine systems herald a new era of precision therapy by coupling the natural therapeutic benefits of phytochemicals with advanced stimuli-sensitive nanotechnology, offering improved efficacy, safety, and patient compliance [260].

8.2. Artificial Intelligence and Computational Modeling in Formulation Design

Artificial intelligence (AI) and computational modeling have emerged as indispensable tools accelerating the rational design and optimization of nano-phytomedicine formulations. By leveraging machine learning (ML), deep learning, and advanced simulation algorithms, researchers can predict nanocarrier properties, drug release kinetics, and biological interactions, reducing trial-and-error experimentation and expediting development timelines [261]. AI-driven models analyze vast datasets from formulation experiments, physicochemical characterizations, and biological assays to identify critical parameters influencing nanoparticle size, stability, drug loading, and release profiles [262]. For example, ML algorithms have been successfully applied to optimize polymeric nanoparticle formulations encapsulating quercetin by predicting optimal polymer ratios and processing conditions to maximize encapsulation efficiency and controlled release [263]. Computational fluid dynamics (CFD) simulations and molecular dynamics (MD) modeling provide atomistic insights into nanoparticle assembly, phytochemical-polymer interactions, and membrane penetration mechanisms [264]. Such simulations enable the rational selection of materials and design features to enhance bioavailability and targeting. AI also facilitates predictive toxicology by modeling potential adverse effects based on nanoparticle physicochemical properties, helping to screen formulations before in vivo testing [265]. In the clinical realm, AI models analyze patient data to predict individual responses to nano-phytomedicines, supporting personalized treatment strategies [266]. Integration of AI with high-throughput experimental platforms enables rapid iterative design cycles, drastically reducing development costs and timelines [267]. Furthermore, natural language processing (NLP) tools mine scientific literature and patent databases to identify novel phytochemicals and nanocarrier combinations with therapeutic potential [268]. Challenges include the need for high-quality, standardized datasets and the interpretability of AI models to ensure reliable decision-making [269]. Interdisciplinary collaboration among formulation scientists, data scientists, and clinicians is essential to realize the full potential of AI in nano-phytomedicine design. Overall, AI and computational modeling constitute a paradigm shift toward data-driven, efficient, and precision formulation of nano-phytomedicines, accelerating their translation from concept to clinical reality [270].

8.3. Personalized Nano-Phytotherapy and Precision Medicine Approaches

The concept of personalized medicine tailoring therapeutic interventions to individual genetic, phenotypic, and environmental profiles is gaining momentum in phytomedicine, with nanoformulations playing a crucial enabling role [271]. Personalized nano-phytotherapy seeks to optimize phytochemical dosage, delivery, and combinations based on patient-specific characteristics, maximizing efficacy while minimizing adverse effects. Variability in patient metabolism, gut microbiota composition, immune status, and disease heterogeneity influences phytochemical pharmacokinetics and pharmacodynamics [272]. Nanocarriers can be engineered to address these interindividual differences by modulating release kinetics, targeting capabilities, and combination therapies. Advanced diagnostic tools, including genomic sequencing, metabolomics, and biomarker profiling, facilitate patient stratification and identification of responders to specific nano-phytomedicines [273]. For example, patients with specific polymorphisms affecting curcumin metabolism may benefit from formulations with enhanced stability or targeted delivery to bypass metabolic degradation. Combining nano-phytomedicines with companion diagnostics enables real-time monitoring of therapeutic response and dynamic adjustment of dosing regimens [274]. Theranostic nanocarriers integrating imaging agents with phytochemicals allow simultaneous diagnosis and treatment, enhancing precision [275]. Emerging technologies such as 3D bioprinting and microfluidic organ-on-chip platforms enable preclinical testing of personalized nano-phytomedicines on patient-derived cells and tissues, improving prediction of clinical outcomes [276]. However, challenges include the complexity of integrating multi-omics data into treatment decisions, the need for robust clinical validation, and ethical considerations around patient data privacy [277]. Regulatory frameworks must evolve to accommodate personalized phytotherapy approaches and facilitate their clinical adoption. In conclusion, personalized nano-phytotherapy represents a promising convergence of nanotechnology, phytochemistry, and precision medicine, offering tailored interventions that optimize patient outcomes and heralding a new standard in natural product therapeutics [278].

8.4. Sustainable and Green Nanotechnology Integration

Sustainability and environmental considerations are increasingly critical in the development of nano-phytomedicines, prompting integration of green chemistry principles and eco-friendly nanotechnologies [279]. Sustainable nanotechnology aims to minimize environmental impact throughout the life cycle of nanocarriers—from raw material sourcing and synthesis to manufacturing, usage, and disposal [280]. Green synthesis methods employing plant extracts, bacteria, fungi, or algae as reducing and stabilizing agents have gained prominence for fabricating metallic and polymeric nanoparticles, avoiding toxic chemicals and high-energy processes [281]. Such biogenic nanoparticles often display enhanced biocompatibility and therapeutic efficacy due to their surface biomolecules and eco-friendly synthesis routes [282]. In addition, solvent-free or aqueous-based nanoparticle fabrication methods reduce hazardous waste and occupational risks [283]. Utilizing renewable and biodegradable polymers (e.g., chitosan, alginate, cellulose derivatives) as nanocarrier matrices aligns with circular economy principles and reduces persistence of nano-waste in the environment [284]. Life cycle assessment (LCA) tools are increasingly applied to evaluate the environmental footprint of nano-phytomedicine production, guiding sustainable design and manufacturing decisions [285]. Strategies such as energy-efficient synthesis, waste valorization, and green purification technologies contribute to reducing carbon footprint and resource consumption [286]. Moreover, regulatory agencies and funding bodies encourage development of sustainable nanomedicines, emphasizing responsible innovation and societal impact [287]. Public perception and acceptance also favor environmentally conscious products, enhancing market potential. However, regulatory acceptance of green-synthesized nanoformulations is still evolving, as the reproducibility, consistency, and scalability of biogenic synthesis remain key challenges. Safety concerns arise from batch-to-batch variability and the undefined nature of surface biomolecules, which complicate toxicological assessment and regulatory classification. Standardization of green synthesis protocols and characterization techniques is necessary to ensure compliance with Good Manufacturing Practice (GMP) guidelines and to satisfy the quality control demands of regulatory authorities. Additionally, nanocarriers developed through green routes must undergo rigorous preclinical testing to confirm their long-term safety and environmental impact, especially when biodegradable carriers are involved in systemic administration. Challenges include balancing sustainability with cost, scalability, and maintaining high-quality standards for therapeutic efficacy [288]. Furthermore, internationally harmonized regulatory frameworks specific to green nanomedicine are currently lacking, creating uncertainty in approval pathways. As a result, developers must proactively engage with regulatory bodies during early development stages to align their eco-friendly approaches with existing guidelines and anticipate future requirements. Continuous innovation, interdisciplinary collaboration, and policy support are essential to embed sustainability in nano-phytomedicine development fully. In summary, integrating sustainable and green nanotechnology principles into nano-phytomedicine research and production ensures that advances in natural product therapeutics align with global environmental and societal goals, fostering responsible innovation and long-term viability [289]. A parallel emphasis on regulatory preparedness and quality assurance will be pivotal to transforming green innovations into clinically approved, market-ready nano-phytomedicine products. Table 7 highlights how nano-phytomedicine is evolving beyond conventional delivery systems toward highly intelligent, patient-specific, and environmentally conscious platforms. The integration of external triggers and smart responses reflects a move toward greater therapeutic precision. Meanwhile, computational approaches are reshaping formulation design, offering data-driven efficiency. Personalized strategies underscore the shift toward individualized care, while eco-friendly fabrication methods respond to growing demands for sustainability in healthcare innovation. These trends collectively represent a multidimensional advancement in both scientific rigor and translational potential.

9. Conclusions

In recent years, the convergence of phytochemistry and nanotechnology has ushered in a new paradigm in natural product therapeutics. The encapsulation of plant-derived bioactive compounds within diverse nanocarrier systems has substantially enhanced their solubility, stability, bioavailability, and targeted delivery capabilities. Advances in smart and stimuli-responsive nanocarriers have enabled controlled, site-specific release, maximizing therapeutic efficacy while minimizing systemic toxicity. Additionally, integration of computational modeling and artificial intelligence has accelerated formulation optimization and personalized treatment approaches. Collectively, these innovations have elevated nano-phytomedicines from traditional herbal remedies to precision nanotherapeutics with vast potential across a broad spectrum of diseases. Despite significant progress, challenges remain that hinder the full clinical translation of nano-phytomedicines. Complexities in standardizing botanical extracts, achieving scalable and reproducible manufacturing, and ensuring long-term safety and regulatory compliance persist as key obstacles. Furthermore, limited clinical trial data and variability in patient responses call for more robust validation frameworks. However, these limitations present opportunities for interdisciplinary collaboration, innovative formulation strategies, and integration of emerging technologies such as machine learning and advanced diagnostic tools. Continued efforts to address these gaps will unlock the full therapeutic potential of nano-phytomedicines and enable their broader acceptance within mainstream healthcare. The future integration of nano-phytomedicines into global healthcare systems promises to redefine natural product-based therapy by combining traditional wisdom with cutting-edge nanotechnology and personalized medicine. By overcoming current barriers and aligning development with sustainability and accessibility goals, nano-phytomedicines can offer cost-effective, safe, and efficacious treatment alternatives, especially for chronic and complex diseases. Their successful adoption will depend on rigorous scientific validation, clear regulatory pathways, and education to foster acceptance among clinicians and patients. Ultimately, the synergy of phytochemistry and nanomedicine holds great promise for advancing holistic, patient-centered care worldwide.

Author Contributions

N.P. and M.A. jointly led the conceptual framework of the study, prepared the initial manuscript draft, contributed to software implementation, and were actively involved in its critical revision and refinement. S.W.J. handled the validation of the Review, curated visual illustrations, and played a key role in the manuscript’s revision and editorial review. T.K.M. contributed to the conceptual planning, supported software and data validation, designed visual representations, and wrote substantial portions of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Research Foundation of Korea (NRF), supported by the Korean government, under the grant number NRF-2019R1A5A8080290.

Conflicts of Interest

The authors declare that there are no competing financial or personal interests that could have influenced the work reported in this manuscript.

