Your new experience awaits. Try the new design now and help us make it even better

MINI REVIEW article

Front. Pharmacol., 29 September 2025

Sec. Experimental Pharmacology and Drug Discovery

Volume 16 - 2025 | https://doi.org/10.3389/fphar.2025.1694752

This article is part of the Research TopicNew Drugs and Future Challenges in Drug Metabolism and TransportView all 26 articles

Phytochemical nanoencapsulation and microfluidics drive gene and tumor microenvironment modulation

  • Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador

Phytochemicals are plant-derived bioactive compounds with promising anticancer properties, but their clinical use is limited by poor solubility, instability, rapid metabolism, and restricted tumor penetration. Nanoencapsulation strategies address these barriers by enhancing bioavailability, stability, and tissue-specific delivery, thereby improving therapeutic efficacy and reducing systemic toxicity. This mini-review summarizes recent progress in nanoscale phytochemical delivery systems engineered for gene modulation and tumor microenvironment targeting, including lipid-based, polymeric, hybrid, and biogenic nanocarriers that improve biodistribution and enhance cellular uptake. Notably, the functional performance of nanoscale delivery systems depends on precisely controlled physicochemical characteristics. Consequently, microfluidics has emerged as a powerful tool to fine-tune and fabricate phytochemical-based nanocarriers in a reproducible manner. Beyond fabrication, microfluidic lab-on-a-chip platforms recreate physiological and tumor-specific microenvironments, providing dynamic, real-time assessment of drug transport, metabolism, and tumor–vascular interactions in biomimetic conditions that surpass conventional static models. These innovations expand mechanistic understanding and support more predictive preclinical evaluations. Remaining challenges include variability of natural sources, limited pharmacokinetic and toxicological data, and hurdles in scale-up and standardization. By integrating nanoscale engineering with microfluidic innovation, phytochemical-based nanomedicine is positioned to advance toward more effective, safer, and clinically translatable cancer therapies.

1 Introduction

Phytochemicals, a diverse group of bioactive compounds found in plants, are the subject of renewed interest in biomedical research for their chemopreventive and chemotherapeutic properties (George et al., 2021; Subramaniam et al., 2019). Despite the widespread use of synthetic pharmaceuticals (Campos et al., 2019; Tamatam and Mohammed, 2024), these natural bioactives have long been valued in traditional medicine and are increasingly recognized for their multi-targeted biological mechanisms and generally favorable safety profiles (Hoenders et al., 2024; Wink, 2022; Yang and Ling, 2025). Their anticancer potential has been supported by preclinical studies and, in some cases, by clinical evidence (Choudhari et al., 2019). However, the transition of phytochemicals from bench to bedside remains challenging due to factors such as low water solubility, chemical instability, limited tissue penetration, and rapid metabolism, all of which restrict bioavailability and therapeutic impact in vivo.

To overcome these barriers, advanced formulation strategies have been developed to enhance solubility, protect functional integrity, and achieve controlled, site-specific delivery (Chen et al., 2024) to support clinical translation (Aljabali et al., 2025). Among these, encapsulation strategies for phytochemicals encompass both micro- and nanoscale systems. Microencapsulation techniques, such as spray-drying and freeze-drying, are commonly employed to protect bioactive molecules from environmental degradation and to improve handling, shelf stability, and palatability (Ijod et al., 2024; Bińkowska et al., 2024; Ingale et al., 2025). Some are engineered to respond to physiological cues, such as pH or enzymatic activity, which allow for site-specific release of bioactive compounds or secondary delivery systems that mediate therapeutic effects (Ang et al., 2019; Ma Y. et al., 2021). However, their role in targeted gene or microenvironmental modulation remains limited. In contrast, nanoscale delivery systems, including liposomes, polymeric nanoparticles, and exosome-based carriers, are specifically designed to interact with biological systems, providing control over biodistribution, cellular uptake, and enhanced therapeutic performance (Das et al., 2020). Their functional performance depends on precisely controlled physicochemical characteristics, requiring reproducible, scalable fabrication (Ly et al., 2024; Hui et al., 2025).

Traditional batch-based synthesis often suffers from limitations such as batch-to-batch variability and reduced control over particle uniformity and surface characteristics (Mülhopt et al., 2018). In contrast, microfluidic technologies enable precise, reproducible fabrication of phytochemical nanocarriers by controlling formulation parameters such as flow rate, concentration, and mixing dynamics. This approach produces uniform nanoparticles with defined surface and compositional properties, supporting scalable and translational nanomedicine development (Bezelya et al., 2023; Sebastian, 2022). Beyond synthesis, microfluidic technologies support functional evaluation through biomimetic platforms replicating tissue-specific microenvironments with greater fidelity than conventional 2D or static 3D cultures. These systems offer dynamic control over nutrient delivery, waste removal, and oxygen gradients under flow (Kim et al., 2021; Ayuso et al., 2022). In cancer research, they allow reconstruction of complex tumor architectures, incorporating vasculature, stromal barriers, hypoxic zones, and even microbiota (Ayuso et al., 2022; Farooqi et al., 2023; Zhai et al., 2024). Their efficiency with minimal cell input makes them ideal for patient-derived material, while precise flow control supports real-time assessment of metastasis, drug response, and tumor–vascular niche interactions (Farooqi et al., 2023; Ngo et al., 2023; Zhai et al., 2024).

This minireview focuses on nanoscale phytochemical delivery systems, highlighting microfluidic-based synthesis and functional evaluation (Figure 1), with emphasis on gene modulation and tumor microenvironment targeting.

Figure 1
A circular chart categorizes phytochemicals used in nanoformulations, distinguishing FDA-approved compounds from those in preclinical trials, with images of at least 10 different natural sources from which the phytochemicals are derived, such as grapes and ginger. To the right, nanocarriers such as liposomes and polymeric nanoparticles are displayed, targeting genes and the tumor microenvironment. Below, microfluidic tools like Lab-on-a-chip and Tumor-on-a-chip devices depict modeling of tumor complexity and evaluation of nanoparticle transport, penetration, and efficacy.

Figure 1. Phytochemicals used in oncology, their sources, and their delivery and evaluation systems.

2 Phytochemical-based nanosystems for gene and tumor microenvironment targeting

2.1 Formulation challenges and delivery strategies

While plant-derived drugs such as vincristine, paclitaxel, and docetaxel are established in oncology, they represent only a small fraction of phytochemicals with demonstrated preclinical anticancer activity that have yet to translate into routine clinical use (Figure 1) (Asma et al., 2022; Mazumder et al., 2022; Kim et al., 2024). The majority of plant-derived agents exhibit poor solubility and bioavailability, requiring higher doses that increase toxicity risk. Assumption of inherent safety is common (Jităreanu et al., 2022; Mugale et al., 2024), yet many plant-based compounds, especially alkaloids, saponins, and anthraquinones, can cause organ-specific toxicity and genotoxicity, may alter drug metabolism, and exacerbate adverse effects (Quan et al., 2020; Rao et al., 2022; Brewer and Chen, 2017). Consequently, establishing no-observed-adverse-effect levels (NOAEL) (Dorato and Engelhardt, 2005) through standardized toxicokinetic studies is essential for clinical advancement (Al-Naqeb et al., 2024). These challenges have driven the development of nanoscale delivery strategies that enhance pharmacokinetics and formulation versatility by solubilizing poorly water-soluble compounds, protecting them from enzymatic degradation, extending circulation time, and ultimately improving bioavailability while reducing toxicity (Dhupal and Chowdhury, 2020; Bilia et al., 2019).

Liposomes remain a cornerstone in phytochemical nanoformulation due to their biocompatibility and amphiphilic nature. Liposomal formulations of resveratrol and epigallocatechin gallate improved bioavailability and reduced systemic toxicity in colorectal and liver cancer models (Wilson et al., 2023). Other lipid-based systems (Jacob et al., 2025), such as solid lipid nanoparticles and nanostructured lipid carriers, enhance absorption and plasma retention of betulinic acid and andrographolide in various tumor models (Wang et al., 2024; Li H. et al., 2022). Phytosomes (Talebi et al., 2025), formed by complexing phytochemicals with phospholipids, improve membrane permeability and metabolic stability, particularly benefiting poorly absorbed polyphenols, silymarin, and catechin (Babazadeh et al., 2018). In contrast to phytosomes, polymeric and hybrid nanocarriers offer versatility for structurally diverse phytochemicals. Systems based in poly(lactic-co-glycolic acid) (PLGA) and hybrid carriers like lecithin–chitosan, often combined with polyethylene glycol (PEG)-modified surfactants, further enhance solubility, systemic retention, and cellular uptake of compounds such as epicatechin, quercetin, ursolic acid, thymoquinone, naringenin, and berberine in various cancer models (Perez-Ruiz et al., 2018; Wang et al., 2021; Shivani et al., 2023; Hsu et al., 2024; Selmi et al., 2023; Bhia et al., 2021). Beyond improving solubility, nanosystems can also overcome structural barriers to reach tumors more effectively. In melanoma models, transferosomes, ultradeformable liposomes that can squeeze through narrow intercellular gaps (Mohaddish et al., 2025), enabled increased skin penetration and boosted antitumor activity of flavonoid-rich extracts (Motawea et al., 2024). Additionally, incorporating matrix-modulating agents such as hyaluronidase further enhances intratumoral penetration of quercetin and resveratrol (Sun et al., 2025; Yu et al., 2024).

Controlled-release strategies exploit tumor-specific cues to trigger release, maintaining therapeutic levels while reducing systemic toxicity and dosing (Park et al., 2021). Chemical-responsive systems use acid-labile or disulfide linkages to destabilize in the acidic, glutathione-rich tumor milieu. Examples include quercetin in pH-responsive graphene-oxide/polymer nanocarriers, epigallocatechin-3-gallate in pH-sensitive nanoparticles, and ursolic acid delivered either in chitosan-modified liposomes (pH-responsive) or as a redox-responsive polymeric prodrug, all showing enhanced intracellular release under tumor-mimicking conditions (Matiyani et al., 2022; Bhattacharya et al., 2024; Wang et al., 2017; Fu et al., 2021). Enzyme-responsive carriers, such as matrix metalloproteinases (MMP)-cleavable block copolymers, exploit selective cleavage by tumor-overexpressed proteases (Vizovisek et al., 2021), achieving faster release than non-cleavable controls (Padmavathy et al., 2018). Similarly, dendrimers and hydrogels can be engineered for the progressive degradation of their structural networks via proteolysis to sustain polyphenol release and improve dispersibility (Ben-Zichri et al., 2022), and localized delivery of silibinin while minimizing burst-related toxicity (Yin et al., 2023). Targeted nanodelivery augments passive accumulation by decorating carriers with ligands for tumor-overexpressed receptors or organ-specific uptake, thereby boosting in-tumor exposure of phytochemicals while sparing normal tissues (Kalia and Kaur, 2020). Collectively, strategies such as folate, RGD (Arg-Gly-Asp) peptide, apolipoprotein E (ApoE), galactose, and epithelial cell adhesion molecule (EpCAM) have been applied to resveratrol, quercetin, epigallocatechin-3-gallate, curcumin, betulinic acid, paclitaxel, and others across diverse tumor types, in some cases also eliciting anti-tumorigenic immune responses (Barbarisi et al., 2018; Kazi et al., 2020; Dutta et al., 2024; Chen et al., 2022; Hu et al., 2021). These have enabled delivery to hard-to-reach tumors such as gliomas (via transferrin, lactoferrin, or sialic acid-mediated blood–brain barrier (BBB) transport) and to cancer stem cell niches through CD44-targeted systems (Guo et al., 2013; Yang et al., 2017; Kuo et al., 2019; Mittal et al., 2023). These ligand-mediated approaches expand the range of tumors accessible to phytochemical-based nanotherapy, including those protected by physiological barriers or with low passive accumulation.