References

  1. Paul, J.K.; Azmal, M.; Haque, A.S.N.B.; Talukder, O.F.; Meem, M.; Ghosh, A. Phytochemical-Mediated Modulation of Signaling Pathways: A Promising Avenue for Drug Discovery. Adv. Redox Res. 2024, 13, 100113. [Google Scholar] [CrossRef]
  2. Chaachouay, N.; Zidane, L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. Drugs Drug Candidates 2024, 3, 184–207. [Google Scholar] [CrossRef]
  3. Garcia-Oliveira, P.; Otero, P.; Pereira, A.G.; Chamorro, F.; Carpena, M.; Echave, J.; Fraga-Corral, M.; Simal-Gandara, J.; Prieto, M.A. Status and Challenges of Plant-Anticancer Compounds in Cancer Treatment. Pharmaceuticals 2021, 14, 157. [Google Scholar] [CrossRef]
  4. Herdiana, Y. Nanoparticles of Natural Product-Derived Medicines: Beyond the Pandemic. Heliyon 2025, 11, e42739. [Google Scholar] [CrossRef] [PubMed]
  5. Nicolaescu, O.E.; Belu, I.; Mocanu, A.G.; Manda, V.C.; Rău, G.; Pîrvu, A.S.; Ionescu, C.; Ciulu-Costinescu, F.; Popescu, M.; Ciocîlteu, M.V. Cyclodextrins: Enhancing Drug Delivery, Solubility and Bioavailability for Modern Therapeutics. Pharmaceutics 2025, 17, 288. [Google Scholar] [CrossRef] [PubMed]
  6. Devaraji, M.; Thanikachalam, P.V. Phytoconstituents as Emerging Therapeutics for Breast Cancer: Mechanistic Insights and Clinical Implications. Cancer Pathog. Ther. 2025, in press. [CrossRef]
  7. El-Saadony, M.T.; Yang, T.; Korma, S.A.; Sitohy, M.; Abd El-Mageed, T.A.; Selim, S.; Al Jaouni, S.K.; Salem, H.M.; Mahmmod, Y.; Soliman, S.M.; et al. Impacts of Turmeric and Its Principal Bioactive Curcumin on Human Health: Pharmaceutical, Medicinal, and Food Applications: A Comprehensive Review. Front. Nutr. 2023, 9, 1040259. [Google Scholar] [CrossRef]
  8. de Luna, F.C.F.; Ferreira, W.A.S.; Casseb, S.M.M.; de Oliveira, E.H.C. Anticancer Potential of Flavonoids: An Overview with an Emphasis on Tangeretin. Pharmaceuticals 2023, 16, 1229. [Google Scholar] [CrossRef]
  9. Goktas, Z.; Zu, Y.; Abbasi, M.; Galyean, S.; Wu, D.; Fan, Z.; Wang, S. Recent Advances in Nanoencapsulation of Phytochemicals to Combat Obesity and Its Comorbidities. J. Agric. Food Chem. 2020, 68, 8119–8131. [Google Scholar] [CrossRef]
  10. Zhuo, Y.; Zhao, Y.-G.; Zhang, Y. Enhancing Drug Solubility, Bioavailability, and Targeted Therapeutic Applications through Magnetic Nanoparticles. Molecules 2024, 29, 4854. [Google Scholar] [CrossRef]
  11. Parvin, N.; Mandal, T.; Roy, P. Soluble Nanoparticles Versatile Tools for Plant Tissue Imaging. J. Bionanoscience 2013, 7, 256–259. [Google Scholar] [CrossRef]
  12. Parvin, N.; Kumar, V.; Joo, S.W.; Park, S.-S.; Mandal, T.K. Recent Advances in the Characterized Identification of Mono-to-Multi-Layer Graphene and Its Biomedical Applications: A Review. Electronics 2022, 11, 3345. [Google Scholar] [CrossRef]
  13. Parvin, N.; Mandal, T.K. Dually Emissive P,N-Co-Doped Carbon Dots for Fluorescent and Photoacoustic Tissue Imaging in Living Mice. Microchim. Acta 2017, 184, 1117–1125. [Google Scholar] [CrossRef]
  14. Parvin, N.; Mandal, T.K. Synthesis of a Highly Fluorescence Nitrogen-Doped Carbon Quantum Dots Bioimaging Probe and Its In Vivo Clearance and Printing Applications. RSC Adv. 2016, 6, 18134–18140. [Google Scholar] [CrossRef]
  15. Mandal, T.K.; Parvin, N. Rapid Detection of Bacteria by Carbon Quantum Dots. J. Biomed. Nanotechnol. 2011, 7, 846–848. [Google Scholar] [CrossRef]
  16. Mandal, T.K.; Parvin, N.; Joo, S.W. PH-Responsive Biocompatible Fluorescent Core-Shell Nanogel for Intracellular Imaging and Control Drug Release. Part. Part. Syst. Charact. 2021, 38, 2100110. [Google Scholar] [CrossRef]
  17. Islam, S.; Ahmed, M.M.S.; Islam, M.A.; Hossain, N.; Chowdhury, M.A. Advances in Nanoparticles in Targeted Drug Delivery–A Review. Results Surf. Interfaces 2025, 19, 100529. [Google Scholar] [CrossRef]
  18. Parvin, N.; Mandal, T.K.; Joo, S.-W. The Impact of COVID-19 on RNA Therapeutics: A Surge in Lipid Nanoparticles and Alternative Delivery Systems. Pharmaceutics 2024, 16, 1366. [Google Scholar] [CrossRef]
  19. Parvin, N.; Joo, S.W.; Mandal, T.K. Enhancing Vaccine Efficacy and Stability: A Review of the Utilization of Nanoparticles in MRNA Vaccines. Biomolecules 2024, 14, 1036. [Google Scholar] [CrossRef]
  20. Chen, Y.; Lu, Y.; Lee, R.J.; Xiang, G. Nano Encapsulated Curcumin: And Its Potential for Biomedical Applications. Int. J. Nanomed. 2020, 15, 3099–3120. [Google Scholar] [CrossRef]
  21. Bajracharya, R.; Song, J.G.; Patil, B.R.; Lee, S.H.; Noh, H.-M.; Kim, D.-H.; Kim, G.-L.; Seo, S.-H.; Park, J.-W.; Jeong, S.H.; et al. Functional Ligands for Improving Anticancer Drug Therapy: Current Status and Applications to Drug Delivery Systems. Drug Deliv. 2022, 29, 1959–1970. [Google Scholar] [CrossRef] [PubMed]
  22. Rauf, A.; Ahmad, Z.; Ajaj, R.; Zhang, H.; Ibrahim, M.; Muhammad, N.; Al-Awthan, Y.S.; Bahattab, O.S.; Ullah, I. Green Synthesis an Eco-Friendly Route for the Synthesis of Iron Oxide Nanoparticles Using Aqueous Extract of Thevetia Peruviana and Their Biological Activities. Sci. Rep. 2025, 15, 18316. [Google Scholar] [CrossRef]
  23. Parvin, N.; Joo, S.W.; Mandal, T.K. Biodegradable and Stimuli-Responsive Nanomaterials for Targeted Drug Delivery in Autoimmune Diseases. J. Funct. Biomater. 2025, 16, 24. [Google Scholar] [CrossRef] [PubMed]
  24. Kashyap, B.K.; Singh, V.V.; Solanki, M.K.; Kumar, A.; Ruokolainen, J.; Kesari, K.K. Smart Nanomaterials in Cancer Theranostics: Challenges and Opportunities. ACS Omega 2023, 8, 14290–14320. [Google Scholar] [CrossRef] [PubMed]
  25. Teli, D.; Satasia, R.; Patel, V.; Nair, R.; Khatri, R.; Gala, D.; Balar, P.C.; Patel, K.; Sharma, A.; Vadodariya, P.; et al. Nature Meets Technology: Harnessing Nanotechnology to Unleash the Power of Phytochemicals. Clin. Tradit. Med. Pharmacol. 2024, 5, 200139. [Google Scholar] [CrossRef]
  26. Ciupei, D.; Colişar, A.; Leopold, L.; Stănilă, A.; Diaconeasa, Z.M. Polyphenols: From Classification to Therapeutic Potential and Bioavailability. Foods 2024, 13, 4131. [Google Scholar] [CrossRef]
  27. Jomova, K.; Alomar, S.Y.; Valko, R.; Liska, J.; Nepovimova, E.; Kuca, K.; Valko, M. Flavonoids and Their Role in Oxidative Stress, Inflammation, and Human Diseases. Chem. Biol. Interact. 2025, 413, 111489. [Google Scholar] [CrossRef]
  28. Bhambhani, S.; Kondhare, K.R.; Giri, A.P. Diversity in Chemical Structures and Biological Properties of Plant Alkaloids. Molecules 2021, 26, 3374. [Google Scholar] [CrossRef]
  29. Kamran, S.; Sinniah, A.; Abdulghani, M.A.M.; Alshawsh, M.A. Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A Scoping Review. Cancers 2022, 14, 1100. [Google Scholar] [CrossRef]
  30. Kubatka, P.; Mazurakova, A.; Samec, M.; Koklesova, L.; Zhai, K.; AL-Ishaq, R.; Kajo, K.; Biringer, K.; Vybohova, D.; Brockmueller, A.; et al. Flavonoids against Non-Physiologic Inflammation Attributed to Cancer Initiation, Development, and Progression—3PM Pathways. EPMA J. 2021, 12, 559–587. [Google Scholar] [CrossRef]
  31. Botelho, A.F.M.; Pierezan, F.; Soto-Blanco, B.; Melo, M.M. A Review of Cardiac Glycosides: Structure, Toxicokinetics, Clinical Signs, Diagnosis and Antineoplastic Potential. Toxicon 2019, 158, 63–68. [Google Scholar] [CrossRef]
  32. Bhuyan, U.; Handique, J.G. Plant Polyphenols as Potent Antioxidants: Highlighting the Mechanism of Antioxidant Activity and Synthesis/Development of Some Polyphenol Conjugates. In Bioactive Natural Products; Elsevier: Amsterdam, The Netherlands, 2022; pp. 243–266. [Google Scholar]
  33. Mokra, D.; Joskova, M.; Mokry, J. Therapeutic Effects of Green Tea Polyphenol (−)-Epigallocatechin-3-Gallate (EGCG) in Relation to Molecular Pathways Controlling Inflammation, Oxidative Stress, and Apoptosis. Int. J. Mol. Sci. 2022, 24, 340. [Google Scholar] [CrossRef] [PubMed]
  34. Cheng, C.; Sui, B.; Wang, M.; Hu, X.; Shi, S.; Xu, P. Carrier-Free Nanoassembly of Curcumin–Erlotinib Conjugate for Cancer Targeted Therapy. Adv. Healthc. Mater. 2020, 9, e2001128. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, M.; Chen, Z.; Huang, D.; Sun, C.; Xie, J.; Chen, T.; Zhao, X.; Huang, Y.; Li, D.; Wu, B.; et al. Myricetin Inhibits TNF-α-Induced Inflammation in A549 Cells via the SIRT1/NF-ΚB Pathway. Pulm. Pharmacol. Ther. 2020, 65, 102000. [Google Scholar] [CrossRef] [PubMed]
  36. Cheng, S.-C.; Huang, W.-C.; Pang, J.-H.S.; Wu, Y.-H.; Cheng, C.-Y. Quercetin Inhibits the Production of IL-1β-Induced Inflammatory Cytokines and Chemokines in ARPE-19 Cells via the MAPK and NF-ΚB Signaling Pathways. Int. J. Mol. Sci. 2019, 20, 2957. [Google Scholar] [CrossRef]
  37. Khare, T.; Anand, U.; Dey, A.; Assaraf, Y.G.; Chen, Z.-S.; Liu, Z.; Kumar, V. Exploring Phytochemicals for Combating Antibiotic Resistance in Microbial Pathogens. Front. Pharmacol. 2021, 12, 720726. [Google Scholar] [CrossRef]
  38. Singh, M.; Devi, S.; Rana, V.S.; Mishra, B.B.; Kumar, J.; Ahluwalia, V. Delivery of Phytochemicals by Liposome Cargos: Recent Progress, Challenges and Opportunities. J. Microencapsul. 2019, 36, 215–235. [Google Scholar] [CrossRef]
  39. Stănciuc, N.; Râpeanu, G. Kinetics of Phytochemicals Degradation During Thermal Processing of Fruits Beverages. In Non-Alcoholic Beverages; Elsevier: Amsterdam, The Netherlands, 2019; pp. 407–440. [Google Scholar]
  40. Nikou, T.; Sakavitsi, M.E.; Kalampokis, E.; Halabalaki, M. Metabolism and Bioavailability of Olive Bioactive Constituents Based on In Vitro, In Vivo and Human Studies. Nutrients 2022, 14, 3773. [Google Scholar] [CrossRef]
  41. Rahat, I.; Yadav, P.; Singhal, A.; Fareed, M.; Purushothaman, J.R.; Aslam, M.; Balaji, R.; Patil-Shinde, S.; Rizwanullah, M. Polymer Lipid Hybrid Nanoparticles for Phytochemical Delivery: Challenges, Progress, and Future Prospects. Beilstein J. Nanotechnol. 2024, 15, 1473–1497. [Google Scholar] [CrossRef]
  42. Zhao, Q.; Chen, Y.; Huang, W.; Zhou, H.; Zhang, W. Drug-Microbiota Interactions: An Emerging Priority for Precision Medicine. Signal Transduct. Target. Ther. 2023, 8, 386. [Google Scholar] [CrossRef]
  43. Lu, H.; Zhang, S.; Wang, J.; Chen, Q. A Review on Polymer and Lipid-Based Nanocarriers and Its Application to Nano-Pharmaceutical and Food-Based Systems. Front. Nutr. 2021, 8, 783831. [Google Scholar] [CrossRef]
  44. Vosoughi, P.; Naghib, S.M.; Jafari, T.; Kangarshahi, B.M. Chitosan-Encapsulated Lipid-Based Nanovesicles for Therapeutic Applications and Tissue Engineering: A Review. Carbohydr. Polym. Technol. Appl. 2025, 10, 100805. [Google Scholar] [CrossRef]
  45. Liu, Y.; Liang, Y.; Yuhong, J.; Xin, P.; Han, J.L.; Du, Y.; Yu, X.; Zhu, R.; Zhang, M.; Chen, W.; et al. Advances in Nanotechnology for Enhancing the Solubility and Bioavailability of Poorly Soluble Drugs. Drug Des. Dev. Ther. 2024, 18, 1469–1495. [Google Scholar] [CrossRef]
  46. Ahmad, K.; Zhang, Y.; Chen, P.; Yang, X.; Hou, H. Chitosan Interaction with Stomach Mucin Layer to Enhances Gastric Retention and Mucoadhesive Properties. Carbohydr. Polym. 2024, 333, 121926. [Google Scholar] [CrossRef]
  47. Mura, P.; Maestrelli, F.; Cirri, M.; Mennini, N. Multiple Roles of Chitosan in Mucosal Drug Delivery: An Updated Review. Mar. Drugs 2022, 20, 335. [Google Scholar] [CrossRef] [PubMed]
  48. Bavnhøj, C.G.; Knopp, M.M.; Löbmann, K. Effect of Drug Loading in Mesoporous Silica on Amorphous Stability and Performance. Pharmaceutics 2024, 16, 163. [Google Scholar] [CrossRef] [PubMed]
  49. Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, Composition, Types, and Clinical Applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef] [PubMed]
  50. Schmitt, C.; Lechanteur, A.; Cossais, F.; Bellefroid, C.; Arnold, P.; Lucius, R.; Held-Feindt, J.; Piel, G.; Hattermann, K. Liposomal Encapsulated Curcumin Effectively Attenuates Neuroinflammatory and Reactive Astrogliosis Reactions in Glia Cells and Organotypic Brain Slices. Int. J. Nanomed. 2020, 15, 3649–3667. [Google Scholar] [CrossRef]
  51. Hegde, M.; Girisa, S.; BharathwajChetty, B.; Vishwa, R.; Kunnumakkara, A.B. Curcumin Formulations for Better Bioavailability: What We Learned from Clinical Trials Thus Far? ACS Omega 2023, 8, 10713–10746. [Google Scholar] [CrossRef]
  52. Akanda, M.; Mithu, M.S.H.; Douroumis, D. Solid Lipid Nanoparticles: An Effective Lipid-Based Technology for Cancer Treatment. J. Drug Deliv. Sci. Technol. 2023, 86, 104709. [Google Scholar] [CrossRef]
  53. Viegas, C.; Patrício, A.B.; Prata, J.M.; Nadhman, A.; Chintamaneni, P.K.; Fonte, P. Solid Lipid Nanoparticles vs. Nanostructured Lipid Carriers: A Comparative Review. Pharmaceutics 2023, 15, 1593. [Google Scholar] [CrossRef]
  54. Subroto, E.; Andoyo, R.; Indiarto, R. Solid Lipid Nanoparticles: Review of the Current Research on Encapsulation and Delivery Systems for Active and Antioxidant Compounds. Antioxidants 2023, 12, 633. [Google Scholar] [CrossRef]
  55. Mall, J.; Naseem, N.; Haider, M.F.; Rahman, M.A.; Khan, S.; Siddiqui, S.N. Nanostructured Lipid Carriers as a Drug Delivery System: A Comprehensive Review with Therapeutic Applications. Intell. Pharm. 2024, in press. [CrossRef]
  56. Panwar, P.; Kumar, S.; Chand, P.; Chauhan, A.S.; Jakhmola, V. Nanostructured Lipid Carriers (NLCs): A Comprehensive Review of Drug Delivery Advancements. J. Appl. Pharm. Res. 2025, 13, 20–38. [Google Scholar] [CrossRef]
  57. Uti, D.E.; Alum, E.U.; Atangwho, I.J.; Ugwu, O.P.-C.; Egbung, G.E.; Aja, P.M. Lipid-Based Nano-Carriers for the Delivery of Anti-Obesity Natural Compounds: Advances in Targeted Delivery and Precision Therapeutics. J. Nanobiotechnology 2025, 23, 336. [Google Scholar] [CrossRef] [PubMed]
  58. Patel, D.; Solanki, J.; Kher, M.M.; Azagury, A. A Review: Surface Engineering of Lipid-Based Drug Delivery Systems. Small 2024, 20, e2401990. [Google Scholar] [CrossRef] [PubMed]
  59. Fatima, N.; Qazi, U.Y.; Mansha, A.; Bhatti, I.A.; Javaid, R.; Abbas, Q.; Nadeem, N.; Rehan, Z.A.; Noreen, S.; Zahid, M. Recent Developments for Antimicrobial Applications of Graphene-Based Polymeric Composites: A Review. J. Ind. Eng. Chem. 2021, 100, 40–58. [Google Scholar] [CrossRef]
  60. Elmowafy, M.; Shalaby, K.; Elkomy, M.H.; Alsaidan, O.A.; Gomaa, H.A.M.; Abdelgawad, M.A.; Mostafa, E.M. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers 2023, 15, 1123. [Google Scholar] [CrossRef]
  61. Bhardwaj, H.; Jangde, R.K. Current Updated Review on Preparation of Polymeric Nanoparticles for Drug Delivery and Biomedical Applications. Next Nanotechnol. 2023, 2, 100013. [Google Scholar] [CrossRef]
  62. Geszke-Moritz, M.; Moritz, M. Biodegradable Polymeric Nanoparticle-Based Drug Delivery Systems: Comprehensive Overview, Perspectives and Challenges. Polymers 2024, 16, 2536. [Google Scholar] [CrossRef]
  63. Gómez-Guillén, M.C.; Montero, M.P. Enhancement of Oral Bioavailability of Natural Compounds and Probiotics by Mucoadhesive Tailored Biopolymer-Based Nanoparticles: A Review. Food Hydrocoll. 2021, 118, 106772. [Google Scholar] [CrossRef]
  64. Martău, G.A.; Mihai, M.; Vodnar, D.C. The Use of Chitosan, Alginate, and Pectin in the Biomedical and Food Sector—Biocompatibility, Bioadhesiveness, and Biodegradability. Polymers 2019, 11, 1837. [Google Scholar] [CrossRef] [PubMed]
  65. Kunde, S.S.; Wairkar, S. Targeted Delivery of Albumin Nanoparticles for Breast Cancer: A Review. Colloids Surf. B Biointerfaces 2022, 213, 112422. [Google Scholar] [CrossRef] [PubMed]
  66. 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. [Google Scholar] [CrossRef]
  67. Farjaminejad, S.; Farjaminejad, R.; Hasani, M.; Garcia-Godoy, F.; Abdouss, M.; Marya, A.; Harsoputranto, A.; Jamilian, A. Advances and Challenges in Polymer-Based Scaffolds for Bone Tissue Engineering: A Path Towards Personalized Regenerative Medicine. Polymers 2024, 16, 3303. [Google Scholar] [CrossRef]
  68. Pawlik, J.; Łukowicz, K.; Cholewa-Kowalska, K.; Osyczka, A.M. New Insights into the PLGA and PCL Blending: Physico-Mechanical Properties and Cell Response. Mater. Res. Express 2019, 6, 085344. [Google Scholar] [CrossRef]
  69. Misir, J.; Kassama, L. Nanoencapsulation of Lycopene by PLGA: Effect on Physicochemical Properties and Extended-Release Kinetics in a Simulated GIT System. LWT 2025, 225, 117747. [Google Scholar] [CrossRef]
  70. Arpagaus, C. PLA/PLGA Nanoparticles Prepared by Nano Spray Drying. J. Pharm. Investig. 2019, 49, 405–426. [Google Scholar] [CrossRef]
  71. Operti, M.C.; Bernhardt, A.; Grimm, S.; Engel, A.; Figdor, C.G.; Tagit, O. PLGA-Based Nanomedicines Manufacturing: Technologies Overview and Challenges in Industrial Scale-Up. Int. J. Pharm. 2021, 605, 120807. [Google Scholar] [CrossRef]
  72. Sivadasan, D.; Sultan, M.H.; Madkhali, O.; Almoshari, Y.; Thangavel, N. Polymeric Lipid Hybrid Nanoparticles (PLNs) as Emerging Drug Delivery Platform—A Comprehensive Review of Their Properties, Preparation Methods, and Therapeutic Applications. Pharmaceutics 2021, 13, 1291. [Google Scholar] [CrossRef]
  73. Gajbhiye, K.R.; Salve, R.; Narwade, M.; Sheikh, A.; Kesharwani, P.; Gajbhiye, V. Lipid Polymer Hybrid Nanoparticles: A Custom-Tailored Next-Generation Approach for Cancer Therapeutics. Mol. Cancer 2023, 22, 160. [Google Scholar] [CrossRef] [PubMed]
  74. Alsehli, M. Polymeric Nanocarriers as Stimuli-Responsive Systems for Targeted Tumor (Cancer) Therapy: Recent Advances in Drug Delivery. Saudi Pharm. J. 2020, 28, 255–265. [Google Scholar] [CrossRef] [PubMed]
  75. Das, S.S.; Bharadwaj, P.; Bilal, M.; Barani, M.; Rahdar, A.; Taboada, P.; Bungau, S.; Kyzas, G.Z. Stimuli-Responsive Polymeric Nanocarriers for Drug Delivery, Imaging, and Theragnosis. Polymers 2020, 12, 1397. [Google Scholar] [CrossRef] [PubMed]
  76. Nadaf, S.J.; Jadhav, N.R.; Naikwadi, H.S.; Savekar, P.L.; Sapkal, I.D.; Kambli, M.M.; Desai, I.A. Green Synthesis of Gold and Silver Nanoparticles: Updates on Research, Patents, and Future Prospects. OpenNano 2022, 8, 100076. [Google Scholar] [CrossRef]
  77. Serdar, G.; Gül Kılınç, G.; Mazlum Şen, T. Green One-Pot Synthesis of Silver and Gold Nanoparticles Using Catechin Extracts: Influence of Temperature and Antioxidant Activity Evaluation. Plasmonics 2025, 1–20. [Google Scholar] [CrossRef]
  78. Sadalage, P.S.; Patil, R.V.; Havaldar, D.V.; Gavade, S.S.; Santos, A.C.; Pawar, K.D. Optimally Biosynthesized, PEGylated Gold Nanoparticles Functionalized with Quercetin and Camptothecin Enhance Potential Anti-Inflammatory, Anti-Cancer and Anti-Angiogenic Activities. J. Nanobiotechnology 2021, 19, 84. [Google Scholar] [CrossRef]
  79. More, P.R.; Pandit, S.; De Filippis, A.; Franci, G.; Mijakovic, I.; Galdiero, M. Silver Nanoparticles: Bactericidal and Mechanistic Approach against Drug Resistant Pathogens. Microorganisms 2023, 11, 369. [Google Scholar] [CrossRef]
  80. Rodrigues, A.S.; Batista, J.G.S.; Rodrigues, M.Á.V.; Thipe, V.C.; Minarini, L.A.R.; Lopes, P.S.; Lugão, A.B. Advances in Silver Nanoparticles: A Comprehensive Review on Their Potential as Antimicrobial Agents and Their Mechanisms of Action Elucidated by Proteomics. Front. Microbiol. 2024, 15, 1440065. [Google Scholar] [CrossRef]
  81. Perera, W.P.T.D.; Dissanayake, R.K.; Ranatunga, U.I.; Hettiarachchi, N.M.; Perera, K.D.C.; Unagolla, J.M.; De Silva, R.T.; Pahalagedara, L.R. Curcumin Loaded Zinc Oxide Nanoparticles for Activity-Enhanced Antibacterial and Anticancer Applications. RSC Adv. 2020, 10, 30785–30795. [Google Scholar] [CrossRef]
  82. Turrina, C.; Schoenen, M.; Milani, D.; Klassen, A.; Rojas Gonzaléz, D.M.; Cvirn, G.; Mela, P.; Berensmeier, S.; Slabu, I.; Schwaminger, S.P. Application of Magnetic Iron Oxide Nanoparticles: Thrombotic Activity, Imaging and Cytocompatibility of Silica-Coated and Carboxymethyl Dextrane-Coated Particles. Colloids Surf. B Biointerfaces 2023, 228, 113428. [Google Scholar] [CrossRef]
  83. Aulifa, D.L.; Amarilis, B.; Ichsani, L.N.; Maharani, D.S.; Shabrina, A.M.; Hanifah, H.; Wulandari, R.P.; Rusdin, A.; Subra, L.; Budiman, A. A Comprehensive Review: Mesoporous Silica Nanoparticles Greatly Improve Pharmacological Effectiveness of Phytoconstituent in Plant Extracts. Pharmaceuticals 2024, 17, 1684. [Google Scholar] [CrossRef]
  84. Razavi, S.; Janfaza, S.; Tasnim, N.; Gibson, D.L.; Hoorfar, M. Nanomaterial-Based Encapsulation for Controlled Gastrointestinal Delivery of Viable Probiotic Bacteria. Nanoscale Adv. 2021, 3, 2699–2709. [Google Scholar] [CrossRef] [PubMed]
  85. Shi, Z.; Zhou, Y.; Fan, T.; Lin, Y.; Zhang, H.; Mei, L. Inorganic Nano-Carriers Based Smart Drug Delivery Systems for Tumor Therapy. Smart Mater. Med. 2020, 1, 32–47. [Google Scholar] [CrossRef]
  86. Unnikrishnan, G.; Joy, A.; Megha, M.; Kolanthai, E.; Senthilkumar, M. Exploration of Inorganic Nanoparticles for Revolutionary Drug Delivery Applications: A Critical Review. Discov. Nano 2023, 18, 157. [Google Scholar] [CrossRef]
  87. Dave, V.; Tak, K.; Sohgaura, A.; Gupta, A.; Sadhu, V.; Reddy, K.R. Lipid-Polymer Hybrid Nanoparticles: Synthesis Strategies and Biomedical Applications. J. Microbiol. Methods 2019, 160, 130–142. [Google Scholar] [CrossRef] [PubMed]
  88. Shi, Y.; Yu, Q.; Tan, L.; Wang, Q.; Zhu, W. Tumor Microenvironment-Responsive Polymer Delivery Platforms for Cancer Therapy. Angew. Chem. Int. Ed. Engl. 2025, 64, e202503776. [Google Scholar] [CrossRef] [PubMed]
  89. Chang, D.; Ma, Y.; Xu, X.; Xie, J.; Ju, S. Stimuli-Responsive Polymeric Nanoplatforms for Cancer Therapy. Front. Bioeng. Biotechnol. 2021, 9, 707319. [Google Scholar] [CrossRef]
  90. Gajbhiye, K.R.; Pawar, A.; Mahadik, K.R.; Gajbhiye, V. PEGylated Nanocarriers: A Promising Tool for Targeted Delivery to the Brain. Colloids Surf. B Biointerfaces 2020, 187, 110770. [Google Scholar] [CrossRef]
  91. Sadeghzadeh, F.; Motavalizadehkakhky, A.; Mehrzad, J.; Zhiani, R.; Homayouni Tabrizi, M. Folic Acid Conjugated-Chitosan Modified Nanostructured Lipid Carriers as Promising Carriers for Delivery of Umbelliprenin to Cancer Cells: In Vivo and In Vitro. Eur. Polym. J. 2023, 186, 111849. [Google Scholar] [CrossRef]
  92. Kim, S.M.; Patel, M.; Patel, R. PLGA Core-Shell Nano/Microparticle Delivery System for Biomedical Application. Polymers 2021, 13, 3471. [Google Scholar] [CrossRef]
  93. Idoudi, S.; Ismail, R.; Rachid, O.; Elhissi, A.; Alkilany, A.M. The Golden Liposomes: Preparation and Biomedical Applications of Gold-Liposome Nanocomposites. J. Nanotheranostics 2023, 4, 201–227. [Google Scholar] [CrossRef]
  94. Koga, K.; Tagami, T.; Ozeki, T. Gold Nanoparticle-Coated Thermosensitive Liposomes for the Triggered Release of Doxorubicin, and Photothermal Therapy Using a near-Infrared Laser. Colloids Surf. A Physicochem. Eng. Asp. 2021, 626, 127038. [Google Scholar] [CrossRef]
  95. Luiz, M.T.; Dutra, J.A.P.; Viegas, J.S.R.; de Araújo, J.T.C.; Tavares Junior, A.G.; Chorilli, M. Hybrid Magnetic Lipid-Based Nanoparticles for Cancer Therapy. Pharmaceutics 2023, 15, 751. [Google Scholar] [CrossRef] [PubMed]
  96. Duda-Madej, A.; Viscardi, S.; Szewczyk, W.; Topola, E. Natural Alkaloids in Cancer Therapy: Berberine, Sanguinarine and Chelerythrine against Colorectal and Gastric Cancer. Int. J. Mol. Sci. 2024, 25, 8375. [Google Scholar] [CrossRef]
  97. Isaković, S.; Senćanski, M.; Perović, V.; Stevanović, K.; Prodić, I. Bioinformatic Selection of Mannose-Specific Lectins from Allium Genus as SARS-CoV-2 Inhibitors Analysing Protein–Protein Interaction. Life 2025, 15, 162. [Google Scholar] [CrossRef]
  98. Nieto, C.; Vega, M.A.; Martín del Valle, E. Nature-Inspired Nanoparticles as Paclitaxel Targeted Carrier for the Treatment of HER2-Positive Breast Cancer. Cancers 2021, 13, 2526. [Google Scholar] [CrossRef]
  99. Chen, B.-Q.; Zhao, Y.; Zhang, Y.; Pan, Y.-J.; Xia, H.-Y.; Kankala, R.K.; Wang, S.-B.; Liu, G.; Chen, A.-Z. Immune-Regulating Camouflaged Nanoplatforms: A Promising Strategy to Improve Cancer Nano-Immunotherapy. Bioact. Mater. 2023, 21, 1–19. [Google Scholar] [CrossRef]
  100. Vadevoo, S.M.P.; Gurung, S.; Lee, H.-S.; Gunassekaran, G.R.; Lee, S.-M.; Yoon, J.-W.; Lee, Y.-K.; Lee, B. Peptides as Multifunctional Players in Cancer Therapy. Exp. Mol. Med. 2023, 55, 1099–1109. [Google Scholar] [CrossRef]
  101. Rejman, J.; Oberle, V.; Zuhorn, I.S.; Hoekstra, D. Size-Dependent Internalization of Particles via the Pathways of Clathrin- and Caveolae-Mediated Endocytosis. Biochem. J. 2004, 377, 159–169. [Google Scholar] [CrossRef]
  102. Wang, J.; Ding, Y.; Chong, K.; Cui, M.; Cao, Z.; Tang, C.; Tian, Z.; Hu, Y.; Zhao, Y.; Jiang, S. Recent Advances in Lipid Nanoparticles and Their Safety Concerns for MRNA Delivery. Vaccines 2024, 12, 1148. [Google Scholar] [CrossRef]
  103. Shirsat, S.D.; Londhe, P.V.; Gaikwad, A.P.; Rizwan, M.; Laha, S.S.; Khot, V.M.; Achal, V.; Tabish, T.A.; Thorat, N.D. Endosomal Escape in Magnetic Nanostructures: Recent Advances and Future Perspectives. Mater. Today Adv. 2024, 22, 100484. [Google Scholar] [CrossRef]
  104. Ni, J.; Tian, F.; Dahmani, F.Z.; Yang, H.; Yue, D.; He, S.; Zhou, J.; Yao, J. Curcumin-Carboxymethyl Chitosan (CNC) Conjugate and CNC/LHR Mixed Polymeric Micelles as New Approaches to Improve the Oral Absorption of P-Gp Substrate Drugs. Drug Deliv. 2016, 23, 3424–3435. [Google Scholar] [CrossRef]
  105. Majrashi, T.A.; Alshehri, S.A.; Alsayari, A.; Bin Muhsinah, A.; Alrouji, M.; Alshahrani, A.M.; Shamsi, A.; Atiya, A. Insight into the Biological Roles and Mechanisms of Phytochemicals in Different Types of Cancer: Targeting Cancer Therapeutics. Nutrients 2023, 15, 1704. [Google Scholar] [CrossRef]
  106. Karnwal, A.; Jassim, A.Y.; Mohammed, A.A.; Sharma, V.; Al-Tawaha, A.R.M.S.; Sivanesan, I. Nanotechnology for Healthcare: Plant-Derived Nanoparticles in Disease Treatment and Regenerative Medicine. Pharmaceuticals 2024, 17, 1711. [Google Scholar] [CrossRef] [PubMed]
  107. Unnikrishnan Meenakshi, D.; Narde, G.K.; Ahuja, A.; Al Balushi, K.; Francis, A.P.; Khan, S.A. Therapeutic Applications of Nanoformulated Resveratrol and Quercetin Phytochemicals in Colorectal Cancer—An Updated Review. Pharmaceutics 2024, 16, 761. [Google Scholar] [CrossRef] [PubMed]
  108. Cano, A.; Ettcheto, M.; Chang, J.-H.; Barroso, E.; Espina, M.; Kühne, B.A.; Barenys, M.; Auladell, C.; Folch, J.; Souto, E.B.; et al. Dual-Drug Loaded Nanoparticles of Epigallocatechin-3-Gallate (EGCG)/Ascorbic Acid Enhance Therapeutic Efficacy of EGCG in a APPswe/PS1dE9 Alzheimer’s Disease Mice Model. J. Control. Release 2019, 301, 62–75. [Google Scholar] [CrossRef] [PubMed]
  109. Thakur, S.; Sharma, R.; Sharma, S.; Mittal, A.; Deodhar, N. Drug–Polymer Conjugate Tailoring by Disulfide Linkage for Controlled and Targeted Drug Delivery. In Polymer-Drug Conjugates; Elsevier: Amsterdam, The Netherlands, 2023; pp. 171–192. [Google Scholar]
  110. Solanki, R.; Jodha, B.; Prabina, K.E.; Aggarwal, N.; Patel, S. Recent Advances in Phytochemical Based Nano-Drug Delivery Systems to Combat Breast Cancer: A Review. J. Drug Deliv. Sci. Technol. 2022, 77, 103832. [Google Scholar] [CrossRef]
  111. Chowdhury, S.; Kumari, G.; Jit, T. Nano-Formulation of Phytochemicals to Combat Neurodegenerative Diseases: Current and Thorough Strategies. Pharmacol. Res.—Nat. Prod. 2025, 7, 100249. [Google Scholar] [CrossRef]
  112. Cherian, E.; Rosamma; Anju, K.A.; Naushad, B. Nanotechnology in Energy Supplements: Sustained Endurance. In Nanofuel: The Future of Sports Nutrition; Springer: Singapore, 2025; pp. 201–208. [Google Scholar]
  113. Sun, S.; Lv, W.; Li, S.; Zhang, Q.; He, W.; Min, Z.; Teng, C.; Chen, Y.; Liu, L.; Yin, J.; et al. Smart Liposomal Nanocarrier Enhanced the Treatment of Ischemic Stroke through Neutrophil Extracellular Traps and Cyclic Guanosine Monophosphate-Adenosine Monophosphate Synthase-Stimulator of Interferon Genes (CGAS-STING) Pathway Inhibition of Ischemic Pen. ACS Nano 2023, 17, 17845–17857. [Google Scholar] [CrossRef]
  114. Trofin, A.-M.; Scripcariu, D.V.; Filipiuc, S.-I.; Neagu, A.-N.; Filipiuc, L.-E.; Tamba, B.-I.; Palaghia, M.M.; Uritu, C.M. From Nature to Nanomedicine: Enhancing the Antitumor Efficacy of Rhein, Curcumin, and Resveratrol. Medicina 2025, 61, 981. [Google Scholar] [CrossRef]
  115. Cavalcante de Freitas, P.G.; Rodrigues Arruda, B.; Araújo Mendes, M.G.; Barroso de Freitas, J.V.; da Silva, M.E.; Sampaio, T.L.; Petrilli, R.; Eloy, J.O. Resveratrol-Loaded Polymeric Nanoparticles: The Effects of D-α-Tocopheryl Polyethylene Glycol 1000 Succinate (TPGS) on Physicochemical and Biological Properties against Breast Cancer In Vitro and In Vivo. Cancers 2023, 15, 2802. [Google Scholar] [CrossRef] [PubMed]
  116. Yin, H.; Guo, Q.; Li, X.; Tang, T.; Li, C.; Wang, H.; Sun, Y.; Feng, Q.; Ma, C.; Gao, C.; et al. Curcumin Suppresses IL-1β Secretion and Prevents Inflammation through Inhibition of the NLRP3 Inflammasome. J. Immunol. 2018, 200, 2835–2846. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, B.; Hu, S.; Teng, Y.; Chen, J.; Wang, H.; Xu, Y.; Wang, K.; Xu, J.; Cheng, Y.; Gao, X. Current Advance of Nanotechnology in Diagnosis and Treatment for Malignant Tumors. Signal Transduct. Target. Ther. 2024, 9, 200. [Google Scholar] [CrossRef] [PubMed]
  118. Afzal, O.; Rizwanullah, M.; Altamimi, A.S.A.; Alossaimi, M.A.; Kamal, M.; Ahmad, J. Harnessing Natural Polysaccharides-Based Nanoparticles for Oral Delivery of Phytochemicals: Knocking down the Barriers. J. Drug Deliv. Sci. Technol. 2023, 82, 104368. [Google Scholar] [CrossRef]
  119. Zhou, Y.; Chen, D.; Xue, G.; Yu, S.; Yuan, C.; Huang, M.; Jiang, L. Improved Therapeutic Efficacy of Quercetin-Loaded Polymeric Nanoparticles on Triple-Negative Breast Cancer by Inhibiting UPA. RSC Adv. 2020, 10, 34517–34526. [Google Scholar] [CrossRef]
  120. Cunha, C.; Daniel-da-Silva, A.L.; Oliveira, H. Drug Delivery Systems and Flavonoids: Current Knowledge in Melanoma Treatment and Future Perspectives. Micromachines 2022, 13, 1838. [Google Scholar] [CrossRef]
  121. Tu, J.; Xu, Y.; Xu, J.; Ling, Y.; Cai, Y. Chitosan Nanoparticles Reduce LPS-Induced Inflammatory Reaction via Inhibition of NF-ΚB Pathway in Caco-2 Cells. Int. J. Biol. Macromol. 2016, 86, 848–856. [Google Scholar] [CrossRef]
  122. Singh Patel, P.; Srivastava, R.; Panchawat, S. Role of Apoptotic-Targeted Phytoconstitutent-Loaded Antipsoriatic Nanobiocomposites. Recent Pat. Nanotechnol. 2024, 18, 220–236. [Google Scholar] [CrossRef]