Among these strategies, curcumin, a benchmark compound, has been incorporated into nearly all major nanocarrier platforms, exemplifying design principles that can be applied to other phytochemicals (Zandieh et al., 2023). Similarly, while whole plant extracts remain under exploration, batch variability complicates standardization and regulation compared to isolated compounds (Ukwubile et al., 2025; Alabrahim et al., 2024; Guillén-Meléndez et al., 2024). In addition, plant-derived exosome-like nanoparticles have emerged as biogenic delivery platforms that naturally encapsulate and transport phytochemicals across biological barriers, offering high biocompatibility, therapeutic potential, targeting capability, and efficient cellular uptake (Kim et al., 2022; Sha et al., 2024).

2.2 Mechanistic insights and clinical translation

Phytochemicals delivered through nanocarriers produce antiproliferative and pro-apoptotic effects by modulating core oncogenic cascades as well as oxidative-stress sensing via Kelch-like ECH-associated protein 1–nuclear factor erythroid 2–related factor 2 (Keap1–Nrf2) (Parvin et al., 2025; Aljabali et al., 2025; Situmorang et al., 2024). Certain subclasses display mechanistic preferences: polyphenols like resveratrol and curcumin frequently downregulate nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3), impairing inflammatory and survival cascades (Dhupal and Chowdhury, 2020); flavonoids including quercetin and epigallocatechin gallate modulate phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, restricting proliferation and metabolic adaptation (Melim et al., 2022); terpenoids such as ursolic acid and oridonin directly interfere with STAT3, heat shock protein 70 (HSP70), and other protein effectors (Yao et al., 2023); and alkaloids like berberine predominantly target AMP-activated protein kinase (AMPK)-dependent metabolic reprogramming, indirectly suppressing mTOR-driven growth (Hashim et al., 2024; Cheng and Ji, 2020). In parallel, phytochemicals also alter glycolysis, glutamine dependence, and lipid biosynthesis, reducing tumor metabolic plasticity (Dey et al., 2021; Hashim et al., 2024; Shuvalov et al., 2023; Wu et al., 2021). Epigenetically, phytochemicals, notably polyphenols and terpenoids, can alter histone acetylation and DNA methylation, as well as modulate miRNA expression, restoring silenced tumor suppressor genes and reducing oncogene expression, yielding antiproliferative, pro-apoptotic, and resensitizing effects (Aljabali et al., 2025; Dandawate et al., 2016; Roy and Datta, 2019). These molecular-level interventions converge to restrict both bulk tumor populations and therapy-persistent subclones, including cancer stem-like cells (Dandawate et al., 2016; Melim et al., 2022; Naujokat and McKee, 2021).

Beyond intrinsic signaling, nanocarrier-delivered phytochemicals modulate the tumor microenvironment (TME) by regulating fibroblast activation, extracellular matrix remodeling, angiogenesis, and immune cell function (Melim et al., 2022; Aljabali et al., 2025). Polyphenols blunt epithelial-to-mesenchymal transition (EMT)–metastasis programs and hypoxia-inducible factor 1α/vascular endothelial growth factor (HIF-1α/VEGF)-driven angiogenesis, flavonoids modulate stemness pathways such as Wingless/Integrated (Wnt)/β-catenin, Hedgehog, and Notch, and terpenoids engage apoptosis and p53-centered DNA-damage checkpoints (Situmorang et al., 2024; Aljabali et al., 2025; Liu et al., 2023; Ci et al., 2016; Kim et al., 2024; Pan et al., 2025; Shuvalov et al., 2023). Importantly, phytochemicals enhance antigen presentation and cytotoxic T-cell infiltration and modulate immunosuppressive mediators such as programmed death-ligand 1 (PD-L1) and transforming growth factor-β (TGF-β) in a compound- and context-dependent manner (Parvin et al., 2025; Dhupal and Chowdhury, 2020). They also promote macrophage polarization from an M2-like to an M1-like phenotype and suppress the accumulation of myeloid-derived suppressor cells, thereby restoring antitumor immune surveillance (Yao et al., 2023; Aljabali et al., 2025). Resveratrol, curcumin, and catechins synergize with checkpoint inhibitors by reprogramming the immunosuppressive microenvironment, while ursolic acid and quercetin modulate inflammatory factors to shift TME balance toward tumor rejection (Chen et al., 2020; Guven et al., 2022; Li et al., 2023b). Combination nanoformulations, widely explored for curcumin, resveratrol, quercetin, and other phytochemicals, have enabled the co-delivery of chemotherapeutics or radiotherapy to achieve synergistic antitumor effects (Kang et al., 2018; Cheng and Ji, 2020; Lv et al., 2016; Wongrakpanich et al., 2024; Afereydoon et al., 2022; AbouAitah et al., 2022; Afshari et al., 2023; Corte-Real et al., 2024), counteracting multidrug resistance by downregulating efflux transporters and disrupting pro-survival metabolic adaptations (Parvin et al., 2025; Melim et al., 2022; Li et al., 2019). More recently, nanocarriers have been engineered to co-deliver therapeutic genes, such as p53, regulatory RNAs (siRNA and miRNA targeting PD-L1, survivin, VEGF, B-cell lymphoma 2 (Bcl-2), and others), and naturally occurring cytolytic peptides, thereby enhancing therapeutic efficacy through gene silencing, apoptosis induction, and immune modulation (Ashrafizadeh et al., 2020; Bhagavatheeswaran and Balakrishnan, 2023; Li X. et al., 2022; Eksi et al., 2025; Wang et al., 2025; Xu et al., 2018; Sun et al., 2023). Collectively, these combination approaches leverage multiple signaling and metabolic pathways, reprogram the tumor microenvironment, and mitigate therapeutic resistance.

Several phytochemical nanoformulations, such as paclitaxel (Abraxane®), irinotecan (Onivyde®), vinorelbine (NanoVNB®), vincristine (Marqibo®), and docetaxel (DoceAqualip®), are already FDA-approved for cancer therapy (Dhupal and Chowdhury, 2020), while multiple curcumin-, camptothecin-, ursolic acid-, mangiferin-, and quercetin-based platforms are undergoing clinical evaluation in liposomal, polymeric, micellar, and plant-derived nanocarriers (Devan et al., 2023; Lekhak and Bhattarai, 2024; Kumar et al., 2024). Patent activity highlights curcumin-, resveratrol-, and quercetin-based nanosystems, such as liposomes, polymeric nanoparticles, and plant-derived vesicles, designed for improved solubility, stability, and tumor targeting (Table 1). Translation remains limited by pharmacokinetic, scale-up, and regulatory hurdles; standardized characterization and robust preclinical models and clinical design will be critical for bringing these promising nanosystems from bench to bedside.

Table 1
www.frontiersin.org

Table 1. Advances in phytochemical-based nanosystems for cancer therapy: patent trends and microfluidic production approaches.

3 Microfluidic technologies driving innovation in design and evaluation

3.1 Microfluidic synthesis of phytochemical nanocarriers: precision and scalability

One of the unique properties of microfluidics is laminar flow, an ordered parallel flow devoid of any fluid layer disruption, which confers constant continuous mixing through the process (Jaradat et al., 2022). In this profile, diffusion plays a key role in evening out concentration differences at a molecular level (Cullen and Misra, 2015). Microfluidics creates steep spatial and temporal solubility gradients, critical for uniform, well-defined nanoparticles (Nunziata et al., 2025). These properties confer a superior mixing quality, assure the same production quality over time with minimal intra-batch, reduced batch-to-batch, and operator variability during scale-up (Cai et al., 2022).

Nanoparticle synthesis comprises three stages: nucleation, growth, and particle separation (Siavashy et al., 2021). Batch nanoparticle synthesis methods lack control in particle growth, mixing, and separation to prevent agglomeration, ultimately causing fluctuations in size distribution and a diverse particle assortment with varying chemical and physical traits, which restricts the synthesis of core–shell nanoparticles and diminishes encapsulation efficiencies (EE). On the other hand, microfluidics enables precise flow rate control (Cai et al., 2022). Then, it can overcome some of the large-scale reactor intricate hurdles as it tackles variability and scalability issues, which are major conventional batch technique concerns (Nunziata et al., 2025). In the nanocarrier synthesis realm, these advantages have been exploited to control attributes such as size, size distribution, and drug loading (Table 1).

Regarding polymeric nanoparticles, chitosan is an FDA-recognized biopolymer cleared for use in wound-healing devices and with limited Generally Recognized As Safe (GRAS) status in specific food applications, also widely investigated for drug delivery systems (Shah et al., 2025; Naghib et al., 2024). Beyond its prior use in nanomedicine, it has more recently been integrated into microfluidic platforms for the controlled synthesis of nanoparticles, underscoring its translational relevance (Siavashy et al., 2021). More broadly, microfluidic approaches have enabled the synthesis of other polymeric nanoparticles as well. For example, using the innovative multi-stage microfluidic TrH chip, hybrid nanoparticles co-encapsulating paclitaxel-simvastatin and paclitaxel-lenvatinib were successfully produced, demonstrating the versatility of microfluidic platforms for multi-drug-loaded nanomedicines (Li et al., 2025).