  123. Hedayati, N.; Safari, M.H.; Milasi, Y.E.; Kahkesh, S.; Farahani, N.; Khoshnazar, S.M.; Dorostgou, Z.; Alaei, E.; Alimohammadi, M.; Rahimzadeh, P.; et al. Modulation of the PI3K/Akt Signaling Pathway by Resveratrol in Cancer: Molecular Mechanisms and Therapeutic Opportunity. Discov. Oncol. 2025, 16, 669. [Google Scholar] [CrossRef]
  124. Nurkolis, F.; Taslim, N.A.; Syahputra, R.A.; d’Arqom, A.; Tjandrawinata, R.R.; Purba, A.K.R.; Mustika, A. Food Phytochemicals as Epigenetic Modulators in Diabetes: A Systematic Review. J. Agric. Food Res. 2025, 21, 101873. [Google Scholar] [CrossRef]
  125. Wang, G.; Wang, J.; Momeni, M.R. Epigallocatechin-3-Gallate and Its Nanoformulation in Cervical Cancer Therapy: The Role of Genes, MicroRNA and DNA Methylation Patterns. Cancer Cell Int. 2023, 23, 335. [Google Scholar] [CrossRef] [PubMed]
  126. Minafra, L.; Porcino, N.; Bravatà, V.; Gaglio, D.; Bonanomi, M.; Amore, E.; Cammarata, F.P.; Russo, G.; Militello, C.; Savoca, G.; et al. Radiosensitizing Effect of Curcumin-Loaded Lipid Nanoparticles in Breast Cancer Cells. Sci. Rep. 2019, 9, 11134. [Google Scholar] [CrossRef] [PubMed]
  127. Hu, Y.; Wang, Z.; Qiu, Y.; Liu, Y.; Ding, M.; Zhang, Y. Anti-MiRNA21 and Resveratrol-Loaded Polysaccharide-Based Mesoporous Silica Nanoparticle for Synergistic Activity in Gastric Carcinoma. J. Drug Target. 2019, 27, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
  128. Di Pompo, G.; Cortini, M.; Palomba, R.; Di Francesco, V.; Bellotti, E.; Decuzzi, P.; Baldini, N.; Avnet, S. Curcumin-Loaded Nanoparticles Impair the Pro-Tumor Activity of Acid-Stressed MSC in an In Vitro Model of Osteosarcoma. Int. J. Mol. Sci. 2021, 22, 5760. [Google Scholar] [CrossRef]
  129. Roy, S.; Deka, D.; Kondaveeti, S.B.; Ayyadurai, P.; Siripragada, S.; Philip, N.; Pathak, S.; Duttaroy, A.K.; Banerjee, A. An Overview of Potential of Natural Compounds to Regulate Epigenetic Modifications in Colorectal Cancer: A Recent Update. Epigenetics 2025, 20, 2491316. [Google Scholar] [CrossRef]
  130. Alven, S.; Aderibigbe, B.A. Efficacy of Polymer-Based Nanocarriers for Co-Delivery of Curcumin and Selected Anticancer Drugs. Nanomaterials 2020, 10, 1556. [Google Scholar] [CrossRef]
  131. Caon, T.; Mazzarino, L.; Simões, C.M.O.; Senna, E.L.; Silva, M.A.S. Lipid- and Polymer-Based Nanostructures for Cutaneous Delivery of Curcumin. AAPS PharmSciTech 2017, 18, 920–925. [Google Scholar] [CrossRef]
  132. Wongrakpanich, A.; Bui Thi Thu, H.; Sakchaisri, K.; Taresco, V.; Crucitti, V.C.; Bunsupa, S.; Suksiriworapong, J. Co-Delivery of Curcumin and Resveratrol by Folic Acid-Conjugated Poly(Glycerol Adipate) Nanoparticles for Enhanced Synergistic Anticancer Effect against Osteosarcoma. J. Drug Deliv. Sci. Technol. 2024, 95, 105610. [Google Scholar] [CrossRef]
  133. Sitia, L.; Sevieri, M.; Signati, L.; Bonizzi, A.; Chesi, A.; Mainini, F.; Corsi, F.; Mazzucchelli, S. HER-2-Targeted Nanoparticles for Breast Cancer Diagnosis and Treatment. Cancers 2022, 14, 2424. [Google Scholar] [CrossRef]
  134. Zhu, Z.; Wu, X.; Zhang, J.; Zhu, M.; Tian, M.; Zhao, P. Advances in Understanding Ferroptosis Mechanisms and Their Impact on Immune Cell Regulation and Tumour Immunotherapy. Discov. Oncol. 2025, 16, 153. [Google Scholar] [CrossRef]
  135. Koklesova, L.; Jakubikova, J.; Cholujova, D.; Samec, M.; Mazurakova, A.; Šudomová, M.; Pec, M.; Hassan, S.T.S.; Biringer, K.; Büsselberg, D.; et al. Phytochemical-Based Nanodrugs Going beyond the State-of-the-Art in Cancer Management—Targeting Cancer Stem Cells in the Framework of Predictive, Preventive, Personalized Medicine. Front. Pharmacol. 2023, 14, 1121950. [Google Scholar] [CrossRef]
  136. Gaur, S.; Stein, E.B.; Schneider, D.K.; Masotti, M.; Davenport, M.S.; George, A.K.; Ellis, J.H. Gold Nanoshells for Prostate Cancer Treatment: Evidence for Deposition in Abdominal Organs. Abdom. Radiol. 2024, 49, 1929–1939. [Google Scholar] [CrossRef] [PubMed]
  137. Jia, W.; Zhou, L.; Li, L.; Zhou, P.; Shen, Z. Nano-Based Drug Delivery of Polyphenolic Compounds for Cancer Treatment: Progress, Opportunities, and Challenges. Pharmaceuticals 2023, 16, 101. [Google Scholar] [CrossRef] [PubMed]
  138. Tie, S.; Tan, M. Current Advances in Multifunctional Nanocarriers Based on Marine Polysaccharides for Colon Delivery of Food Polyphenols. J. Agric. Food Chem. 2022, 70, 903–915. [Google Scholar] [CrossRef]
  139. Rezaei, B.; Yari, P.; Sanders, S.M.; Wang, H.; Chugh, V.K.; Liang, S.; Mostufa, S.; Xu, K.; Wang, J.; Gómez-Pastora, J.; et al. Magnetic Nanoparticles: A Review on Synthesis, Characterization, Functionalization, and Biomedical Applications. Small 2024, 20, e2304848. [Google Scholar] [CrossRef] [PubMed]
  140. Bhia, M.; Motallebi, M.; Abadi, B.; Zarepour, A.; Pereira-Silva, M.; Saremnejad, F.; Santos, A.C.; Zarrabi, A.; Melero, A.; Jafari, S.M.; et al. Naringenin Nano-Delivery Systems and Their Therapeutic Applications. Pharmaceutics 2021, 13, 291. [Google Scholar] [CrossRef]
  141. Javid-Naderi, M.J.; Abbasi, Z.; Fathi-karkan, S.; Shahgolzari, M.; Maleki-baladi, R.; Shayegh, F.; Ebrahimzadeh, A.; Banimohamad-Shotorbani, B.; Rahdar, A.; Babaei, M.; et al. Advancements in Nanocarrier-Mediated Sunitinib Delivery: Addressing Obstacles and Revealing Its Therapeutic Promise in Oncological Treatment. J. Drug Deliv. Sci. Technol. 2024, 100, 106107. [Google Scholar] [CrossRef]
  142. Farasati Far, B.; Naimi-Jamal, M.R.; Sedaghat, M.; Hoseini, A.; Mohammadi, N.; Bodaghi, M. Combinational System of Lipid-Based Nanocarriers and Biodegradable Polymers for Wound Healing: An Updated Review. J. Funct. Biomater. 2023, 14, 115. [Google Scholar] [CrossRef]
  143. Teixeira, M.I.; Lopes, C.M.; Amaral, M.H.; Costa, P.C. Surface-Modified Lipid Nanocarriers for Crossing the Blood-Brain Barrier (BBB): A Current Overview of Active Targeting in Brain Diseases. Colloids Surf. B Biointerfaces 2023, 221, 112999. [Google Scholar] [CrossRef]
  144. Yavarpour-Bali, H.; Ghasemi-Kasman, M.; Pirzadeh, M. Curcumin-Loaded Nanoparticles: A Novel Therapeutic Strategy in Treatment of Central Nervous System Disorders. Int. J. Nanomed. 2019, 14, 4449–4460. [Google Scholar] [CrossRef]
  145. Payne, A.; Nahashon, S.; Taka, E.; Adinew, G.M.; Soliman, K.F.A. Epigallocatechin-3-Gallate (EGCG): New Therapeutic Perspectives for Neuroprotection, Aging, and Neuroinflammation for the Modern Age. Biomolecules 2022, 12, 371. [Google Scholar] [CrossRef]
  146. Patel, V.; Chavda, V.; Shah, J. Nanotherapeutics in Neuropathologies: Obstacles, Challenges and Recent Advancements in CNS Targeted Drug Delivery Systems. Curr. Neuropharmacol. 2021, 19, 693–710. [Google Scholar] [CrossRef]
  147. Detsi, A.; Kavetsou, E.; Kostopoulou, I.; Pitterou, I.; Pontillo, A.R.N.; Tzani, A.; Christodoulou, P.; Siliachli, A.; Zoumpoulakis, P. Nanosystems for the Encapsulation of Natural Products: The Case of Chitosan Biopolymer as a Matrix. Pharmaceutics 2020, 12, 669. [Google Scholar] [CrossRef]
  148. Behzadifar, S.; Barras, A.; Plaisance, V.; Pawlowski, V.; Szunerits, S.; Abderrahmani, A.; Boukherroub, R. Polymer-Based Nanostructures for Pancreatic Beta-Cell Imaging and Non-Invasive Treatment of Diabetes. Pharmaceutics 2023, 15, 1215. [Google Scholar] [CrossRef] [PubMed]
  149. Thiruvengadam, R.; Kondapavuluri, B.K.; Thangavelu, L.; Thiruvengadam, M.; Hatami, M.; Kim, J.H. Nanoparticle-Based Strategies with Bioactive Compounds for Targeting Oxidative Stress in Therapeutic Interventions: A Comprehensive Review. Ind. Crops Prod. 2025, 227, 120804. [Google Scholar] [CrossRef]
  150. Lacatusu, I.; Badea, N.; Udeanu, D.; Coc, L.; Pop, A.; Cioates Negut, C.; Tanase, C.; Stan, R.; Meghea, A. Improved Anti-Obesity Effect of Herbal Active and Endogenous Lipids Co-Loaded Lipid Nanocarriers: Preparation, In Vitro and In Vivo Evaluation. Mater. Sci. Eng. C 2019, 99, 12–24. [Google Scholar] [CrossRef] [PubMed]
  151. Padmanaban, S.; Baek, J.-W.; Chamarthy, S.S.; Chandrasekaran, S.; Samrot, A.V.; Gosu, V.; Park, I.-K.; Radhakrishnan, K.; Kim, D.-K. Nanoparticle-Based Therapeutic Strategies for Chronic Liver Diseases: Advances and Insights. Liver Res. 2025, 9, 104–117. [Google Scholar] [CrossRef] [PubMed]
  152. Chehelgerdi, M.; Chehelgerdi, M.; Allela, O.Q.B.; Pecho, R.D.C.; Jayasankar, N.; Rao, D.P.; Thamaraikani, T.; Vasanthan, M.; Viktor, P.; Lakshmaiya, N.; et al. Progressing Nanotechnology to Improve Targeted Cancer Treatment: Overcoming Hurdles in Its Clinical Implementation. Mol. Cancer 2023, 22, 169. [Google Scholar] [CrossRef]
  153. Sharma, G.; Song, L.F.; Merz, K.M. Effect of an Inhibitor on the ACE2-Receptor-Binding Domain of SARS-CoV-2. J. Chem. Inf. Model. 2022, 62, 6574–6585. [Google Scholar] [CrossRef]
  154. Hajibonabi, A.; Yekani, M.; Sharifi, S.; Nahad, J.S.; Dizaj, S.M.; Memar, M.Y. Antimicrobial Activity of Nanoformulations of Carvacrol and Thymol: New Trend and Applications. OpenNano 2023, 13, 100170. [Google Scholar] [CrossRef]
  155. Granata, G.; Stracquadanio, S.; Leonardi, M.; Napoli, E.; Consoli, G.M.L.; Cafiso, V.; Stefani, S.; Geraci, C. Essential Oils Encapsulated in Polymer-Based Nanocapsules as Potential Candidates for Application in Food Preservation. Food Chem. 2018, 269, 286–292. [Google Scholar] [CrossRef] [PubMed]
  156. Singh, L.S. Developing Nanocarrier-Based Formulations of Antidiabetic Drugs Derived from Medicinal Plants: A Systemic Review. Pharmacol. Res. Nat. Prod. 2024, 2, 100004. [Google Scholar] [CrossRef]
  157. Dube, E.; Okuthe, G.E. Silver Nanoparticle-Based Antimicrobial Coatings: Sustainable Strategies for Microbial Contamination Control. Microbiol. Res. 2025, 16, 110. [Google Scholar] [CrossRef]
  158. Hallan, S.S.; Ferrara, F.; Cortesi, R.; Sguizzato, M. Potential of the Nano-Encapsulation of Antioxidant Molecules in Wound Healing Applications: An Innovative Strategy to Enhance the Bio-Profile. Molecules 2025, 30, 641. [Google Scholar] [CrossRef]
  159. Pattnaik, S.; Mohanty, S.; Sahoo, S.K.; Mohanty, C. A Mechanistic Perspective on the Role of Phytoconstituents-Based Pharmacotherapeutics and Their Topical Formulations in Chronic Wound Management. J. Drug Deliv. Sci. Technol. 2023, 84, 104546. [Google Scholar] [CrossRef]
  160. Spleis, H.; Sandmeier, M.; Claus, V.; Bernkop-Schnürch, A. Surface Design of Nanocarriers: Key to More Efficient Oral Drug Delivery Systems. Adv. Colloid Interface Sci. 2023, 313, 102848. [Google Scholar] [CrossRef]
  161. Su, Y.; Zhang, B.; Sun, R.; Liu, W.; Zhu, Q.; Zhang, X.; Wang, R.; Chen, C. PLGA-Based Biodegradable Microspheres in Drug Delivery: Recent Advances in Research and Application. Drug Deliv. 2021, 28, 1397–1418. [Google Scholar] [CrossRef]
  162. Pareek, A.; Kapoor, D.U.; Yadav, S.K.; Rashid, S.; Fareed, M.; Akhter, M.S.; Muteeb, G.; Gupta, M.M.; Prajapati, B.G. Advancing Lipid Nanoparticles: A Pioneering Technology in Cosmetic and Dermatological Treatments. Colloid Interface Sci. Commun. 2025, 64, 100814. [Google Scholar] [CrossRef]
  163. Li, X.; Peng, X.; Zoulikha, M.; Boafo, G.F.; Magar, K.T.; Ju, Y.; He, W. Multifunctional Nanoparticle-Mediated Combining Therapy for Human Diseases. Signal Transduct. Target. Ther. 2024, 9, 1. [Google Scholar] [CrossRef]
  164. 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, 2400280. [Google Scholar] [CrossRef]
  165. Formica, M.L.; Real, D.A.; Picchio, M.L.; Catlin, E.; Donnelly, R.F.; Paredes, A.J. On a Highway to the Brain: A Review on Nose-to-Brain Drug Delivery Using Nanoparticles. Appl. Mater. Today 2022, 29, 101631. [Google Scholar] [CrossRef]
  166. Cheng, X.; Han, X.; Si, J.; Dong, C.; Ji, Z.; Zhao, S.; Wu, X.; Li, H.; Jin, X. Cationic Curcumin Nanocrystals Liposomes for Improved Oral Bioavailability: Formulation Development, Optimization, In Vitro and In Vivo Evaluation. Pharmaceutics 2024, 16, 1155. [Google Scholar] [CrossRef] [PubMed]
  167. Bertoncini-Silva, C.; Vlad, A.; Ricciarelli, R.; Giacomo Fassini, P.; Suen, V.M.M.; Zingg, J.-M. Enhancing the Bioavailability and Bioactivity of Curcumin for Disease Prevention and Treatment. Antioxidants 2024, 13, 331. [Google Scholar] [CrossRef] [PubMed]
  168. Zhu, M.; Whittaker, A.K.; Han, F.Y.; Smith, M.T. Journey to the Market: The Evolution of Biodegradable Drug Delivery Systems. Appl. Sci. 2022, 12, 935. [Google Scholar] [CrossRef]
  169. Zandieh, M.A.; Farahani, M.H.; Daryab, M.; Motahari, A.; Gholami, S.; Salmani, F.; Karimi, F.; Samaei, S.S.; Rezaee, A.; Rahmanian, P.; et al. Stimuli-Responsive (Nano)Architectures for Phytochemical Delivery in Cancer Therapy. Biomed. Pharmacother. 2023, 166, 115283. [Google Scholar] [CrossRef]
  170. Mi, P. Stimuli-Responsive Nanocarriers for Drug Delivery, Tumor Imaging, Therapy and Theranostics. Theranostics 2020, 10, 4557–4588. [Google Scholar] [CrossRef]
  171. Lu, S.; Zhang, C.; Wang, J.; Zhao, L.; Li, G. Research Progress in Nano-Drug Delivery Systems Based on the Characteristics of the Liver Cancer Microenvironment. Biomed. Pharmacother. 2024, 170, 116059. [Google Scholar] [CrossRef]
  172. Shi, Y.; Li, X.; Li, Z.; Sun, J.; Gao, T.; Wei, G.; Guo, Q. Nano-Formulations in Disease Therapy: Designs, Advances, Challenges, and Future Directions. J. Nanobiotechnology 2025, 23, 396. [Google Scholar] [CrossRef]
  173. Vagena, I.-A.; Malapani, C.; Gatou, M.-A.; Lagopati, N.; Pavlatou, E.A. Enhancement of EPR Effect for Passive Tumor Targeting: Current Status and Future Perspectives. Appl. Sci. 2025, 15, 3189. [Google Scholar] [CrossRef]
  174. Shi, D.; Beasock, D.; Fessler, A.; Szebeni, J.; Ljubimova, J.Y.; Afonin, K.A.; Dobrovolskaia, M.A. To PEGylate or Not to PEGylate: Immunological Properties of Nanomedicine’s Most Popular Component, Polyethylene Glycol and Its Alternatives. Adv. Drug Deliv. Rev. 2022, 180, 114079. [Google Scholar] [CrossRef]
  175. Shi, L.; Zhang, J.; Zhao, M.; Tang, S.; Cheng, X.; Zhang, W.; Li, W.; Liu, X.; Peng, H.; Wang, Q. Effects of Polyethylene Glycol on the Surface of Nanoparticles for Targeted Drug Delivery. Nanoscale 2021, 13, 10748–10764. [Google Scholar] [CrossRef]
  176. Shakori Poshteh, S.; Alipour, S.; Varamini, P. Harnessing Curcumin and Nanotechnology for Enhanced Treatment of Breast Cancer Bone Metastasis. Discov. Nano 2024, 19, 177. [Google Scholar] [CrossRef] [PubMed]
  177. Madamsetty, V.S.; Pal, K.; Keshavan, S.; Caulfield, T.R.; Dutta, S.K.; Wang, E.; Fadeel, B.; Mukhopadhyay, D. Development of Multi-Drug Loaded PEGylated Nanodiamonds to Inhibit Tumor Growth and Metastasis in Genetically Engineered Mouse Models of Pancreatic Cancer. Nanoscale 2019, 11, 22006–22018. [Google Scholar] [CrossRef] [PubMed]
  178. Ahmadi, M.; Ritter, C.A.; von Woedtke, T.; Bekeschus, S.; Wende, K. Package Delivered: Folate Receptor-Mediated Transporters in Cancer Therapy and Diagnosis. Chem. Sci. 2024, 15, 1966–2006. [Google Scholar] [CrossRef] [PubMed]