Regarding liposome preparation, microhydrodynamic focusing (MHF) (Weaver et al., 2022) and herringbone micromixer (Pisani et al., 2022) have been utilized. MHF presents promising results in producing liposomal formulations with low polydispersity index (PDI) by a one-step procedure (Bochicchio et al., 2020). In MHF, two fluid streams are introduced into a microchannel, where one stream flows at the center and is enveloped by another. This mixture relies on the diffusion mechanism to blend two reagents, wrestling in a lower throughput of around a hundred μL/min. In contrast, herringbone micromixers boast a higher throughput of approximately a few mL/min. Most studies utilizing this configuration rely on a commercialized chip that requires specialized equipment, the NanoAssemblr™ bench-top instrument (Precision Nano Systems Inc., Vancouver, Canada). This device is designed for nanocarrier production and is commercially accessible for research endeavors. In fact, the characteristics of eleven liposomal docetaxel formulations prepared using the NanoAssemblr™ bench-top instrument were assessed by (Dacos et al., 2024). Indeed, the liposomal delivery system for curcumin (Lipo-Cur) was developed utilizing automated microfluidics. When administered alongside cisplatin to mice with tumors, Lipo-Cur strengthened the cisplatin antitumor effectiveness across various mouse tumor models while mitigating nephrotoxicity (Hamano et al., 2019). In this connection, umbelliprenin was co-encapsulated with doxorubicin in liposomes, and this combination, prepared with microfluidics, induced higher toxicity than liposomes prepared with the thin-film method, with an IC50 (half-maximal inhibitory concentration) at least 2-fold lower. This feature was attributed to different release kinetics. Furthermore, they discovered that umbelliprenin affected the viscoelastic behaviour and the lipid biomembrane fluidity (Gkionis et al., 2020).

In this regard, (Jaradat et al., 2022), prepared liposomes to encapsulate paclitaxel and determined the best lipid candidates for nanocarrier synthesis. They found that microfluidics has a significant effect in improving the EE of paclitaxel compared to other conventional methods, such as film hydration and extrusion (EE% <50%). Furthermore, they observed that MHF enhanced 1,2-Dimyristoyl-sn-glycero-3-phosphocholine (DMPC) and 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) EE%. In addition, these lipids provided a smaller particle size due to their short acyl length. Besides, paclitaxel loading in both DMPC and DPPC liposomes exhibits higher packing with DPPC and shows a sustained release profile. Importantly, the delayed release (after 24 h) can be an advantage in limiting the collateral toxicity to normal tissue due to the reduced premature release.

In fact, continuous flow achieved through microfluidics provides better heat mass transport, and it enables multiple unit operations (Guo et al., 2023). These properties were applied to successfully load curcumin, a hydrophobic polyphenol extracted from the rhizomes of Curcuma longa, into zein-SH nanoparticles by establishing a robust and controllable solvent-antisolvent laminar diffusion, achieving a millisecond short mixing time and a homogeneous particle size distribution. They improved EE and loading capacity (LC) (Table 1, Part B) by applying microfluidics when compared to bulk mixing prepared nanoparticles (EE% = 7.7 ± 0.5, LC% = 0.4 ± 0.5). They found that at a high flow ratio, the nucleated nanoparticles are rapidly diluted in the antisolvent and kinetically locked, preventing further size growth. When prepared by bulk mixing, they observed spherical nanoparticles connected by dendritic structures that form due to turbulent mixing in some areas (Guo et al., 2023).

Furthermore, microfluidics offers outstanding opportunities for the nanodrug delivery systems production processes as it enhances controllability and uniformity (Zhang et al., 2023). In spite of the remarkable progress made in the creation and assessment of these systems through microfluidics, the shift of this innovative technology into actual industrial applications remains a hurdle. Achieving a kilogram production output or beyond each day is essential for both clinical investigations and large-scale manufacturing (Liu et al., 2018). However, the daily microfluidic-assisted-nanoparticle-production rate is usually in the milligram range (Kim et al., 2012). In order to address this issue, a nanoparticle production rate up to 3 kg/day was achieved by developing a coaxial turbulent jet mixer, which is suitable for industrial-scale production of nanodrug delivery systems (Lim et al., 2014). Other explored approaches are incrementing the channel dimensions (Gomez et al., 2014), and microfluidic channel parallelization (Shepherd et al., 2021). In this context, using an immobilized liquid lubricant perfluorodecalin layer was proposed by (Hwang et al., 2025) to prevent RNA-loaded lipid nanoparticles fouling. This technology was applied to a staggered herringbone microfluidic (SHM) mixing chip and reached more than 3 h of stable operation. Furthermore, they demonstrated this strategy’s compatibility with a parallelized microfluidic platform that gathers 256 SHM mixers, which assures stable production at L/h production rates suitable for commercial-scale applications.

3.2 Lab-on-chip tools for modeling tumor complexity and response

Microfluidic devices have emerged as an efficient tool for modeling tumor and normal tissue microenvironments (Nasiri et al., 2025b). Their ability to replicate tissue physiology and integrate biomechanical factors such as extracellular matrix and fluid dynamics parameters as flow rate, pressure, viscosity, surface tension, shear stress, and wettability (Liu et al., 2021; Sunildutt et al., 2023), has allowed them to create platforms to mimic the heterogeneity and complex cell organization and study cancer cell treatments, tumor evolution, chemosensibility, metastasis, and cell migration (Dsouza et al., 2022; Nejati et al., 2025; Ding et al., 2025). This performance is further enhanced by nanomaterials, which mainly improve detection sensitivity and biocompatibility (Tian et al., 2025).

Building on these advances, organ-on-a-chip (Ooc) represents a next-generation approach, capable of recreating controlled micro- and nanoenvironments in real time (Dsouza et al., 2022). These advanced biomimetic systems combine two or more cell types, including those derived from patients (Liu et al., 2025; Nejati et al., 2025), with microfluidics to replicate tumor heterogeneity, vascular networks, and three-dimensional architecture, facilitating the evaluation of drugs (Ma C. et al., 2021; Tian et al., 2025; Ayuso et al., 2022). In recent years, Ooc platforms have been used to assess the anticancer activity and phytochemical toxicity, for instance, Spatholobi Caulis tannin in cervical cancer (Wang et al., 2018), cis-stilbene glycoside and emodin-8-O-β-D-glucoside from Polygonum multiflorum in liver cancer (Deng et al., 2025), and panaxatriol from Panax ginseng C.A. Mayer in lung adenocarcinoma (Nasiri et al., 2025a). Additionally, this technology enables the study of complementary TME such as cell morphology, inflammatory process, migration process, protein expression, enzymatic activity, and oxygen and nutrient supply (Farooqi, 2022; Feng et al., 2025; Boul et al., 2021).

The integration of nanotechnology further enhances the phytochemical bioactivity, which has been evaluated in Ooc platforms. For instance, (Sharifi et al., 2020), used a liver-chip model to evaluate and compare anticancer activity of thymoquinone from Nigella sativa, both in its free form and encapsulated in chitosan-based nanoparticles. The results emphasize the potential of these systems to improve the analysis of antimetastatic activity, proliferation, migration, and colonization of tumor cells when encapsulated phytochemicals are delivered in tumor microenvironments.

Specialized Ooc platforms have been developed to address specific cancer-related processes. Vascular-on-a-chip models mimic angiogenesis and vascular responses; (Fayazbakhsh et al., 2023); used such a system to mimic blood vessels structure in angiogenesis process and assess the antioxidant effects of resveratrol-loaded gold nanoparticles on human umbilical vein endothelial cells under hyperglycemic conditions. The system enabled precise collagen level modulation and reactive oxygen species (ROS) real-time monitoring, revealing their reduction. BBB-on-a-chip system replicates central nervous system barriers to test drug delivery to brain tumors and evaluate parameters such as homeostasis and permeability. For instance, (Shi et al., 2023), developed a BBB-on-a-chip model using microvascular endothelial cells, pericytes, and mast cells to mimic the glioma microenvironment, enabling synergistic effect evaluation from traditional Chinese medicine phytochemicals to improve drug delivery and efficacy (Garcia et al., 2023) created a model combining 3 cell types: brain cells, human astrocytes-hippocampal (Ha-h), and Human brain vascular pericytes (HBVP), to test permeability and internalization of PLGA-encapsulated ferulic acid. Results showed improved internalization of this hydrophobic antioxidant, reduced ROS levels, and suggested nanoparticle size influences BBB permeability.

In addition, tumor-on-a-chip integrates tumor and stromal cells instead of healthy tissue cells, to mimic invasive tumor behaviour and TME dynamics (Li et al., 2023a; Tian et al., 2022), enabling a close evaluation of the anticancer potential of phytochemical extracts under conditions compared to 2D cultures. (Farooqi et al., 2022). used this model to evaluate anticancer activity of A. cappadocicum methanolic extract as well as to study ROS real-time monitoring, superoxide dismutase activity, and tumor biomarkers (e.g., urea, albumin) in the liver. Similarly, (Martins et al., 2023), designed a single-channel microfluidic devices with human glioblastoma cells to test the efficacy of free and nanoparticle-encapsulated docetaxel, reporting up to 50-fold lower IC50 values compared to conventional 2D monolayers, indicating higher tumor susceptibility under microfluidic conditions. Finally, spheroid on a chip combines multicellular tumor spheroids with microfluidics to assess long-term drug responses under perfusion, allowing compression of in vivo tumorigenesis and processes such as apoptosis and cell viability (Uzabakiriho et al., 2025; Nashimoto et al., 2020). For instance, a model with endothelial cells revealed that perfusion modulates paclitaxel sensitivity, underscored the role of stromal cells in angiogenesis, and identified flow rate as a determinant of drug efficacy and therapeutic response (Nashimoto et al., 2020).

Altogether, Ooc platforms provide a versatile and physiologically relevant technology for studying anticancer phytochemicals, nanoparticle-based therapies, tumor heterogeneity, and TME processes, bridging the gap between conventional 2D in vitro assays and in vivo models.

4 Conclusion

Phytochemical-based nanosystems enhance tumor-targeted therapy through multi-pathway modulation, improved bioavailability, stability, sustained and controlled release and efficacy. Among the different approaches, liposomes appear to be the most promising nanosystems in phytochemical delivery due to their properties such as: functional surface, low toxicity and minimal impact on healthy tissues. In this context, microfluidics enables the synthesis of precise-controlled size nanocarriers with potent activity for both laboratory research and industry settings. Although large-scale manufacturing remains challenging, parallelization emerged as a key strategy to scale production from milliliters to liters per hour. Lab-on-a-chip platforms complement microfluidic synthesis by providing biomimetic evaluation of transport, penetration, and response, facilitating patient-derived assays, increasing fidelity, lowering costs, and enhancing translational potential. Although several phytochemical nanomedicines have achieved FDA approval, microfluidic applications remain preclinical; nevertheless, ongoing innovation and patent activity highlight their promise for clinical translation.