  179. Perumal, V.; Ravula, A.R.; Agas, A.; Kannan, M.; Liu, X.; I, S.S.; Vijayaraghavalu, S.; Haorah, J.; Zhang, Y.; Chandra, N. Transferrin-Grafted Albumin Nanoparticles for the Targeted Delivery of Apocynin and Neuroprotection in an In Vitro Model of the BBB. Micro 2023, 3, 84–106. [Google Scholar] [CrossRef]
  180. Ow, V.; Lin, Q.; Wong, J.H.M.; Sim, B.; Tan, Y.L.; Leow, Y.; Goh, R.; Loh, X.J. Understanding the Interplay between PH and Charges for Theranostic Nanomaterials. Nanoscale 2025, 17, 6960–6980. [Google Scholar] [CrossRef]
  181. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  182. Rarokar, N.; Yadav, S.; Saoji, S.; Bramhe, P.; Agade, R.; Gurav, S.; Khedekar, P.; Subramaniyan, V.; Wong, L.S.; Kumarasamy, V. Magnetic Nanosystem a Tool for Targeted Delivery and Diagnostic Application: Current Challenges and Recent Advancement. Int. J. Pharm. X 2024, 7, 100231. [Google Scholar] [CrossRef]
  183. Wang, W.; Huang, Z.; Huang, Y.; Zhang, X.; Huang, J.; Cui, Y.; Yue, X.; Ma, C.; Fu, F.; Wang, W.; et al. Pulmonary Delivery Nanomedicines towards Circumventing Physiological Barriers: Strategies and Characterization Approaches. Adv. Drug Deliv. Rev. 2022, 185, 114309. [Google Scholar] [CrossRef]
  184. Chow, J.C.L. Nanomaterial-Based Molecular Imaging in Cancer: Advances in Simulation and AI Integration. Biomolecules 2025, 15, 444. [Google Scholar] [CrossRef]
  185. Kumar, M.; Kulkarni, P.; Liu, S.; Chemuturi, N.; Shah, D.K. Nanoparticle Biodistribution Coefficients: A Quantitative Approach for Understanding the Tissue Distribution of Nanoparticles. Adv. Drug Deliv. Rev. 2023, 194, 114708. [Google Scholar] [CrossRef] [PubMed]
  186. Sarcan, E.T. Nanoparticles for Dual Imaging: Pet and Fluorescence Imaging. Ank. Univ. Eczac. Fak. Derg. 2024, 48, 658–671. [Google Scholar] [CrossRef]
  187. Skotland, T.; Iversen, T.G.; Llorente, A.; Sandvig, K. Biodistribution, Pharmacokinetics and Excretion Studies of Intravenously Injected Nanoparticles and Extracellular Vesicles: Possibilities and Challenges. Adv. Drug Deliv. Rev. 2022, 186, 114326. [Google Scholar] [CrossRef] [PubMed]
  188. Kurul, F.; Turkmen, H.; Cetin, A.E.; Topkaya, S.N. Nanomedicine: How Nanomaterials Are Transforming Drug Delivery, Bio-Imaging, and Diagnosis. Next Nanotechnol. 2025, 7, 100129. [Google Scholar] [CrossRef]
  189. Sairam, A.B.; Sanmugam, A.; Pushparaj, A.; Mahesh Kumar, G.; Sundarapandian, N.; Balaji, S.; Nallal, M.; Park, K.H. Toxicity of Polymeric Nanodrugs as Drug Carriers. ACS Chem. Health Saf. 2023, 30, 236–250. [Google Scholar] [CrossRef]
  190. Herdiana, Y.; Wathoni, N.; Shamsuddin, S.; Joni, I.M.; Muchtaridi, M. Chitosan-Based Nanoparticles of Targeted Drug Delivery System in Breast Cancer Treatment. Polymers 2021, 13, 1717. [Google Scholar] [CrossRef]
  191. Sukubo, N.G.; Bigini, P.; Morelli, A. Nanocarriers and Macrophage Interaction: From a Potential Hurdle to an Alternative Therapeutic Strategy. Beilstein J. Nanotechnol. 2025, 16, 97–118. [Google Scholar] [CrossRef]
  192. Gutiérrez-Cruz, S.G.; Muñoz-Diosdado, A.; Gutiérrez-Calleja, R.A.; Rodríguez-Cortés, O.; Ortiz-Reyez, A.E.; Flores-Mejía, R. Influence of Physicochemical Factors on the Interaction of Metallic Nanoparticles with Immune System Cells. Front. Nanotechnol. 2025, 6, 1496230. [Google Scholar] [CrossRef]
  193. Makharadze, D.; del Valle, L.J.; Katsarava, R.; Puiggalí, J. The Art of PEGylation: From Simple Polymer to Sophisticated Drug Delivery System. Int. J. Mol. Sci. 2025, 26, 3102. [Google Scholar] [CrossRef]
  194. Miao, G.; He, Y.; Lai, K.; Zhao, Y.; He, P.; Tan, G.; Wang, X. Accelerated Blood Clearance of PEGylated Nanoparticles Induced by PEG-Based Pharmaceutical Excipients. J. Control. Release 2023, 363, 12–26. [Google Scholar] [CrossRef]
  195. Yang, L.; Lin, X.; Zhou, J.; Hou, S.; Fang, Y.; Bi, X.; Yang, L.; Li, L.; Fan, Y. Cell Membrane-Biomimetic Coating via Click-Mediated Liposome Fusion for Mitigating the Foreign-Body Reaction. Biomaterials 2021, 271, 120768. [Google Scholar] [CrossRef]
  196. de la Harpe, K.; Kondiah, P.; Choonara, Y.; Marimuthu, T.; du Toit, L.; Pillay, V. The Hemocompatibility of Nanoparticles: A Review of Cell–Nanoparticle Interactions and Hemostasis. Cells 2019, 8, 1209. [Google Scholar] [CrossRef]
  197. Mehrizi, T.Z.; Kafiabad, S.A.; Eshghi, P. Effects and Treatment Applications of Polymeric Nanoparticles on Improving Platelets’ Storage Time: A Review of the Literature from 2010 to 2020. Blood Res. 2021, 56, 215–228. [Google Scholar] [CrossRef] [PubMed]
  198. Pizzoli, G.; Gargaro, M.; Drava, G.; Voliani, V. Inorganic Nanomaterials Meet the Immune System: An Intricate Balance. Adv. Healthc. Mater. 2025, 14, e2404795. [Google Scholar] [CrossRef] [PubMed]
  199. Sharma, N.; Saifi, M.A.; Singh, S.B.; Godugu, C. In Vivo Studies: Toxicity and Biodistribution of Nanocarriers in Organisms. In Nanotoxicity; Elsevier: Amsterdam, The Netherlands, 2020; pp. 41–70. [Google Scholar]
  200. Available online: https://www.iso.org/standard/68936.html (accessed on 29 June 2025).
  201. Ramos, T.I.; Villacis-Aguirre, C.A.; López-Aguilar, K.V.; Santiago Padilla, L.; Altamirano, C.; Toledo, J.R.; Santiago Vispo, N. The Hitchhiker’s Guide to Human Therapeutic Nanoparticle Development. Pharmaceutics 2022, 14, 247. [Google Scholar] [CrossRef] [PubMed]
  202. Guo, D.; Ji, X.; Luo, J. Rational Nanocarrier Design towards Clinical Translation of Cancer Nanotherapy. Biomed. Mater. 2021, 16, 032005. [Google Scholar] [CrossRef]
  203. Ahmad, J.; Ameeduzzafar; Ahmad, M.Z.; Akhter, H. Surface-Engineered Cancer Nanomedicine: Rational Design and Recent Progress. Curr. Pharm. Des. 2020, 26, 1181–1190. [Google Scholar] [CrossRef]
  204. Rodríguez-Gómez, F.D.; Monferrer, D.; Penon, O.; Rivera-Gil, P. Regulatory Pathways and Guidelines for Nanotechnology-Enabled Health Products: A Comparative Review of EU and US Frameworks. Front. Med. 2025, 12, 1544393. [Google Scholar] [CrossRef]
  205. Kumar, A.; P, N.; Kumar, M.; Jose, A.; Tomer, V.; Oz, E.; Proestos, C.; Zeng, M.; Elobeid, T.; K, S.; et al. Major Phytochemicals: Recent Advances in Health Benefits and Extraction Method. Molecules 2023, 28, 887. [Google Scholar] [CrossRef]
  206. DA, B. Exploring Diverse Techniques for Phytochemical Extraction from Plant Sources a Comprehensive Review. Int. J. Pharmacogn. Chin. Med. 2024, 8, 1–11. [Google Scholar] [CrossRef]
  207. Havelikar, U.; Ghorpade, K.B.; Kumar, A.; Patel, A.; Singh, M.; Banjare, N.; Gupta, P.N. Comprehensive Insights into Mechanism of Nanotoxicity, Assessment Methods and Regulatory Challenges of Nanomedicines. Discov. Nano 2024, 19, 165. [Google Scholar] [CrossRef]
  208. Chen, X.; Li, H.; Zhang, B.; Deng, Z. The Synergistic and Antagonistic Antioxidant Interactions of Dietary Phytochemical Combinations. Crit. Rev. Food Sci. Nutr. 2022, 62, 5658–5677. [Google Scholar] [CrossRef]
  209. Powell, L.G.; Gillies, S.; Fernandes, T.F.; Murphy, F.; Giubilato, E.; Cazzagon, V.; Hristozov, D.; Pizzol, L.; Blosi, M.; Costa, A.L.; et al. Developing Integrated Approaches for Testing and Assessment (IATAs) in Order to Support Nanomaterial Safety. Nanotoxicology 2022, 16, 484–499. [Google Scholar] [CrossRef]
  210. Hu, L.; Luo, Y.; Yang, J.; Cheng, C. Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability. Molecules 2025, 30, 1184. [Google Scholar] [CrossRef] [PubMed]
  211. Petcov, T.E.; Silberschmidt, V.V.; Pandele, M.A.; Chiticaru, E.A.; Ioniță, M.; Manole, M. Nanostructures: An Efficient Drug Delivery Platform for Therapy of Multiple Myeloma. Eur. J. Med. Chem. Rep. 2025, 14, 100263. [Google Scholar] [CrossRef]
  212. Ma, X.; Tian, Y.; Yang, R.; Wang, H.; Allahou, L.W.; Chang, J.; Williams, G.; Knowles, J.C.; Poma, A. Nanotechnology in Healthcare, and Its Safety and Environmental Risks. J. Nanobiotechnology 2024, 22, 715. [Google Scholar] [CrossRef] [PubMed]
  213. Wu, C.; Lee, S.-L.; Taylor, C.; Li, J.; Chan, Y.-M.; Agarwal, R.; Temple, R.; Throckmorton, D.; Tyner, K. Scientific and Regulatory Approach to Botanical Drug Development: A U.S. FDA Perspective. J. Nat. Prod. 2020, 83, 552–562. [Google Scholar] [CrossRef]
  214. Takechi-Haraya, Y.; Ohgita, T.; Demizu, Y.; Saito, H.; Izutsu, K.; Sakai-Kato, K. Current Status and Challenges of Analytical Methods for Evaluation of Size and Surface Modification of Nanoparticle-Based Drug Formulations. AAPS PharmSciTech 2022, 23, 150. [Google Scholar] [CrossRef]
  215. Caputo, F.; Favre, G.; Borchard, G.; Calzolai, L.; Fisicaro, P.; Frejafon, E.; Günday-Türeli, N.; Koltsov, D.; Minelli, C.; Nelson, B.C.; et al. Toward an International Standardisation Roadmap for Nanomedicine. Drug Deliv. Transl. Res. 2024, 14, 2578–2588. [Google Scholar] [CrossRef]
  216. Ali, F.; Neha, K.; Parveen, S. Current Regulatory Landscape of Nanomaterials and Nanomedicines: A Global Perspective. J. Drug Deliv. Sci. Technol. 2023, 80, 104118. [Google Scholar] [CrossRef]
  217. Thomas, J.; Kumar, V.; Sharma, N.; John, N.; Umesh, M.; Kumar Dasarahally Huligowda, L.; Kaur, K.; Utreja, D. Recent Approaches in Nanotoxicity Assessment for Drug Delivery Applications: Challenges and Prospects. Med. Drug Discov. 2025, 25, 100204. [Google Scholar] [CrossRef]
  218. Abdel-Mageed, H.M.; AbuelEzz, N.Z.; Ali, A.A.; Abdelaziz, A.E.; Nada, D.; Abdelraouf, S.M.; Fouad, S.A.; Bishr, A.; Radwan, R.A. Newly Designed Curcumin-Loaded Hybrid Nanoparticles: A Multifunctional Strategy for Combating Oxidative Stress, Inflammation, and Infections to Accelerate Wound Healing and Tissue Regeneration. BMC Biotechnol. 2025, 25, 49. [Google Scholar] [CrossRef] [PubMed]
  219. Burns, J.; Buck, A.C.; D’ Souza, S.; Dube, A.; Bardien, S. Nanophytomedicines as Therapeutic Agents for Parkinson’s Disease. ACS Omega 2023, 8, 42045–42061. [Google Scholar] [CrossRef] [PubMed]
  220. Li, P.; Liu, S.; Zhuang, H.; Niu, M.; Pan, F.; Wang, N.; Sha, S.; Wang, Q.; Wang, J. LC–MS/MS Quantification of 20(S)-Protopanaxadiol in Complex Biological Matrices for Bioanalytical Method Validation and Pharmacokinetic Analysis. Sci. Rep. 2025, 15, 16640. [Google Scholar] [CrossRef]
  221. Dannhorn, A.; Kazanc, E.; Hamm, G.; Swales, J.G.; Strittmatter, N.; Maglennon, G.; Goodwin, R.J.A.; Takats, Z. Correlating Mass Spectrometry Imaging and Liquid Chromatography-Tandem Mass Spectrometry for Tissue-Based Pharmacokinetic Studies. Metabolites 2022, 12, 261. [Google Scholar] [CrossRef]
  222. Bimonte, S.; Barbieri, A.; Leongito, M.; Piccirillo, M.; Giudice, A.; Pivonello, C.; De Angelis, C.; Granata, V.; Palaia, R.; Izzo, F. Curcumin AntiCancer Studies in Pancreatic Cancer. Nutrients 2016, 8, 433. [Google Scholar] [CrossRef]
  223. Conte, R.; Valentino, A.; Sepe, F.; Gianfreda, F.; Condò, R.; Cerroni, L.; Calarco, A.; Peluso, G. Resveratrol-Loaded Solid Lipid Nanoparticles Reinforced Hyaluronic Hydrogel: Multitarget Strategy for the Treatment of Diabetes-Related Periodontitis. Biomedicines 2025, 13, 1059. [Google Scholar] [CrossRef]
  224. Zhang, X.; Chan, H.W.; Shao, Z.; Wang, Q.; Chow, S.; Chow, S.F. Navigating Translational Research in Nanomedicine: A Strategic Guide to Formulation and Manufacturing. Int. J. Pharm. 2025, 671, 125202. [Google Scholar] [CrossRef]
  225. Singh, G.; Thakur, N.; Kumar, R. Nanoparticles in Drinking Water: Assessing Health Risks and Regulatory Challenges. Sci. Total Environ. 2024, 949, 174940. [Google Scholar] [CrossRef]
  226. Saji, C.V.; Mohanakumar, K.P.; Shenoi, R.A. Phytomedicine Nanoformulations for Parkinson’s Disease. Phytomedicine Plus 2025, 5, 100704. [Google Scholar] [CrossRef]
  227. Chavda, V.P.; Nalla, L.V.; Balar, P.; Bezbaruah, R.; Apostolopoulos, V.; Singla, R.K.; Khadela, A.; Vora, L.; Uversky, V.N. Advanced Phytochemical-Based Nanocarrier Systems for the Treatment of Breast Cancer. Cancers 2023, 15, 1023. [Google Scholar] [CrossRef]
  228. Anas, M.; Falak, A.; Khan, A.; Khattak, W.A.; Nisa, S.G.; Aslam, Q.; Khan, K.A.; Saleem, M.H.; Fahad, S. Therapeutic Potential and Agricultural Benefits of Curcumin: A Comprehensive Review of Health and Sustainability Applications. J. Umm Al-Qura Univ. Appl. Sci. 2024, 1–16. [Google Scholar] [CrossRef]
  229. Majumder, J.; Minko, T. Multifunctional and Stimuli-Responsive Nanocarriers for Targeted Therapeutic Delivery. Expert Opin. Drug Deliv. 2021, 18, 205–227. [Google Scholar] [CrossRef] [PubMed]
  230. Pandey, P.; Dureja, H. Recent Patents on Polymeric Nanoparticles for Cancer Therapy. Recent Pat. Nanotechnol. 2018, 12, 155–169. [Google Scholar] [CrossRef] [PubMed]
  231. Mehan, S.; Arora, N.; Bhalla, S.; Khan, A.; Rehman, M.U.; Alghamdi, B.S.; Al Zughaibi, T.; Ashraf, G.M. Involvement of Phytochemical-Encapsulated Nanoparticles’ Interaction with Cellular Signalling in the Amelioration of Benign and Malignant Brain Tumours. Molecules 2022, 27, 3561. [Google Scholar] [CrossRef] [PubMed]
  232. Benderski, K.; Lammers, T.; Sofias, A.M. Analysis of Multi-Drug Cancer Nanomedicine. Nat. Nanotechnol. 2025. Online ahead of print. [Google Scholar] [CrossRef]
  233. Awlqadr, F.H.; Majeed, K.R.; Altemimi, A.B.; Hassan, A.M.; Qadir, S.A.; Saeed, M.N.; Faraj, A.M.; Salih, T.H.; Abd Al-Manhel, A.J.; Najm, M.A.A.; et al. Nanotechnology-Based Herbal Medicine: Preparation, Synthesis, and Applications in Food and Medicine. J. Agric. Food Res. 2025, 19, 101661. [Google Scholar] [CrossRef]
  234. Datta, D.; Priyanka Bandi, S.; Colaco, V.; Dhas, N.; Siva Reddy, D.; Vora, L.K. Fostering the Unleashing Potential of Nanocarriers-Mediated Delivery of Ocular Therapeutics. Int. J. Pharm. 2024, 658, 124192. [Google Scholar] [CrossRef]
  235. Pérez-Pérez, V.; Jiménez-Martínez, C.; González-Escobar, J.L.; Corzo-Ríos, L.J. Exploring the Impact of Encapsulation on the Stability and Bioactivity of Peptides Extracted from Botanical Sources: Trends and Opportunities. Front. Chem. 2024, 12, 1423500. [Google Scholar] [CrossRef]
  236. Nunziata, G.; Borroni, A.; Rossi, F. Advanced Microfluidic Strategies for Core-Shell Nanoparticles: The Next-Generation of Polymeric and Lipid-Based Drug Nanocarriers. Chem. Eng. J. Adv. 2025, 22, 100759. [Google Scholar] [CrossRef]
  237. Bezelya, A.; Küçüktürkmen, B.; Bozkır, A. Microfluidic Devices for Precision Nanoparticle Production. Micro 2023, 3, 822–866. [Google Scholar] [CrossRef]
  238. Das, A.; Prajapati, P. Navigating Pharmaceuticals: Microfluidic Devices in Analytical and Formulation Sciences. Discov. Chem. 2025, 2, 49. [Google Scholar] [CrossRef]
  239. Nogueira, S.S.; Samaridou, E.; Simon, J.; Frank, S.; Beck-Broichsitter, M.; Mehta, A. Analytical Techniques for the Characterization of Nanoparticles for MRNA Delivery. Eur. J. Pharm. Biopharm. 2024, 198, 114235. [Google Scholar] [CrossRef] [PubMed]
  240. Gerzon, G.; Sheng, Y.; Kirkitadze, M. Process Analytical Technologies—Advances in Bioprocess Integration and Future Perspectives. J. Pharm. Biomed. Anal. 2022, 207, 114379. [Google Scholar] [CrossRef] [PubMed]
  241. Ogbuagu, O.O.; Mbata, A.O.; Balogun, O.D.; Oladapo, O.; Ojo, O.O.; Muonde, M. Quality Assurance in Pharmaceutical Manufacturing: Bridging the Gap between Regulations, Supply Chain, and Innovations. Int. J. Multidiscip. Res. Growth Eval. 2023, 4, 823–831. [Google Scholar] [CrossRef]