Author contributions

ABP-P: Writing – original draft, Writing – review and editing. MC: Writing – original draft, Writing – review and editing. RG-P: Conceptualization, Writing – original draft, Writing – review and editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

The authors gratefully acknowledge Carlos Ulloa for his help during information gathering. Figures were created with BioRender.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

AbouAitah, K., Soliman, A. A. F., Swiderska-Sroda, A., Nassrallah, A., Smalc-Koziorowska, J., Gierlotka, S., et al. (2022). Co-Delivery system of curcumin and colchicine using functionalized mesoporous Silica nanoparticles promotes anticancer and apoptosis effects. Pharmaceutics 14, 2770. doi:10.3390/pharmaceutics14122770

PubMed Abstract | CrossRef Full Text | Google Scholar

Afereydoon, S., Haghiralsadat, F., Hamzian, N., Shams, A., Hemati, M., Naghib, S. M., et al. (2022). Multifunctional PEGylated niosomal nanoparticle-loaded herbal drugs as a novel nano-radiosensitizer and stimuli-sensitive nanocarrier for synergistic cancer therapy. Front. Bioeng. Biotechnol. 10, 917368. doi:10.3389/fbioe.2022.917368

PubMed Abstract | CrossRef Full Text | Google Scholar

Afshari, A. R., Sanati, M., Kesharwani, P., and Sahebkar, A. (2023). Recent advances in curcumin-based combination nanomedicines for cancer therapy. J. Funct. Biomater. 14, 408. doi:10.3390/jfb14080408

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Naqeb, G., Kalmpourtzidou, A., Giampieri, F., De Giuseppe, R., and Cena, H. (2024). Genotoxic and antigenotoxic medicinal plant extracts and their main phytochemicals: “A review”. Front. Pharmacol. 15, 1448731. doi:10.3389/fphar.2024.1448731

PubMed Abstract | CrossRef Full Text | Google Scholar

Alabrahim, O. A. A., Lababidi, J. M., Fritzsche, W., and Azzazy, H. M. E.-S. (2024). Beyond aromatherapy: can essential oil loaded nanocarriers revolutionize cancer treatment? Nanoscale Adv. 6, 5511–5562. doi:10.1039/d4na00678j

PubMed Abstract | CrossRef Full Text | Google Scholar

Aljabali, A. A. A., Obeid, M. A., Bashatwah, R. M., Qnais, E., Gammoh, O., Alqudah, A., et al. (2025). Phytochemicals in cancer therapy: a structured review of mechanisms, challenges, and progress in personalized treatment. Chem. Biodivers. 22, e202402479. doi:10.1002/cbdv.202402479

PubMed Abstract | CrossRef Full Text | Google Scholar

Ang, L. F., Darwis, Y., Por, L. Y., and Yam, M. F. (2019). Microencapsulation curcuminoids for effective delivery in pharmaceutical application. Pharmaceutics 11, 451. doi:10.3390/pharmaceutics11090451

PubMed Abstract | CrossRef Full Text | Google Scholar

Ashrafizadeh, M., Zarrabi, A., Hushmandi, K., Hashemi, F., Rahmani Moghadam, E., Raei, M., et al. (2020). Progress in natural Compounds/siRNA Co-delivery employing nanovehicles for cancer therapy. ACS Comb. Sci. 22, 669–700. doi:10.1021/acscombsci.0c00099

PubMed Abstract | CrossRef Full Text | Google Scholar

Asma, S. T., Acaroz, U., Imre, K., Morar, A., Shah, S. R. A., Hussain, S. Z., et al. (2022). Natural products/bioactive compounds as a source of anticancer drugs. Cancers (Basel) 14, 6203. doi:10.3390/cancers14246203

PubMed Abstract | CrossRef Full Text | Google Scholar

Ayuso, J. M., Virumbrales-Muñoz, M., Lang, J. M., and Beebe, D. J. (2022). A role for microfluidic systems in precision medicine. Nat. Commun. 13, 3086. doi:10.1038/s41467-022-30384-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Babazadeh, A., Zeinali, M., and Hamishehkar, H. (2018). Nano-Phytosome: a developing platform for herbal anti-cancer agents in cancer therapy. Curr. Drug Targets 19, 170–180. doi:10.2174/1389450118666170508095250

PubMed Abstract | CrossRef Full Text | Google Scholar

Barbarisi, M., Iaffaioli, R. V., Armenia, E., Schiavo, L., De Sena, G., Tafuto, S., et al. (2018). Novel nanohydrogel of hyaluronic acid loaded with quercetin alone and in combination with temozolomide as new therapeutic tool, CD44 targeted based, of glioblastoma multiforme. J. Cell. Physiol. 233, 6550–6564. doi:10.1002/jcp.26238

PubMed Abstract | CrossRef Full Text | Google Scholar

Ben-Zichri, S., Meltzer, M., Lacham-Hartman, S., Kolusheva, S., Hadad, U., Papo, N., et al. (2022). Synergistic activity of anticancer polyphenols embedded in amphiphilic dendrimer nanoparticles. ACS Appl. Polym. Mater. 4, 8913–8925. doi:10.1021/acsapm.2c01316

CrossRef Full Text | Google Scholar

Bezelya, A., Küçüktürkmen, B., and Bozkır, A. (2023). Microfluidic devices for precision nanoparticle production. Micro 3, 822–866. doi:10.3390/micro3040058

CrossRef Full Text | Google Scholar

Bhagavatheeswaran, S., and Balakrishnan, A. (2023). “Phytochemicals and miRNA-Based therapeutic opportunities for cancer stem cell-mediated tumor angiogenesis,” in Handbook of oncobiology: from basic to clinical sciences. Editors R. C. Sobti, N. K. Ganguly, and R. Kumar (Singapore: Springer Nature Singapore), 1–11. doi:10.1007/978-981-99-2196-6_32-1

CrossRef Full Text | Google Scholar

Bhattacharya, S., Sangave, P. C., Belemkar, S., and Anjum, M. M. (2024). pH-Sensitive nanoparticles of Epigallocatechin-3-Gallate in enhanced colorectal cancer therapy. Nanomedicine (Lond) 19, 459–481. doi:10.2217/nnm-2023-0342

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhia, M., Motallebi, M., Abadi, B., Zarepour, A., Pereira-Silva, M., Saremnejad, F., et al. (2021). Naringenin nano-delivery systems and their therapeutic applications. Pharmaceutics 13, 291. doi:10.3390/pharmaceutics13020291

PubMed Abstract | CrossRef Full Text | Google Scholar

Bilia, A. R., Piazzini, V., Risaliti, L., Vanti, G., Casamonti, M., Wang, M., et al. (2019). Nanocarriers: a successful tool to increase solubility, stability and optimise bioefficacy of natural constituents. Curr. Med. Chem. 26, 4631–4656. doi:10.2174/0929867325666181101110050

PubMed Abstract | CrossRef Full Text | Google Scholar

Bińkowska, W., Szpicer, A., Stelmasiak, A., Wojtasik-Kalinowska, I., and Półtorak, A. (2024). Microencapsulation of polyphenols and their application in food technology. Appl. Sci. 14, 11954. doi:10.3390/app142411954

CrossRef Full Text | Google Scholar

Bochicchio, S., Dalmoro, A., De Simone, V., Bertoncin, P., Lamberti, G., and Barba, A. A. (2020). Simil-Microfluidic nanotechnology in manufacturing of liposomes as hydrophobic antioxidants skin release systems. Cosmetics 7, 22. doi:10.3390/cosmetics7020022

CrossRef Full Text | Google Scholar

Boul, M., Benzoubir, N., Messina, A., Ghasemi, R., Mosbah, I. B., Duclos-Vallée, J.-C., et al. (2021). A versatile microfluidic tool for the 3D culture of HepaRG cells seeded at various stages of differentiation. Sci. Rep. 11, 14075. doi:10.1038/s41598-021-92011-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Brewer, C. T., and Chen, T. (2017). Hepatotoxicity of herbal supplements mediated by modulation of cytochrome P450. Int. J. Mol. Sci. 18, 2353. doi:10.3390/ijms18112353

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, Q., Castagnola, V., Boselli, L., Moura, A., Lopez, H., Zhang, W., et al. (2022). A microfluidic approach for synthesis and kinetic profiling of branched gold nanostructures. Nanoscale Horiz. 7, 288–298. doi:10.1039/d1nh00540e

PubMed Abstract | CrossRef Full Text | Google Scholar

Campos, K. R., Coleman, P. J., Alvarez, J. C., Dreher, S. D., Garbaccio, R. M., Terrett, N. K., et al. (2019). The importance of synthetic chemistry in the pharmaceutical industry. Science 363, eaat0805. doi:10.1126/science.aat0805

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Liu, Q., Zhu, Z., Xiang, F., Zhang, M., Wu, R., et al. (2020). Ursolic acid protects against proliferation and inflammatory response in LPS-Treated gastric tumour model and cells by inhibiting NLRP3 inflammasome activation. Cancer Manag. Res. 12, 8413–8424. doi:10.2147/CMAR.S264070

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, B., Liu, X., Li, Y., Shan, T., Bai, L., Li, C., et al. (2022). iRGD tumor-penetrating peptide-modified nano-delivery System based on a marine sulfated polysaccharide for enhanced anti-tumor efficiency against breast cancer. Int. J. Nanomedicine 17, 617–633. doi:10.2147/IJN.S343902

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Tang, Y., Li, Y., Rui, Y., and Zhang, P. (2024). Enhancing the efficacy of active pharmaceutical ingredients in medicinal plants through nanoformulations: a promising field. Nanomater. (Basel) 14, 1598. doi:10.3390/nano14191598

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, Y., and Ji, Y. (2020). Mitochondria-targeting nanomedicine self-assembled from GSH-responsive paclitaxel-ss-berberine conjugate for synergetic cancer treatment with enhanced cytotoxicity. J. Control. Release 318, 38–49. doi:10.1016/j.jconrel.2019.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Choudhari, A. S., Mandave, P. C., Deshpande, M., Ranjekar, P., and Prakash, O. (2019). Phytochemicals in cancer treatment: from preclinical studies to clinical practice. Front. Pharmacol. 10, 1614. doi:10.3389/fphar.2019.01614

PubMed Abstract | CrossRef Full Text | Google Scholar

Ci, Y., Qiao, J., and Han, M. (2016). Molecular mechanisms and metabolomics of natural polyphenols interfering with breast cancer metastasis. Molecules 21, 1634. doi:10.3390/molecules21121634

PubMed Abstract | CrossRef Full Text | Google Scholar

Corte-Real, M., Veiga, F., Paiva-Santos, A. C., and Pires, P. C. (2024). Improving skin cancer treatment by dual drug Co-Encapsulation into liposomal Systems-An integrated approach towards anticancer synergism and targeted delivery. Pharmaceutics 16, 1200. doi:10.3390/pharmaceutics16091200

PubMed Abstract | CrossRef Full Text | Google Scholar

Cullen, P. J., and Misra, N. N. (2015). “Laminar mixing fundamentals,” in Pharmaceutical blending and mixing. Editors P. J. Cullen, R. J. Romañach, N. Abatzoglou, and C. D. Rielly (Wiley), 43–56. doi:10.1002/9781118682692.ch3