  242. Carroccio, S.C.; Scarfato, P.; Bruno, E.; Aprea, P.; Dintcheva, N.T.; Filippone, G. Impact of Nanoparticles on the Environmental Sustainability of Polymer Nanocomposites Based on Bioplastics or Recycled Plastics—A Review of Life-Cycle Assessment Studies. J. Clean. Prod. 2022, 335, 130322. [Google Scholar] [CrossRef]
  243. Elayaperumal, S.; Sivamani, Y.; Agarwal, P.; Srivastava, N. Plant-Based Nanotherapeutics: A New Frontier in Disease Management and Prevention. Nano TransMed 2025, 4, 100086. [Google Scholar] [CrossRef]
  244. Haleem, A.; Javaid, M.; Singh, R.P.; Rab, S.; Suman, R. Applications of Nanotechnology in Medical Field: A Brief Review. Glob. Health J. 2023, 7, 70–77. [Google Scholar] [CrossRef]
  245. Khanna, N.N.; Maindarkar, M.A.; Viswanathan, V.; Fernandes, J.F.E.; Paul, S.; Bhagawati, M.; Ahluwalia, P.; Ruzsa, Z.; Sharma, A.; Kolluri, R.; et al. Economics of Artificial Intelligence in Healthcare: Diagnosis vs. Treatment. Healthcare 2022, 10, 2493. [Google Scholar] [CrossRef]
  246. Dutta, N.; Dhar, D. Understanding Medical Technology Innovation in Low- and Middle-Income Countries: Factors, Impact, and a Model Proposal. She Ji J. Des. Econ. Innov. 2024, 10, 192–222. [Google Scholar] [CrossRef]
  247. Cheng, T.-M.; Chu, H.-Y.; Huang, H.-M.; Li, Z.-L.; Chen, C.-Y.; Shih, Y.-J.; Whang-Peng, J.; Cheng, R.H.; Mo, J.-K.; Lin, H.-Y.; et al. Toxicologic Concerns with Current Medical Nanoparticles. Int. J. Mol. Sci. 2022, 23, 7597. [Google Scholar] [CrossRef]
  248. Damasco, J.A.; Ravi, S.; Perez, J.D.; Hagaman, D.E.; Melancon, M.P. Understanding Nanoparticle Toxicity to Direct a Safe-by-Design Approach in Cancer Nanomedicine. Nanomaterials 2020, 10, 2186. [Google Scholar] [CrossRef]
  249. Shajar, F.; Saleem, S.; Mushtaq, N.U.; Shah, W.H.; Rasool, A.; Padder, S.A.; Tahir, I.; Rehman, R.U. Regulatory and Ethical Issues Raised by the Utilization of Nanomaterials. In Interaction of Nanomaterials with Living Cells; Springer Nature: Singapore, 2023; pp. 899–924. [Google Scholar]
  250. Gupta, A.P.; Pathak, A.; Pandey, P. Challenges and Future of Nanotechnology in Global Herbal Medicine Practices. In Herbal Medicine Phytochemistry; Springer: Cham, Switzerland, 2024; pp. 1627–1653. [Google Scholar]
  251. Tian, M.; Xin, X.; Wu, R.; Guan, W.; Zhou, W. Advances in Intelligent-Responsive Nanocarriers for Cancer Therapy. Pharmacol. Res. 2022, 178, 106184. [Google Scholar] [CrossRef]
  252. Sabit, H.; Pawlik, T.M.; Radwan, F.; Abdel-Hakeem, M.; Abdel-Ghany, S.; Wadan, A.-H.S.; Elzawahri, M.; El-Hashash, A.; Arneth, B. Precision Nanomedicine: Navigating the Tumor Microenvironment for Enhanced Cancer Immunotherapy and Targeted Drug Delivery. Mol. Cancer 2025, 24, 160. [Google Scholar] [CrossRef]
  253. Palanikumar, L.; Al-Hosani, S.; Kalmouni, M.; Nguyen, V.P.; Ali, L.; Pasricha, R.; Barrera, F.N.; Magzoub, M. PH-Responsive High Stability Polymeric Nanoparticles for Targeted Delivery of Anticancer Therapeutics. Commun. Biol. 2020, 3, 95. [Google Scholar] [CrossRef]
  254. Meng, X.; Shen, Y.; Zhao, H.; Lu, X.; Wang, Z.; Zhao, Y. Redox-Manipulating Nanocarriers for Anticancer Drug Delivery: A Systematic Review. J. Nanobiotechnology 2024, 22, 587. [Google Scholar] [CrossRef] [PubMed]
  255. Graham, W.; Torbett-Dougherty, M.; Islam, A.; Soleimani, S.; Bruce-Tagoe, T.A.; Johnson, J.A. Magnetic Nanoparticles and Drug Delivery Systems for Anti-Cancer Applications: A Review. Nanomaterials 2025, 15, 285. [Google Scholar] [CrossRef] [PubMed]
  256. Sun, H.; Zhang, Q.; Li, J.; Peng, S.; Wang, X.; Cai, R. Near-Infrared Photoactivated Nanomedicines for Photothermal Synergistic Cancer Therapy. Nano Today 2021, 37, 101073. [Google Scholar] [CrossRef]
  257. Li, L.; Jiang, W.; Qiu, S. Nanomaterials in Stimulus-Responsive Drug Delivery Systems Facilitate Precise Therapy for Hematologic Diseases. J. Mater. Chem. B 2025, 13, 7953–7972. [Google Scholar] [CrossRef]
  258. Mousavi-Kiasary, S.M.S.; Senabreh, A.; Zandi, A.; Pena, R.; Cruz, F.; Adibi, A.; Hooshmand, N. Synergistic Cancer Therapies Enhanced by Nanoparticles: Advancing Nanomedicine Through Multimodal Strategies. Pharmaceutics 2025, 17, 682. [Google Scholar] [CrossRef]
  259. Wu, F.; Zhang, Z.; Ma, S.; He, Y.; He, Y.; Ma, L.; Lei, N.; Deng, W.; Wang, F. Microenvironment-Responsive Nanosystems for Ischemic Stroke Therapy. Theranostics 2024, 14, 5571–5595. [Google Scholar] [CrossRef]
  260. Han, H.S.; Koo, S.Y.; Choi, K.Y. Emerging Nanoformulation Strategies for Phytocompounds and Applications from Drug Delivery to Phototherapy to Imaging. Bioact. Mater. 2022, 14, 182–205. [Google Scholar] [CrossRef] [PubMed]
  261. Kopac, T. Leveraging Artificial Intelligence and Machine Learning for Characterizing Protein Corona, Nanobiological Interactions, and Advancing Drug Discovery. Bioengineering 2025, 12, 312. [Google Scholar] [CrossRef] [PubMed]
  262. Kapoor, D.U.; Sharma, J.B.; Gandhi, S.M.; Prajapati, B.G.; Thanawuth, K.; Limmatvapirat, S.; Sriamornsak, P. AI-Driven Design and Optimization of Nanoparticle-Based Drug Delivery Systems. Sci. Eng. Health Stud. 2024, 18, 24010003. [Google Scholar] [CrossRef]
  263. Rezvantalab, S.; Mihandoost, S.; Rezaiee, M. Machine Learning Assisted Exploration of the Influential Parameters on the PLGA Nanoparticles. Sci. Rep. 2024, 14, 1114. [Google Scholar] [CrossRef]
  264. Zeynalov, N.; Taghiyev, D.; Taghiyev, S.; Mikayilov, E. Role of Computational Modeling in the Design and Development of Nanotechnology-Based Drug Delivery Systems. Chem. Biochem. Eng. Q. 2024, 38, 97–110. [Google Scholar] [CrossRef]
  265. Chou, W.-C.; Chen, Q.; Yuan, L.; Cheng, Y.-H.; He, C.; Monteiro-Riviere, N.A.; Riviere, J.E.; Lin, Z. An Artificial Intelligence-Assisted Physiologically-Based Pharmacokinetic Model to Predict Nanoparticle Delivery to Tumors in Mice. J. Control. Release 2023, 361, 53–63. [Google Scholar] [CrossRef]
  266. Samathoti, P.; Kumarachari, R.K.; Bukke, S.P.N.; Rajasekhar, E.S.K.; Jaiswal, A.A.; Eftekhari, Z. The Role of Nanomedicine and Artificial Intelligence in Cancer Health Care: Individual Applications and Emerging Integrations—A Narrative Review. Discov. Oncol. 2025, 16, 697. [Google Scholar] [CrossRef]
  267. Fu, C.; Chen, Q. The Future of Pharmaceuticals: Artificial Intelligence in Drug Discovery and Development. J. Pharm. Anal. 2025, 101248, in press. [Google Scholar] [CrossRef]
  268. Zeng, J.; Jia, X. Systems Theory-Driven Framework for AI Integration into the Holistic Material Basis Research of Traditional Chinese Medicine. Engineering 2024, 40, 28–50. [Google Scholar] [CrossRef]
  269. Mienye, I.D.; Obaido, G.; Jere, N.; Mienye, E.; Aruleba, K.; Emmanuel, I.D.; Ogbuokiri, B. A Survey of Explainable Artificial Intelligence in Healthcare: Concepts, Applications, and Challenges. Inform. Med. Unlocked 2024, 51, 101587. [Google Scholar] [CrossRef]
  270. Serrano, D.R.; Luciano, F.C.; Anaya, B.J.; Ongoren, B.; Kara, A.; Molina, G.; Ramirez, B.I.; Sánchez-Guirales, S.A.; Simon, J.A.; Tomietto, G.; et al. Artificial Intelligence (AI) Applications in Drug Discovery and Drug Delivery: Revolutionizing Personalized Medicine. Pharmaceutics 2024, 16, 1328. [Google Scholar] [CrossRef]
  271. Shrestha, B.; Tang, L.; Hood, R.L. Nanotechnology for Personalized Medicine. In Nanomedicine; Springer: Singapore, 2022; pp. 1–48. [Google Scholar]
  272. Kim, J.H.; Dareowolabi, B.O.; Thiruvengadam, R.; Moon, E.-Y. Application of Nanotechnology and Phytochemicals in Anticancer Therapy. Pharmaceutics 2024, 16, 1169. [Google Scholar] [CrossRef] [PubMed]
  273. Mansoor, S.; Hamid, S.; Tuan, T.T.; Park, J.-E.; Chung, Y.S. Advance Computational Tools for Multiomics Data Learning. Biotechnol. Adv. 2024, 77, 108447. [Google Scholar] [CrossRef] [PubMed]
  274. Chen, Y.; Cai, S.; Liu, F.-Y.; Liu, M. Advancing Oral Cancer Care: Nanomaterial-Driven Diagnostic and Therapeutic Innovations. Cell Biol. Toxicol. 2025, 41, 90. [Google Scholar] [CrossRef] [PubMed]
  275. Hosseini, S.M.; Mohammadnejad, J.; Salamat, S.; Beiram Zadeh, Z.; Tanhaei, M.; Ramakrishna, S. Theranostic Polymeric Nanoparticles as a New Approach in Cancer Therapy and Diagnosis: A Review. Mater. Today Chem. 2023, 29, 101400. [Google Scholar] [CrossRef]
  276. Rahmani Dabbagh, S.; Rezapour Sarabi, M.; Birtek, M.T.; Mustafaoglu, N.; Zhang, Y.S.; Tasoglu, S. 3D Bioprinted Organ-on-chips. Aggregate 2023, 4, e197. [Google Scholar] [CrossRef]
  277. Wu, X.; Shi, W.; Liu, X.; Gu, Z. Recent Advances in 3D-Printing-Based Organ-on-a-Chip. EngMedicine 2024, 1, 100003. [Google Scholar] [CrossRef]
  278. Wainwright, C.L.; Teixeira, M.M.; Adelson, D.L.; Braga, F.C.; Buenz, E.J.; Campana, P.R.V.; David, B.; Glaser, K.B.; Harata-Lee, Y.; Howes, M.-J.R.; et al. Future Directions for the Discovery of Natural Product-Derived Immunomodulating Drugs: An IUPHAR Positional Review. Pharmacol. Res. 2022, 177, 106076. [Google Scholar] [CrossRef]
  279. Jafarzadeh, S.; Oladzadabbasabadi, N.; Dheyab, M.A.; Lalabadi, M.A.; Sheibani, S.; Ghasemlou, M.; Esmaeili, Y.; Barrow, C.J.; Naebe, M.; Timms, W. Emerging Trends in Smart and Sustainable Nano-Biosensing: The Role of Green Nanomaterials. Ind. Crops Prod. 2025, 223, 120108. [Google Scholar] [CrossRef]
  280. Reda, A.T.; Park, Y.T. Sustainable Synthesis of Functional Nanomaterials: Renewable Resources, Energy-Efficient Methods, Environmental Impact and Circular Economy Approaches. Chem. Eng. J. 2025, 516, 163894. [Google Scholar] [CrossRef]
  281. Kazemi, S.; Hosseingholian, A.; Gohari, S.D.; Feirahi, F.; Moammeri, F.; Mesbahian, G.; Moghaddam, Z.S.; Ren, Q. Recent Advances in Green Synthesized Nanoparticles: From Production to Application. Mater. Today Sustain. 2023, 24, 100500. [Google Scholar] [CrossRef]
  282. Singh, P.; Garg, A.; Pandit, S.; Mokkapati, V.R.S.S.; Mijakovic, I. Antimicrobial Effects of Biogenic Nanoparticles. Nanomaterials 2018, 8, 1009. [Google Scholar] [CrossRef] [PubMed]
  283. Ansari, M.; Ahmed, S.; Abbasi, A.; Khan, M.T.; Subhan, M.; Bukhari, N.A.; Hatamleh, A.A.; Abdelsalam, N.R. Plant Mediated Fabrication of Silver Nanoparticles, Process Optimization, and Impact on Tomato Plant. Sci. Rep. 2023, 13, 18048. [Google Scholar] [CrossRef]
  284. Kučuk, N.; Primožič, M.; Knez, Ž.; Leitgeb, M. Sustainable Biodegradable Biopolymer-Based Nanoparticles for Healthcare Applications. Int. J. Mol. Sci. 2023, 24, 3188. [Google Scholar] [CrossRef] [PubMed]
  285. Nizam, N.U.M.; Hanafiah, M.M.; Woon, K.S. A Content Review of Life Cycle Assessment of Nanomaterials: Current Practices, Challenges, and Future Prospects. Nanomaterials 2021, 11, 3324. [Google Scholar] [CrossRef] [PubMed]
  286. Alli, Y.A.; Bamisaye, A.; Bamidele, M.O.; Etafo, N.O.; Chkirida, S.; Lawal, A.; Hammed, V.O.; Akinfenwa, A.S.; Hanson, E.; Nwakile, C.; et al. Transforming Waste to Wealth: Harnessing Carbon Dioxide for Sustainable Solutions. Results Surf. Interfaces 2024, 17, 100321. [Google Scholar] [CrossRef]
  287. Jeffcoat, P.; Di Lernia, C.; Hardy, C.; New, E.J.; Chrzanowski, W. (Re)Imagining Purpose: A Framework for Sustainable Nanotechnology Innovation. NanoImpact 2024, 35, 100511. [Google Scholar] [CrossRef]
  288. Pokrajac, L.; Abbas, A.; Chrzanowski, W.; Dias, G.M.; Eggleton, B.J.; Maguire, S.; Maine, E.; Malloy, T.; Nathwani, J.; Nazar, L.; et al. Nanotechnology for a Sustainable Future: Addressing Global Challenges with the International Network4Sustainable Nanotechnology. ACS Nano 2021, 15, 18608–18623. [Google Scholar] [CrossRef]
  289. Kumar, A.; Tyagi, P.K.; Tyagi, S.; Ghorbanpour, M. Integrating Green Nanotechnology with Sustainable Development Goals: A Pathway to Sustainable Innovation. Discov. Sustain. 2024, 5, 364. [Google Scholar] [CrossRef]
Figure 1. Emergence of nanotechnology in phytomedicine: from plant-derived bioactives to multifunctional nanoformulations.
Figure 1. Emergence of nanotechnology in phytomedicine: from plant-derived bioactives to multifunctional nanoformulations.
Molecules 30 03177 g001
Figure 2. (a) Overview of plant-derived phytochemicals relevant to human health. (b), Chemical structures of major phytochemical classes polyphenols, alkaloids, terpenoids, flavonoids, and glycosides highlighting their therapeutic relevance. These scaffolds form the basis for nanoformulation strategies in modern phytomedicine.
Figure 2. (a) Overview of plant-derived phytochemicals relevant to human health. (b), Chemical structures of major phytochemical classes polyphenols, alkaloids, terpenoids, flavonoids, and glycosides highlighting their therapeutic relevance. These scaffolds form the basis for nanoformulation strategies in modern phytomedicine.
Molecules 30 03177 g002
Figure 3. Comprehensive classification and functional overview of nanocarrier systems for phytochemical delivery.
Figure 3. Comprehensive classification and functional overview of nanocarrier systems for phytochemical delivery.
Molecules 30 03177 g003
Figure 4. Schematic of the molecular mechanisms of action of nano-phytomedicine: cellular uptake, redox modulation, signaling interference, and epigenetic regulation.
Figure 4. Schematic of the molecular mechanisms of action of nano-phytomedicine: cellular uptake, redox modulation, signaling interference, and epigenetic regulation.
Molecules 30 03177 g004
Figure 5. Therapeutic applications of nano-phytomedicine in human health.
Figure 5. Therapeutic applications of nano-phytomedicine in human health.
Molecules 30 03177 g005
Figure 6. Pharmacokinetic and toxicological framework of nano-phytomedicine.
Figure 6. Pharmacokinetic and toxicological framework of nano-phytomedicine.
Molecules 30 03177 g006
Figure 7. Schematic overview of nano-phytomedicine pharmacology highlighting bioavailability, biodistribution, biocompatibility, and regulatory safety considerations.
Figure 7. Schematic overview of nano-phytomedicine pharmacology highlighting bioavailability, biodistribution, biocompatibility, and regulatory safety considerations.
Molecules 30 03177 g007
Table 1. Comparative overview of major phytochemical classes, therapeutic properties, and delivery challenges.
Table 1. Comparative overview of major phytochemical classes, therapeutic properties, and delivery challenges.
Phytochemical ClassExample CompoundsPrimary Therapeutic PropertiesDelivery ChallengesReferences
PolyphenolsCurcumin, Resveratrol, EGCGAntioxidant, anti-inflammatory, anticancerPoor aqueous solubility, rapid metabolism, low absorption[30,32]
AlkaloidsBerberine, Morphine, VincristineAntibacterial, antidiabetic, anticancerNarrow therapeutic index, high toxicity at higher doses[28,37]
TerpenoidsLimonene, Paclitaxel, CarotenoidsAnticancer, antimicrobial, cardioprotectiveOxidative instability, poor bioavailability[29]
FlavonoidsQuercetin, Kaempferol, NaringeninAntioxidant, neuroprotective, anti-inflammatoryLow membrane permeability, rapid phase II metabolism[30,37]
GlycosidesDigoxin, Sennosides, SteviosideCardiotonic, laxative, metabolic regulationLimited stability in GI tract, slow release kinetics[31]
EGCG = Epigallocatechin gallate, GI tract = Gastrointestinal tract.
Table 2. Comparative analysis of nanocarrier systems for phytochemical delivery.
Table 2. Comparative analysis of nanocarrier systems for phytochemical delivery.
Nanocarrier TypeKey FeaturesAdvantages in Phytochemical DeliveryLimitations/ChallengesRepresentative ApplicationsReferences
LiposomesPhospholipid bilayer vesicles with hydrophilic core and lipophilic shellBiocompatibility, can carry both hydrophilic and lipophilic phytochemicals, enhanced absorptionPhysical instability, oxidation of lipids, rapid clearance without PEGylationCurcumin, resveratrol, EGCG for cancer and inflammation[49]
Solid Lipid Nanoparticles (SLNs)Nanocarriers made of solid lipids stabilized by surfactantsHigh loading of lipophilic phytochemicals, protection from degradation, scalable productionLimited drug loading, potential for polymorphic transitions affecting stabilitySilymarin, quercetin for liver protection and metabolic disorders[52,53,54]
Nanostructured Lipid Carriers (NLCs)Mix of solid and liquid lipids to form a less ordered matrixImproved drug loading, prolonged release, better stability than SLNsComplex formulation optimization needed, possibility of lipid phase separationBerberine, naringenin for metabolic and cardiovascular diseases[55,56]
Natural Polymeric NPsPolysaccharide (e.g., chitosan, alginate) or protein-based carriersBiodegradable, mucoadhesive, enhances intestinal uptake and stabilityBatch variability, lower mechanical strength than synthetic carriersChitosan-curcumin NPs for oral anti-inflammatory therapy[63,64]
Synthetic Polymeric NPsPLGA, PCL, PEG-based systems designed via emulsion or nanoprecipitationCustomizable release profile, high encapsulation efficiency, FDA-accepted polymersRequires organic solvents, potential accumulation of degradation productsPLGA-resveratrol or quercetin NPs for cancer and neurodegenerative diseases[67,68]
Inorganic NPs—MetalGold (AuNPs), Silver (AgNPs), synthesized via green or chemical methodsExcellent stability, imaging compatibility, tunable size/shape, antimicrobial synergy with phytochemicalsPotential cytotoxicity, accumulation risk, surface functionalization required for targetingAgNP–berberine conjugates for antimicrobial activity[76,77]
Inorganic NPs—Metal OxideZnO, Fe3O4, TiO2, often used with plant extract functionalizationIntrinsic bioactivity (e.g., ROS generation), magnetic guidance, enhanced dissolutionLong-term toxicity concerns, environmental persistenceZnO–curcumin for wound healing, Fe3O4–EGCG for brain targeting[81]
Inorganic NPs—Silica (MSNs)Mesoporous silica with tunable pore size and surface areaHigh surface loading, pH-sensitive release, good biocompatibilityNon-biodegradable, risk of particle accumulation and chronic exposureMSNs with polyphenols for anticancer or gut delivery[83]
Hybrid NanocarriersLipid-polymer combinations, metal-lipid or polymer-metal hybridsCombines benefits of each system, controlled release, structural stability, multi-functional capabilityComplex synthesis, scalability challengesPLGA–lipid hybrid loaded with curcumin for dual-release in tumors[87]
Stimuli-Responsive SystemspH, redox, enzyme, temperature, or light-triggered release platformsOn-demand release at disease sites, reduced systemic toxicityStimuli-specific tuning required, clinical validation is limitedpH-sensitive quercetin NPs for tumor microenvironments[88,89]
Surface-Functionalized NPsNanoparticles modified with ligands (folate, peptides, antibodies, aptamers) for active targetingReceptor-specific delivery, improved uptake in diseased cells/tissuesLigand synthesis cost, potential immune response, ligand detachment in vivoFolic acid-decorated NLCs for ovarian cancer therapy[90,91]
Table 3. Comparative insights into the molecular mechanisms of action of nano-phytomedicine.
Table 3. Comparative insights into the molecular mechanisms of action of nano-phytomedicine.
MechanismKey FeaturesNano-Phytomedicine RoleAdvantages of NanoformulationRepresentative PhytochemicalsReferences
Cellular Uptake and Intracellular TraffickingInvolves endocytosis (clathrin, caveolin, macropinocytosis), endosomal escape, and organelle targetingEnables cytosolic/nuclear delivery; bypasses degradation; improves uptake in target cellsEnhanced retention, subcellular targeting, P-gp bypass, prolonged releaseCurcumin, resveratrol, quercetin[101,102]
Redox Balance and Antioxidant PathwaysModulation of oxidative stress via Nrf2 pathway, ROS scavenging, and GSH regulationImproves intracellular antioxidant levels, activates protective genes (e.g., HO-1, NQO1)Site-specific ROS scavenging, redox-responsive release, mitochondrial targetingEGCG, curcumin, sulforaphane[106,107,108,109,110]
Apoptosis, Proliferation and InflammationTargets intrinsic/extrinsic apoptotic pathways, NF-κB, and cytokine networksTriggers caspase cascade, downregulates Bcl-2, inhibits COX-2 and TNF-α productionSelective cancer cell apoptosis, reduced systemic toxicity, enhanced anti-inflammatory effectsBerberine, apigenin, quercetin[111]
Signaling Pathway RegulationInterference with PI3K/Akt, MAPK, and NF-κB cascadesInhibits survival/proliferation signals; enhances apoptosis and immune responsesSustained pathway inhibition, synergistic effects with ligands or co-drugsKaempferol, luteolin, curcumin[118],
Epigenetic and Genomic RegulationInvolves DNA methylation, histone acetylation, miRNA modulationModulates DNMTs, HDACs, and epigenetic markers for long-term gene expression controlNuclear delivery, miRNA-targeted therapy, chromatin remodelingResveratrol, sulforaphane, EGCG[124]
P-gp: P-glycoprotein; ROS: Reactive oxygen species; Nrf2: Nuclear factor erythroid 2-related factor 2; DNMTs: DNA methyltransferases; HDACs: Histone deacetylases; GSH: Glutathione; HO-1: Heme oxygenase-1; NQO1: NAD(P)H quinone dehydrogenase 1.
Table 4. Comparative overview of nano-phytomedicine applications in human health.
Table 4. Comparative overview of nano-phytomedicine applications in human health.
Therapeutic AreaMechanism of Action/BenefitNanocarrier StrategiesRepresentative PhytochemicalsReferences
Cancer TherapyTumor targeting via EPR effect, apoptosis induction, ferroptosis, and anti-angiogenesisLiposomes, polymeric nanoparticles, gold nanoshells; HER2, folate-targeted systemsCurcumin, resveratrol, EGCG, paclitaxel precursors[130,131,132,133,134,135,136]
Cardiovascular HealthAnti-atherogenic (↓ LDL oxidation, ↓ VSMC proliferation), antihypertensive (↑ NO release, ↓ vascular resistance), anti-inflammatoryNanoemulsions, lipid-polymer hybrids, magnetic targeting nanoparticlesResveratrol, catechins, naringenin[137,138,139,140,141,142]
Neurodegenerative DisordersBBB penetration, microglial modulation, anti-amyloid aggregation, antioxidant and anti-inflammatory neuroprotectionLiposomes, polymeric nanoparticles with surfactant/ligand coating (e.g., polysorbate-80, transferrin)Curcumin, resveratrol, EGCG[143,144,145,146]
Metabolic DisordersGlycemic control, insulin sensitization, adipogenesis inhibition, lipid modulation, anti-inflammatory and thermogenic effectsChitosan, silver NPs, lipid-polymer hybrids, nanoencapsulation for GI protection and sustained releaseBerberine, quercetin, curcumin, silymarin, catechins[147,148,149,150,151,152]
Infectious DiseasesAntibacterial (biofilm disruption, membrane damage), antiviral (entry blockade, protease inhibition), antifungal (membrane disruption), resistance mitigation (efflux inhibition)Metal–phytochemical NPs (e.g., Ag, ZnO), polymeric nanocapsules, gold conjugates, inhalable/topical delivery formatsCurcumin, baicalin, quercetin, thymol, eugenol[153,154,155,156]
Table 5. Advanced pharmacokinetic and toxicological determinants of nano-phytomedicine performance.
Table 5. Advanced pharmacokinetic and toxicological determinants of nano-phytomedicine performance.
Pharmacological/Toxicological DimensionScientific Insight and Mechanistic RoleNano-Phytomedicine Strategy/AdvantageEmerging Barriers/Optimization NeedsReferences
Kinetic Modulation via Nano-ArchitectureNanoform morphology (e.g., core-shell, hollow, matrix) influences degradation and release rates through interfacial drug diffusion and structural disintegrationPLGA matrix vs. lipid vesicles: predictable vs. burst release kinetics; core–shell hybrids improve tunabilityNano–drug mismatch (e.g., rapid-release from hydrophobic matrices), cross-linking irregularity[156,159,160]
Gastrointestinal Stability and Transit BehaviorNano-encapsulation delays premature phytochemical degradation by gastric enzymes and bile salts, improving residence time and epithelial transportEnteric-coated or mucin-adhering nanoparticles preserve payload integrity and enhance absorptionGI transit variability, impact of digestive enzymes on carrier integrity[159,163,165]
Epithelial Transport and Endocytosis PathwaysNanoform size/charge influences endocytic route: clathrin/caveolae-mediated or macropinocytosis; smaller particles often cross via paracellular or tight junctionsSurface-tuned nanoparticles exploit paracellular pathways (e.g., chitosan opens tight junctions), enhancing transepithelial fluxExcessive opening of junctions may increase toxicity; variability in transporter expression among individuals[163,165,166]
Organ-Specific Accumulation DynamicsOrganotropism is governed by anatomical microenvironment, vascular permeability, and nanoparticle deformability or elasticityFlexible or elongated nanoparticles exhibit greater tumor margination and accumulation; stealth PEG coating extends circulationShape-induced immune clearance, variability in microvascular permeability in disease states[171,173,181]
Mononuclear Phagocyte System (MPS) InteractionsRecognition by Kupffer cells and splenic macrophages remains a major clearance mechanism, influenced by opsonization and protein corona formationPEGylation or biomimetic cloaking (e.g., RBC membrane) reduces MPS uptake and enhances plasma half-lifeAccelerated clearance after repeated dosing (anti-PEG antibodies); need for personalized stealth coatings[174,176,186]
Cytokine and Complement Response MechanismsNanoparticles can activate TLRs or complement system, leading to cytokine storms or hypersensitivity depending on their physicochemical profileUsing neutral/zwitterionic polymers lowers immune activation; shape and surface control mitigate pro-inflammatory signalingRisk of chronic low-grade inflammation; inter-individual differences in immune recognition pathways[191,193,198]
Metabolic Fate and Excretion PathwaysSurface functionalization and hydrodynamic diameter define renal (≤10 nm) vs. hepatobiliary clearance (≥50 nm); metabolism by macrophage enzymes modifies excretion patternsSize-tuned nanoparticles designed for controlled hepatic metabolism or renal filtration; active efflux modulation is possibleRisk of hepatic overloading or kidney retention with chronic use; lack of data on long-term excretion metabolites[169,184,199]
Standardization for Regulatory ApprovalISO 10993 and ICH M3(R2) guidelines emphasize safety, biodegradation, genotoxicity, and immunotoxicity profiling in nanomedicine developmentNanoformulations undergo systematic hemolysis, cytotoxicity, and cytokine profiling prior to preclinical testingAbsence of phytochemical-specific nanotoxicity thresholds; inter-lab variability in compliance methodologies[28]
PLGA: Poly(lactic-co-glycolic acid); PEG: Polyethylene glycol; MPS: Mononuclear Phagocyte System; RBC: Red Blood Cell; ISO: International Organization for Standardization; TLR: Toll-like receptor.
Table 6. Key clinical and translational insights in nano-phytomedicine development.
Table 6. Key clinical and translational insights in nano-phytomedicine development.
Focus AreaScientific HighlightsChallenges and Strategic NeedsReferences
Preclinical and Clinical TrialsDemonstrated enhanced efficacy in murine models (e.g., liposomal curcumin, polymeric resveratrol); improved ADME profiles via nanoencapsulationLimited human trials, complex regulatory pathways, need for predictive preclinical models and adaptive clinical designs[218,219,220,221,222,223,224,225,226]
Commercial Products and Patent LandscapeSeveral liposomal/polymeric products on market as supplements; rising patents in targeted nanocarriers and controlled release technologiesRegulatory heterogeneity between supplements and drugs; IP protection of complex phytochemical systems remains difficult[227,228,229,230,231,232,233]
Scale-Up and Manufacturing BarriersTechniques like high-pressure homogenization and microfluidics can enhance reproducibility; PAT tools support QbD complianceBatch-to-batch variation, costly GMP production, lack of industrial scalability for many prototype nanoformulations[234,235,236,237,238,239,240,241,242,243]
Cost, Accessibility, and Healthcare AcceptanceNanoformulations may reduce dosing and improve outcomes; potential for long-term cost-effectiveness with fewer side effects and improved patient adherenceHigh initial cost, unequal access in low-income regions, lack of awareness and acceptance among healthcare providers and patients[244,245,246,247,248,249,250]
ADME: Absorption, Distribution, Metabolism, and Excretion; QbD: Quality by Design; PAT: Process Analytical Technology; GMP: Good Manufacturing Practice; IP: Intellectual Property.
Table 7. Emerging trends and translational strategies in nano-phytomedicine.
Table 7. Emerging trends and translational strategies in nano-phytomedicine.
Emerging TrendKey FeaturesTranslational AdvantagesReferences
Smart and Responsive Nano-Phytomedicine SystemsStimuli-responsive release triggered by pH, ROS, enzymes, light, or magnetic fields; enables site-specific and on-demand phytochemical delivery.Enhanced spatiotemporal control, reduced off-target toxicity, and improved efficacy in complex disease microenvironments.[251,252,253,254,255,256,257,258,259,260]
Artificial Intelligence and Computational Modeling in Formulation DesignMachine learning and modeling tools used to predict formulation outcomes, optimize nanocarrier parameters, and reduce trial-and-error cycles.Accelerated development timelines, reduced costs, improved reproducibility, and predictive safety/toxicity screening.[261,262,263,264,265,266,267,268,269,270]
Personalized Nano-Phytotherapy and Precision Medicine ApproachesStratified treatments using biomarker profiling, genomics, and pharmacogenomics; theranostics and companion diagnostics enhance personalization.Customized therapies for heterogeneous patient populations, improved adherence, reduced side effects, and dynamic dose optimization.[271,272,273,274,275,276,277,278]
Sustainable and Green Nanotechnology IntegrationGreen synthesis using biogenic routes; biodegradable carriers; aqueous-based processes and LCA-driven design minimize environmental impact.Improved public acceptance, regulatory alignment, reduced ecological burden, and compliance with sustainability mandates.[279,280,281,282,283,284,285,286,287,288,289]
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