CrossRef Full Text | Google Scholar

Dacos, M., Immordino, B., Diroff, E., Sicard, G., Kosta, A., Rodallec, A., et al. (2024). Pegylated liposome encapsulating docetaxel using microfluidic mixing technique: process optimization and results in breast cancer models. Int. J. Pharm. 656, 124091. doi:10.1016/j.ijpharm.2024.124091

PubMed Abstract | CrossRef Full Text | Google Scholar

Dandawate, P. R., Subramaniam, D., Jensen, R. A., and Anant, S. (2016). Targeting cancer stem cells and signaling pathways by phytochemicals: novel approach for breast cancer therapy. Semin. Cancer Biol. 40 (41), 192–208. doi:10.1016/j.semcancer.2016.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, S., Das, M. K., Deka, T., Singha, L. R., and Das, P. (2020). “Nanomedicines and nanodrug delivery systems: trends and perspectives,” in Nano medicine and nano safety: recent trends and clinical evidences. Editors M. K. Das, and Y. V. Pathak (Singapore: Springer Singapore), 99–141. doi:10.1007/978-981-15-6255-6_6

CrossRef Full Text | Google Scholar

Deng, Q., Qu, Y., Luo, Y., and Zhang, X. (2025). An immune-liver microphysiological system method for evaluation and quality control of hepatotoxicity induced by Polygonum multiflorum thunb. Extract. J. Ethnopharmacol. 345, 119633. doi:10.1016/j.jep.2025.119633

PubMed Abstract | CrossRef Full Text | Google Scholar

Devan, A. R., Nair, B., and Nath, L. R. (2023). Translational phytomedicines against cancer: promise and hurdles. Adv. Pharm. Bull. 13, 210–215. doi:10.34172/apb.2023.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Dey, P., Kundu, A., Kar, B., Bhakta, A., Vishal, V., Keerthana, S., et al. (2021). “Bioactive natural leads targeting cancer cell metabolism,” in Evidence based validation of traditional medicines: a comprehensive approach. Editors S. C. Mandal, R. Chakraborty, and S. Sen (Singapore: Springer Singapore), 29–75. doi:10.1007/978-981-15-8127-4_2

CrossRef Full Text | Google Scholar

Dhupal, M., and Chowdhury, D. (2020). Phytochemical-Based nanomedicine for advanced cancer theranostics: perspectives on clinical trials to clinical use. Int. J. Nanomedicine 15, 9125–9157. doi:10.2147/IJN.S259628

PubMed Abstract | CrossRef Full Text | Google Scholar

Ding, S., Ding, W., Wang, X., Li, Z., Fan, Y., Lin, F., et al. (2025). Sorting and characterization of cancer cells with different migration phenotypes using a microfluidic platform. Sensors Actuators B Chem. 440, 137875. doi:10.1016/j.snb.2025.137875

CrossRef Full Text | Google Scholar

Dorato, M. A., and Engelhardt, J. A. (2005). The no-observed-adverse-effect-level in drug safety evaluations: use, issues, and definition(s). Regul. Toxicol. Pharmacol. 42, 265–274. doi:10.1016/j.yrtph.2005.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Dsouza, V. L., Kuthethur, R., Kabekkodu, S. P., and Chakrabarty, S. (2022). Organ-on-Chip platforms to study tumor evolution and chemosensitivity. Biochim. Biophys. Acta Rev. Cancer 1877, 188717. doi:10.1016/j.bbcan.2022.188717

PubMed Abstract | CrossRef Full Text | Google Scholar

Dutta, D., Al Hoque, A., Paul, B., Park, J. H., Chowdhury, C., Quadir, M., et al. (2024). EpCAM-targeted betulinic acid analogue nanotherapy improves therapeutic efficacy and induces anti-tumorigenic immune response in colorectal cancer tumor microenvironment. J. Biomed. Sci. 31, 81. doi:10.1186/s12929-024-01069-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Eksi, O. B., Guler, A., Akdeniz, M., Atalay, P., Hamurcu, Z., and Aydin, O. (2025). Development of silver-based hybrid nanoparticles loaded with eEF2 K-siRNA and quercetin against triple-negative breast cancer. Drug Deliv. Transl. Res. doi:10.1007/s13346-025-01860-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Farooqi, M. A. (2022). Effect of methanolic extracts of Acer Cappadocicum on HepG2 cancer cell line in a liver microphysiological System.

Google Scholar

Farooqi, H. M. U., Sammantasinghar, A., Kausar, F., Farooqi, M. A., Chethikkattuveli Salih, A. R., Hyun, K., et al. (2022). Study of the anticancer potential of plant extracts using liver tumor microphysiological system. Life (Basel) 12, 135. doi:10.3390/life12020135

PubMed Abstract | CrossRef Full Text | Google Scholar

Farooqi, M. A., Kang, C.-U., and Choi, K. H. (2023). Organ-on-Chip: advancing nutraceutical testing for improved health outcomes. ACS Omega 8, 31632–31647. doi:10.1021/acsomega.3c03155

PubMed Abstract | CrossRef Full Text | Google Scholar

Fayazbakhsh, F., Hataminia, F., Eslam, H. M., Ajoudanian, M., Kharrazi, S., Sharifi, K., et al. (2023). Evaluating the antioxidant potential of resveratrol-gold nanoparticles in preventing oxidative stress in endothelium on a chip. Sci. Rep. 13, 21344. doi:10.1038/s41598-023-47291-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, Q., Chen, H., Ren, M., Qiao, Y., Zou, J., Liang, X., et al. (2025). Protective effect of osmanthus water extract on liver dysfunction caused by DBP based on organoids and organ chips technologies. Food Res. Int. 219, 116976. doi:10.1016/j.foodres.2025.116976

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, D., Ni, Z., Wu, K., Cheng, P., Ji, X., Li, G., et al. (2021). A novel redox-responsive ursolic acid polymeric prodrug delivery system for osteosarcoma therapy. Drug Deliv. 28, 195–205. doi:10.1080/10717544.2020.1870583

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia, L., Palma-Florez, S., Espinosa, V., Soleimani Rokni, F., Lagunas, A., Mir, M., et al. (2023). Ferulic acid-loaded polymeric nanoparticles prepared from nano-emulsion templates facilitate internalisation across the blood-brain barrier in model membranes. Nanoscale 15, 7929–7944. doi:10.1039/d2nr07256d

PubMed Abstract | CrossRef Full Text | Google Scholar

George, B. P., Chandran, R., and Abrahamse, H. (2021). Role of phytochemicals in cancer chemoprevention: insights. Antioxidants 10, 1455. doi:10.3390/antiox10091455

PubMed Abstract | CrossRef Full Text | Google Scholar

Gkionis, L., Campbell, R. A., Aojula, H., Harris, L. K., and Tirella, A. (2020). Manufacturing drug co-loaded liposomal formulations targeting breast cancer: influence of preparative method on liposomes characteristics and in vitro toxicity. Int. J. Pharm. 590, 119926. doi:10.1016/j.ijpharm.2020.119926

PubMed Abstract | CrossRef Full Text | Google Scholar

Gomez, L., Sebastian, V., Irusta, S., Ibarra, A., Arruebo, M., and Santamaria, J. (2014). Scaled-up production of plasmonic nanoparticles using microfluidics: from metal precursors to functionalized and sterilized nanoparticles. Lab. Chip 14, 325–332. doi:10.1039/c3lc50999k

PubMed Abstract | CrossRef Full Text | Google Scholar

Guillén-Meléndez, G. A., Pérez-Hernández, R. A., Chávez-Montes, A., Castillo-Velázquez, U., de Jesús Loera-Arias, M., Montes-de-Oca-Saucedo, C. R., et al. (2024). Nanoencapsulation of extracts and isolated compounds of plant origin and their cytotoxic effects on breast and cervical cancer treatments: advantages and new challenges. Toxicon 244, 107753. doi:10.1016/j.toxicon.2024.107753

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, W., Li, A., Jia, Z., Yuan, Y., Dai, H., and Li, H. (2013). Transferrin modified PEG-PLA-resveratrol conjugates: in vitro and in vivo studies for glioma. Eur. J. Pharmacol. 718, 41–47. doi:10.1016/j.ejphar.2013.09.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, H., Feng, Y., Deng, Y., Yan, T., Liang, Z., Zhou, Y., et al. (2023). Continuous flow modulates zein nanoprecipitation solvent environment to obtain colloidal particles with high curcumin loading. Food Hydrocoll. 134, 108089. doi:10.1016/j.foodhyd.2022.108089

CrossRef Full Text | Google Scholar

Guven, D. C., Sahin, T. K., Rizzo, A., Ricci, A. D., Aksoy, S., and Sahin, K. (2022). The use of phytochemicals to improve the efficacy of immune checkpoint inhibitors: opportunities and challenges. Appl. Sci. 12, 10548. doi:10.3390/app122010548

CrossRef Full Text | Google Scholar

Hamano, N., Böttger, R., Lee, S. E., Yang, Y., Kulkarni, J. A., Ip, S., et al. (2019). Robust microfluidic technology and new lipid composition for fabrication of curcumin-loaded liposomes: effect on the anticancer activity and safety of Cisplatin. Mol. Pharm. 16, 3957–3967. doi:10.1021/acs.molpharmaceut.9b00583

PubMed Abstract | CrossRef Full Text | Google Scholar

Hashim, G. M., Shahgolzari, M., Hefferon, K., Yavari, A., and Venkataraman, S. (2024). Plant-Derived anti-cancer therapeutics and biopharmaceuticals. Bioeng. (Basel) 12, 7. doi:10.3390/bioengineering12010007

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoenders, R., Ghelman, R., Portella, C., Simmons, S., Locke, A., Cramer, H., et al. (2024). A review of the WHO strategy on traditional, complementary, and integrative medicine from the perspective of academic consortia for integrative medicine and health. Front. Med. (Lausanne) 11, 1395698. doi:10.3389/fmed.2024.1395698

PubMed Abstract | CrossRef Full Text | Google Scholar

Hsu, C.-Y., Pallathadka, H., Gupta, J., Ma, H., Al-Shukri, H. H. K., Kareem, A. K., et al. (2024). Berberine and berberine nanoformulations in cancer therapy: focusing on lung cancer. Phytother. Res. 38, 4336–4350. doi:10.1002/ptr.8255