Parvin, N.; Aslam, M.; Joo, S.W.; Mandal, T.K. Nano-Phytomedicine: Harnessing Plant-Derived Phytochemicals in Nanocarriers for Targeted Human Health Applications. Molecules 2025, 30, 3177. https://doi.org/10.3390/molecules30153177

AMA Style

Parvin N, Aslam M, Joo SW, Mandal TK. Nano-Phytomedicine: Harnessing Plant-Derived Phytochemicals in Nanocarriers for Targeted Human Health Applications. Molecules. 2025; 30(15):3177. https://doi.org/10.3390/molecules30153177

Chicago/Turabian Style

Parvin, Nargish, Mohammad Aslam, Sang Woo Joo, and Tapas Kumar Mandal. 2025. "Nano-Phytomedicine: Harnessing Plant-Derived Phytochemicals in Nanocarriers for Targeted Human Health Applications" Molecules 30, no. 15: 3177. https://doi.org/10.3390/molecules30153177

APA Style

Parvin, N., Aslam, M., Joo, S. W., & Mandal, T. K. (2025). Nano-Phytomedicine: Harnessing Plant-Derived Phytochemicals in Nanocarriers for Targeted Human Health Applications. Molecules, 30(15), 3177. https://doi.org/10.3390/molecules30153177

Article Metrics

Back to TopTop