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, X., Zhang, J., Deng, L., Hu, H., Hu, J., and Zheng, G. (2021). Galactose-modified PH-Sensitive niosomes for controlled release and hepatocellular carcinoma target delivery of tanshinone IIA. AAPS PharmSciTech 22, 96. doi:10.1208/s12249-021-01973-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Hui, Y., Liu, Y., Yang, G., Weng, Y., Hou, F., Wang, X., et al. (2025). Critical role of nanomaterial mechanical properties in drug delivery, nanovaccines and beyond. Adv. Mater. 37, e2413779. doi:10.1002/adma.202413779

PubMed Abstract | CrossRef Full Text | Google Scholar

Hwang, Y.-H., Shepherd, S. J., Kim, D., Mukalel, A. J., Mitchell, M. J., Issadore, D. A., et al. (2025). Robust, scalable microfluidic manufacturing of RNA-lipid nanoparticles using immobilized antifouling lubricant coating. ACS Nano 19, 1090–1102. doi:10.1021/acsnano.4c12965

PubMed Abstract | CrossRef Full Text | Google Scholar

Ijod, G., Nawawi, N. I. M., Anwar, F., Rahim, M. H. A., Ismail-Fitry, M. R., Adzahan, N. M., et al. (2024). Recent microencapsulation trends for enhancing the stability and functionality of anthocyanins: a review. Food Sci. Biotechnol. 33, 2673–2698. doi:10.1007/s10068-024-01603-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ingale, O. S., Pravin, B. P., Pawase, P. A., Shams, R., Dash, K. K., Bashir, O., et al. (2025). Enhancing bioactive stability and applications: microencapsulation in fruit and vegetable waste valorization. Discov. Food 5, 148. doi:10.1007/s44187-025-00412-8

CrossRef Full Text | Google Scholar

Jacob, S., Kather, F. S., Boddu, S. H. S., Rao, R., and Nair, A. B. (2025). Vesicular carriers for phytochemical delivery: a comprehensive review of techniques and applications. Pharmaceutics 17, 464. doi:10.3390/pharmaceutics17040464

PubMed Abstract | CrossRef Full Text | Google Scholar

Jaradat, E., Weaver, E., Meziane, A., and Lamprou, D. A. (2022). Microfluidic paclitaxel-loaded lipid nanoparticle formulations for chemotherapy. Int. J. Pharm. 628, 122320. doi:10.1016/j.ijpharm.2022.122320

PubMed Abstract | CrossRef Full Text | Google Scholar

Jităreanu, A., Trifan, A., Vieriu, M., Caba, I.-C., Mârțu, I., and Agoroaei, L. (2022). Current trends in toxicity assessment of herbal medicines: a narrative review. Processes 11, 83. doi:10.3390/pr11010083

CrossRef Full Text | Google Scholar

Kalia, A., and Kaur, G. (2020). “Nano-delivery carriers for enhanced bioavailability of antitumor phytochemicals,” in Pharmacotherapeutic botanicals for cancer chemoprevention. Editors M. Kumar, A. Sharma, and P. Kumar (Singapore: Springer Singapore), 189–196. doi:10.1007/978-981-15-5999-0_8

CrossRef Full Text | Google Scholar

Kang, X., Zheng, Z., Liu, Z., Wang, H., Zhao, Y., Zhang, W., et al. (2018). Liposomal codelivery of doxorubicin and andrographolide inhibits breast cancer growth and metastasis. Mol. Pharm. 15, 1618–1626. doi:10.1021/acs.molpharmaceut.7b01164

PubMed Abstract | CrossRef Full Text | Google Scholar

Kazi, J., Sen, R., Ganguly, S., Jha, T., Ganguly, S., and Chatterjee Debnath, M. (2020). Folate decorated epigallocatechin-3-gallate (EGCG) loaded PLGA nanoparticles; in-vitro and in-vivo targeting efficacy against MDA-MB-231 tumor xenograft. Int. J. Pharm. 585, 119449. doi:10.1016/j.ijpharm.2020.119449

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y., Lee Chung, B., Ma, M., Mulder, W. J. M., Fayad, Z. A., Farokhzad, O. C., et al. (2012). Mass production and size control of lipid-polymer hybrid nanoparticles through controlled microvortices. Nano Lett. 12, 3587–3591. doi:10.1021/nl301253v

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, S. K., Kim, Y. H., Park, S., and Cho, S.-W. (2021). Organoid engineering with microfluidics and biomaterials for liver, lung disease, and cancer modeling. Acta Biomater. 132, 37–51. doi:10.1016/j.actbio.2021.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J., Li, S., Zhang, S., and Wang, J. (2022). Plant-derived exosome-like nanoparticles and their therapeutic activities. Asian J. Pharm. Sci. 17, 53–69. doi:10.1016/j.ajps.2021.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J. H., Dareowolabi, B. O., Thiruvengadam, R., and Moon, E.-Y. (2024). Application of nanotechnology and phytochemicals in anticancer therapy. Pharmaceutics 16, 1169. doi:10.3390/pharmaceutics16091169

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, G., Virmani, T., Chhabra, V., Virmani, R., Pathak, K., Akhtar, M. S., et al. (2024). Transforming cancer treatment: the potential of nanonutraceuticals. Int. J. Pharm. 667, 124919. doi:10.1016/j.ijpharm.2024.124919

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuo, Y.-C., Wang, L.-J., and Rajesh, R. (2019). Targeting human brain cancer stem cells by curcumin-loaded nanoparticles grafted with anti-aldehyde dehydrogenase and sialic acid: colocalization of ALDH and CD44. Mater. Sci. Eng. C Mater. Biol. Appl. 102, 362–372. doi:10.1016/j.msec.2019.04.065

PubMed Abstract | CrossRef Full Text | Google Scholar

Lekhak, N., and Bhattarai, H. K. (2024). Phytochemicals in cancer chemoprevention: preclinical and clinical studies. Cancer control. 31, 10732748241302902. doi:10.1177/10732748241302902

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Q., Xiong, Y., Ji, C., and Yan, Z. (2019). The application of nanotechnology in the codelivery of active constituents of plants and chemotherapeutics for overcoming physiological barriers during antitumor treatment. Biomed. Res. Int. 2019, 9083068. doi:10.1155/2019/9083068

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., Qu, X., Qian, W., Song, Y., Wang, C., and Liu, W. (2022a). Andrographolide-loaded solid lipid nanoparticles enhance anti-cancer activity against head and neck cancer and precancerous cells. Oral Dis. 28, 142–149. doi:10.1111/odi.13751

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Zhou, X., Liu, J., Zhang, J., Feng, Y., Wang, F., et al. (2022b). Liposomal Co-delivery of PD-L1 siRNA/Anemoside B4 for enhanced combinational immunotherapeutic effect. ACS Appl. Mater. Interfaces 14, 28439–28454. doi:10.1021/acsami.2c01123

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Holman, J. B., Shi, Z., Qiu, B., and Ding, W. (2023a). On-chip modeling of tumor evolution: advances, challenges and opportunities. Mater. Today Bio 21, 100724. doi:10.1016/j.mtbio.2023.100724

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Xu, Y., Zhang, J., Zhang, Y., He, W., Ju, J., et al. (2023b). The effect of resveratrol, curcumin and quercetin combination on immuno-suppression of tumor microenvironment for breast tumor-bearing mice. Sci. Rep. 13, 13278. doi:10.1038/s41598-023-39279-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, S., Yang, B., Ye, L., Hu, S., Li, B., Yang, Y., et al. (2025). Multistage microfluidic assisted Co-Delivery platform for dual-agent facile sequential encapsulation. Eur. J. Pharm. Biopharm. 207, 114616. doi:10.1016/j.ejpb.2024.114616

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, J.-M., Swami, A., Gilson, L. M., Chopra, S., Choi, S., Wu, J., et al. (2014). Ultra-high throughput synthesis of nanoparticles with homogeneous size distribution using a coaxial turbulent jet mixer. ACS Nano 8, 6056–6065. doi:10.1021/nn501371n

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, D., Zhang, H., Fontana, F., Hirvonen, J. T., and Santos, H. A. (2018). Current developments and applications of microfluidic technology toward clinical translation of nanomedicines. Adv. Drug Deliv. Rev. 128, 54–83. doi:10.1016/j.addr.2017.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Fang, J., Huang, S., Wu, X., Xie, X., Wang, J., et al. (2021). Tumor-on-a-chip: from bioinspired design to biomedical application. Microsyst. Nanoeng. 7, 50. doi:10.1038/s41378-021-00277-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S., Li, L., and Ren, D. (2023). Anti-cancer potential of phytochemicals: the regulation of the epithelial-mesenchymal transition. Molecules 28, 5069. doi:10.3390/molecules28135069

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Wang, H., Chen, R., Song, Y., Wei, W., Baek, D., et al. (2025). Cancer-on-a-chip for precision cancer medicine. Lab. Chip 25, 3314–3347. doi:10.1039/d4lc01043d

PubMed Abstract | CrossRef Full Text | Google Scholar

Lv, L., Liu, C., Chen, C., Yu, X., Chen, G., Shi, Y., et al. (2016). Quercetin and doxorubicin co-encapsulated biotin receptor-targeting nanoparticles for minimizing drug resistance in breast cancer. Oncotarget 7, 32184–32199. doi:10.18632/oncotarget.8607

PubMed Abstract | CrossRef Full Text | Google Scholar

Ly, P.-D., Ly, K.-N., Phan, H.-L., Nguyen, H. H. T., Duong, V.-A., and Nguyen, H. V. (2024). Recent advances in surface decoration of nanoparticles in drug delivery. Front. Nanotechnol. 6, 1456939. doi:10.3389/fnano.2024.1456939

CrossRef Full Text | Google Scholar

Ma, C., Peng, Y., Li, H., and Chen, W. (2021a). Organ-on-a-Chip: a new paradigm for drug development. Trends Pharmacol. Sci. 42, 119–133. doi:10.1016/j.tips.2020.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, Y., Thurecht, K. J., and Coombes, A. G. A. (2021b). Development of enteric-coated, biphasic chitosan/HPMC microcapsules for colon-targeted delivery of anticancer drug-loaded nanoparticles. Int. J. Pharm. 607, 121026. doi:10.1016/j.ijpharm.2021.121026

PubMed Abstract | CrossRef Full Text | Google Scholar

Martins, A. M., Brito, A., Barbato, M. G., Felici, A., Reis, R. L., Pires, R. A., et al. (2023). Efficacy of molecular and nano-therapies on brain tumor models in microfluidic devices. Biomater. Adv. 144, 213227. doi:10.1016/j.bioadv.2022.213227

PubMed Abstract | CrossRef Full Text | Google Scholar

Matiyani, M., Rana, A., Pal, M., Rana, S., Melkani, A. B., and Sahoo, N. G. (2022). Polymer grafted magnetic graphene oxide as a potential nanocarrier for pH-responsive delivery of sparingly soluble quercetin against breast cancer cells. RSC Adv. 12, 2574–2588. doi:10.1039/d1ra05382e

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazumder, K., Aktar, A., Roy, P., Biswas, B., Hossain, M. E., Sarkar, K. K., et al. (2022). A review on mechanistic insight of plant derived anticancer bioactive phytocompounds and their structure activity relationship. Molecules 27, 3036. doi:10.3390/molecules27093036

PubMed Abstract | CrossRef Full Text | Google Scholar

Melim, C., Magalhães, M., Santos, A. C., Campos, E. J., and Cabral, C. (2022). Nanoparticles as phytochemical carriers for cancer treatment: news of the last decade. Expert Opin. Drug Deliv. 19, 179–197. doi:10.1080/17425247.2022.2041599

PubMed Abstract | CrossRef Full Text | Google Scholar

Mittal, S., Shah, S., Yadav, H. N., Ali, J., Gupta, M. M., and Baboota, S. (2023). Quality by design engineered, enhanced anticancer activity of temozolomide and resveratrol coloaded NLC and brain targeting via lactoferrin conjugation in treatment of glioblastoma. Eur. J. Pharm. Biopharm. 191, 175–188. doi:10.1016/j.ejpb.2023.08.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Mohaddish, M., Jilani, M., Rehman, U., Ramaiah, R., Hani, U., Gupta, G., et al. (2025). Recent advances in the clinical application of transferosomes for skin cancer management. Colloids Surf. B Biointerfaces 254, 114877. doi:10.1016/j.colsurfb.2025.114877

PubMed Abstract | CrossRef Full Text | Google Scholar

Motawea, A., Maria, S. N., Maria, D. N., Jablonski, M. M., and Ibrahim, M. M. (2024). Genistein transfersome-embedded topical delivery system for skin melanoma treatment: in vitro and ex vivo evaluations. Drug Deliv. 31, 2372277. doi:10.1080/10717544.2024.2372277

PubMed Abstract | CrossRef Full Text | Google Scholar

Mugale, M. N., Dev, K., More, B. S., Mishra, V. S., Washimkar, K. R., Singh, K., et al. (2024). A comprehensive review on preclinical safety and toxicity of medicinal plants. Clin. Complementary Med. Pharmacol. 4, 100129. doi:10.1016/j.ccmp.2024.100129

CrossRef Full Text | Google Scholar

Mülhopt, S., Diabaté, S., Dilger, M., Adelhelm, C., Anderlohr, C., Bergfeldt, T., et al. (2018). Characterization of nanoparticle batch-to-batch variability. Nanomater. (Basel) 8, 311. doi:10.3390/nano8050311

PubMed Abstract | CrossRef Full Text | Google Scholar

Naghib, S. M., Ahmadi, B., Mikaeeli Kangarshahi, B., and Mozafari, M. R. (2024). Chitosan-based smart stimuli-responsive nanoparticles for gene delivery and gene therapy: recent progresses on cancer therapy. Int. J. Biol. Macromol. 278, 134542. doi:10.1016/j.ijbiomac.2024.134542

PubMed Abstract | CrossRef Full Text | Google Scholar

Nashimoto, Y., Okada, R., Hanada, S., Arima, Y., Nishiyama, K., Miura, T., et al. (2020). Vascularized cancer on a chip: the effect of perfusion on growth and drug delivery of tumor spheroid. Biomaterials 229, 119547. doi:10.1016/j.biomaterials.2019.119547

PubMed Abstract | CrossRef Full Text | Google Scholar

Nasiri, R., Kurosu Jalil, M., Ibanez Gaspar, V., Flores Perez, A. S., Nguyen, H. T. M., Khan, S., et al. (2025a). A lung tumor-on-a-chip model recapitulates the effect of hypoxia on radiotherapy response and FDG-PET imaging. Lab. Chip 25, 4677–4691. doi:10.1039/d5lc00373c

PubMed Abstract | CrossRef Full Text | Google Scholar

Nasiri, R., Sankaranthi, A., and Pratx, G. (2025b). Organ-on-a-chip systems for modeling tumor and normal tissue microenvironments in radiotherapy research. Trends Biotechnol. doi:10.1016/j.tibtech.2025.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Naujokat, C., and McKee, D. L. (2021). The “big five” phytochemicals targeting cancer stem cells: Curcumin, EGCG, sulforaphane, resveratrol and genistein. Curr. Med. Chem. 28, 4321–4342. doi:10.2174/0929867327666200228110738

PubMed Abstract | CrossRef Full Text | Google Scholar

Nejati, B., Shahhosseini, R., Hajiabbasi, M., Ardabili, N. S., Baktash, K. B., Alivirdiloo, V., et al. (2025). Cancer-on-chip: a breakthrough organ-on-a-chip technology in cancer cell modeling. Med. Biol. Eng. Comput. 63, 321–337. doi:10.1007/s11517-024-03199-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Ngo, H., Amartumur, S., Tran, V. T. A., Tran, M., Diep, Y. N., Cho, H., et al. (2023). In vitro tumor models on chip and integrated microphysiological analysis platform (MAP) for life sciences and high-throughput drug screening. Biosens. (Basel) 13, 231. doi:10.3390/bios13020231

PubMed Abstract | CrossRef Full Text | Google Scholar

Nunziata, G., Borroni, A., and Rossi, F. (2025). Advanced microfluidic strategies for core-shell nanoparticles: the next-generation of polymeric and lipid-based drug nanocarriers. Chem. Eng. J. Adv. 22, 100759. doi:10.1016/j.ceja.2025.100759

CrossRef Full Text | Google Scholar

Padmavathy, N., Das Ghosh, L., Meka, S. R. K., and Chatterjee, K. (2018). Synthesis of a block copolymer exhibiting cell-responsive phytochemical release for cancer therapy. ACS Appl. Mater. Interfaces 10, 21816–21824. doi:10.1021/acsami.8b03521

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, Y., Mary Peter, R., Chou, P., Dave, P. D., Xu, J., Shanner, A., et al. (2025). Cancer-specific regulation of metabolic and epigenetic pathways by dietary phytochemicals. Pharm. Res. 42, 1443–1457. doi:10.1007/s11095-025-03898-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, H., Saravanakumar, G., Kim, J., Lim, J., and Kim, W. J. (2021). Tumor microenvironment sensitive nanocarriers for bioimaging and therapeutics. Adv. Healthc. Mater. 10, e2000834. doi:10.1002/adhm.202000834

PubMed Abstract | CrossRef Full Text | Google Scholar

Parvin, N., Aslam, M., Joo, S. W., and Mandal, T. K. (2025). Nano-phytomedicine: harnessing plant-derived phytochemicals in nanocarriers for targeted human health applications. Molecules 30, 3177. doi:10.3390/molecules30153177

PubMed Abstract | CrossRef Full Text | Google Scholar

Perez-Ruiz, A. G., Ganem, A., Olivares-Corichi, I. M., and García-Sánchez, J. R. (2018). Lecithin-chitosan-TPGS nanoparticles as nanocarriers of (-)-epicatechin enhanced its anticancer activity in breast cancer cells. RSC Adv. 8, 34773–34782. doi:10.1039/c8ra06327c

PubMed Abstract | CrossRef Full Text | Google Scholar

Pisani, S., Chiesa, E., Genta, I., Dorati, R., Gregorini, M., Grignano, M. A., et al. (2022). Liposome formulation and in vitro testing in non-physiological conditions addressed to Ex Vivo kidney perfusion. Int. J. Mol. Sci. 23, 7999. doi:10.3390/ijms23147999

PubMed Abstract | CrossRef Full Text | Google Scholar

Quan, N. V., Dang Xuan, T., and Teschke, R. (2020). Potential hepatotoxins found in herbal medicinal products: a systematic review. Int. J. Mol. Sci. 21, 5011. doi:10.3390/ijms21145011

PubMed Abstract | CrossRef Full Text | Google Scholar

Rao, J., Peng, T., Li, N., Wang, Y., Yan, C., Wang, K., et al. (2022). Nephrotoxicity induced by natural compounds from herbal medicines - a challenge for clinical application. Crit. Rev. Toxicol. 52, 757–778. doi:10.1080/10408444.2023.2168178

PubMed Abstract | CrossRef Full Text | Google Scholar

Roy, M., and Datta, A. (2019). “Molecular mechanisms of phytochemical actions in cancer,” in Cancer genetics and therapeutics: focus on phytochemicals (Singapore: Springer Singapore), 111–138. doi:10.1007/978-981-13-9471-3_5

CrossRef Full Text | Google Scholar

Sebastian, V. (2022). Toward continuous production of high-quality nanomaterials using microfluidics: nanoengineering the shape, structure and chemical composition. Nanoscale 14, 4411–4447. doi:10.1039/d1nr06342a

PubMed Abstract | CrossRef Full Text | Google Scholar

Selmi, M., Salek, A., Barboura, M., Njim, L., Trabelsi, A., Lahmar, A., et al. (2023). Thymoquinone-loaded lipid nanocapsules with promising anticancer activity for colorectal cancer. Nanoscale Adv. 5, 5390–5398. doi:10.1039/d3na00445g

PubMed Abstract | CrossRef Full Text | Google Scholar

Sha, A., Luo, Y., Xiao, W., He, J., Chen, X., Xiong, Z., et al. (2024). Plant-derived exosome-like nanoparticles: a comprehensive overview of their composition, biogenesis, isolation, and biological applications. Int. J. Mol. Sci. 25, 12092. doi:10.3390/ijms252212092

PubMed Abstract | CrossRef Full Text | Google Scholar

Shah, J., Patel, D., Rananavare, D., Hudson, D., Tran, M., Schloss, R., et al. (2025). Recent advancements in chitosan-based biomaterials for wound healing. J. Funct. Biomater. 16, 45. doi:10.3390/jfb16020045

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharifi, F., Yesil-Celiktas, O., Kazan, A., Maharjan, S., Saghazadeh, S., Firoozbakhsh, K., et al. (2020). A hepatocellular carcinoma–bone metastasis-on-a-chip model for studying thymoquinone-loaded anticancer nanoparticles. Bio-des. Manuf. 3, 189–202. doi:10.1007/s42242-020-00074-8

CrossRef Full Text | Google Scholar

Shepherd, S. J., Warzecha, C. C., Yadavali, S., El-Mayta, R., Alameh, M.-G., Wang, L., et al. (2021). Scalable mRNA and siRNA lipid nanoparticle production using a parallelized microfluidic device. Nano Lett. 21, 5671–5680. doi:10.1021/acs.nanolett.1c01353

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, Y., He, X., Wang, H., Dai, J., Fang, J., He, Y., et al. (2023). Construction of a novel blood brain barrier-glioma microfluidic chip model: applications in the evaluation of permeability and anti-glioma activity of traditional Chinese medicine components. Talanta 253, 123971. doi:10.1016/j.talanta.2022.123971

PubMed Abstract | CrossRef Full Text | Google Scholar

Shivani, G., Singh, G., Narwal, S., Chopra, B., and Dhingra, A. K. (2023). Quercetin-based nanoformulation: a potential approach for cancer treatment. Anticancer Agents Med. Chem. 23, 1983–2007. doi:10.2174/1871520623666230817101926

PubMed Abstract | CrossRef Full Text | Google Scholar

Shuvalov, O., Kirdeeva, Y., Daks, A., Fedorova, O., Parfenyev, S., Simon, H.-U., et al. (2023). Phytochemicals target multiple metabolic pathways in cancer. Antioxidants (Basel) 12, 2012. doi:10.3390/antiox12112012

PubMed Abstract | CrossRef Full Text | Google Scholar

Siavashy, S., Soltani, M., Ghorbani-Bidkorbeh, F., Fallah, N., Farnam, G., Mortazavi, S. A., et al. (2021). Microfluidic platform for synthesis and optimization of chitosan-coated magnetic nanoparticles in cisplatin delivery. Carbohydr. Polym. 265, 118027. doi:10.1016/j.carbpol.2021.118027

PubMed Abstract | CrossRef Full Text | Google Scholar

Situmorang, P. C., Ilyas, S., Nugraha, S. E., Syahputra, R. A., and Nik Abd Rahman, N. M. A. (2024). Prospects of compounds of herbal plants as anticancer agents: a comprehensive review from molecular pathways. Front. Pharmacol. 15, 1387866. doi:10.3389/fphar.2024.1387866

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramaniam, S., Selvaduray, K. R., and Radhakrishnan, A. K. (2019). Bioactive compounds: natural defense against cancer? Biomolecules 9, 758. doi:10.3390/biom9120758

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, M., Wu, Y., Zhou, Z., Liu, S., Mao, S., and Li, G. (2023). Co-delivery of EGCG and melittin with self-assembled fluoro-nanoparticles for enhanced cancer therapy. Aging (Albany NY) 15, 4875–4888. doi:10.18632/aging.204769

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, G., Wu, Y., Li, J., Yang, M., Xu, H., Li, Y., et al. (2025). Quercetin liposomes conjugated with hyaluronidase: an efficient drug delivery system to block pancreatic cancer. J. Control. Release 382, 113642. doi:10.1016/j.jconrel.2025.113642

PubMed Abstract | CrossRef Full Text | Google Scholar

Sunildutt, N., Parihar, P., Chethikkattuveli Salih, A. R., Lee, S. H., and Choi, K. H. (2023). Revolutionizing drug development: harnessing the potential of organ-on-chip technology for disease modeling and drug discovery. Front. Pharmacol. 14, 1139229. doi:10.3389/fphar.2023.1139229

PubMed Abstract | CrossRef Full Text | Google Scholar

Talebi, M., Shahbazi, K., Dakkali, M. S., Akbari, M., Almasi Ghale, R., Hashemi, S., et al. (2025). Phytosomes: a promising nanocarrier system for enhanced bioavailability and therapeutic efficacy of herbal products. Phytomedicine Plus 5, 100779. doi:10.1016/j.phyplu.2025.100779

CrossRef Full Text | Google Scholar

Tamatam, R., and Mohammed, A. (2024). Small molecule anticancer drugs approved during 2021-2022: synthesis and clinical applications. Eur. J. Med. Chem. 272, 116441. doi:10.1016/j.ejmech.2024.116441

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, C., Zheng, S., Liu, X., and Kamei, K.-I. (2022). Tumor-on-a-chip model for advancement of anti-cancer nano drug delivery system. J. Nanobiotechnology 20, 338. doi:10.1186/s12951-022-01552-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, Z., Fu, Y., Dang, Z., Guo, T., Li, W., and Zhang, J. (2025). Utilizing nanomaterials in microfluidic devices for disease detection and treatment. Nanomater. (Basel) 15, 434. doi:10.3390/nano15060434

PubMed Abstract | CrossRef Full Text | Google Scholar

Ukwubile, C. A., Malgwi, T. S., and Odu, C. E. (2025). Targeted delivery of Zingiber officinale roscoe extract-loaded chitosan nanoparticles for inhibition of multidrug-resistant escherichia coli-induced HCT-116 Colon cancer. Clin. Phytosci. 11, 6. doi:10.1186/s40816-025-00392-3

CrossRef Full Text | Google Scholar

Uzabakiriho, P. C., Jiajun, F., Nguchu, B. A., Iqbal, S., Manishimwe, C., and Shaw, P. (2025). Spheroid-on-a-chip platforms for tumor microenvironment and drug development. Adv. Mater. Technol. 10, 2401821. doi:10.1002/admt.202401821

CrossRef Full Text | Google Scholar

Vizovisek, M., Ristanovic, D., Menghini, S., Christiansen, M. G., and Schuerle, S. (2021). The tumor proteolytic landscape: a challenging frontier in cancer diagnosis and therapy. Int. J. Mol. Sci. 22, 2514. doi:10.3390/ijms22052514

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, M., Zhao, T., Liu, Y., Wang, Q., Xing, S., Li, L., et al. (2017). Ursolic acid liposomes with chitosan modification: promising antitumor drug delivery and efficacy. Mater. Sci. Eng. C Mater. Biol. Appl. 71, 1231–1240. doi:10.1016/j.msec.2016.11.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, N., Wang, J., Meng, X., Bao, Y., Wang, S., and Li, T. (2018). 3D microfluidic in vitro model and bioinformatics integration to study the effects of spatholobi caulis tannin in cervical cancer. Sci. Rep. 8, 12285. doi:10.1038/s41598-018-29848-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Yin, Q., Liu, C., Tang, Y., Sun, C., and Zhuang, J. (2021). Nanoformulations of ursolic acid: a modern natural anticancer molecule. Front. Pharmacol. 12, 706121. doi:10.3389/fphar.2021.706121

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, K., Shang, J., Tao, C., Huang, M., Wei, D., Yang, L., et al. (2024). Advancements in betulinic acid-loaded nanoformulations for enhanced anti-tumor therapy. Int. J. Nanomedicine 19, 14075–14103. doi:10.2147/IJN.S493489

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Li, S., Wang, Z., Kang, B., and Yan, H. (2025). The Co-Delivery of natural products and small RNAs for cancer therapy: a review. Molecules 30, 1495. doi:10.3390/molecules30071495

PubMed Abstract | CrossRef Full Text | Google Scholar

Weaver, E., O’Connor, E., Cole, D. K., Hooker, A., Uddin, S., and Lamprou, D. A. (2022). Microfluidic-mediated self-assembly of phospholipids for the delivery of biologic molecules. Int. J. Pharm. 611, 121347. doi:10.1016/j.ijpharm.2021.121347

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilson, K., Bala, A., Sindhu, R. K., Jeengar, M. K., Verma, I., Swami, R., et al. (2023). “Nanonutraceuticals and their applications,” in Nanotechnology and drug delivery: principles and applications (New York: Jenny Stanford Publishing), 577–610. doi:10.1201/9781003430407-14

CrossRef Full Text | Google Scholar

Wink, M. (2022). Current understanding of modes of action of multicomponent bioactive phytochemicals: potential for nutraceuticals and antimicrobials. Annu. Rev. Food Sci. Technol. 13, 337–359. doi:10.1146/annurev-food-052720-100326

PubMed Abstract | CrossRef Full Text | Google Scholar

Wongrakpanich, A., Bui Thi Thu, H., Sakchaisri, K., Taresco, V., Crucitti, V. C., Bunsupa, S., et al. (2024). 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. 95, 105610. doi:10.1016/j.jddst.2024.105610

CrossRef Full Text | Google Scholar

Wu, Q., Zhao, B., Sui, G., and Shi, J. (2021). Phytochemicals block glucose utilization and lipid synthesis to counteract metabolic reprogramming in cancer cells. Appl. Sci. 11, 1259. doi:10.3390/app11031259

CrossRef Full Text | Google Scholar

Xu, X., Liu, A., Bai, Y., Li, Y., Zhang, C., Cui, S., et al. (2018). Co-delivery of resveratrol and p53 gene via peptide cationic liposomal nanocarrier for the synergistic treatment of cervical cancer and breast cancer cells. J. Drug Deliv. Sci. Technol. 51, 746–753. doi:10.1016/j.jddst.2018.05.008

CrossRef Full Text | Google Scholar

Yang, Y., and Ling, W. (2025). Health benefits and future research of phytochemicals: a literature review. J. Nutr. 155, 87–101. doi:10.1016/j.tjnut.2024.11.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Z., Sun, N., Cheng, R., Zhao, C., Liu, J., and Tian, Z. (2017). Hybrid nanoparticles coated with hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. J. Mater. Chem. B Mater. Biol. Med. 5, 6762–6775. doi:10.1039/c7tb01510k

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, J., Liu, L., Sun, Q., and Shen, X. (2023). Direct cellular targets and anticancer mechanisms of the natural product oridonin. MedComm – Future Med. 2, e35. doi:10.1002/mef2.35

CrossRef Full Text | Google Scholar

Yin, L., Duan, W., Chen, Y., Chen, D., Wang, Y., Guo, S., et al. (2023). Biodegradable hydrogel from pectin and carboxymethyl cellulose with silibinin loading for lung tumor therapy. Int. J. Biol. Macromol. 243, 125128. doi:10.1016/j.ijbiomac.2023.125128

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, S., Zhang, L., Yang, Y., Wang, M., Liu, T., Ji, W., et al. (2024). Polydopamine-based resveratrol-hyaluronidase nanomedicine inhibited pancreatic cancer cell invasive phenotype in hyaluronic acid enrichment tumor sphere model. ACS Pharmacol. Transl. Sci. 7, 1013–1022. doi:10.1021/acsptsci.3c00304

PubMed Abstract | CrossRef Full Text | Google Scholar

Zandieh, M. A., Farahani, M. H., Daryab, M., Motahari, A., Gholami, S., Salmani, F., et al. (2023). Stimuli-responsive (nano)architectures for phytochemical delivery in cancer therapy. Biomed. Pharmacother. 166, 115283. doi:10.1016/j.biopha.2023.115283

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhai, J., Liu, Y., Ji, W., Huang, X., Wang, P., Li, Y., et al. (2024). Drug screening on digital microfluidics for cancer precision medicine. Nat. Commun. 15, 4363. doi:10.1038/s41467-024-48616-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Yang, J., Sun, R., Han, S., Yang, Z., and Teng, L. (2023). Microfluidics for nano-drug delivery systems: from fundamentals to industrialization. Acta Pharm. Sin. B 13, 3277–3299. doi:10.1016/j.apsb.2023.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: phytochemical nanosystems, cancer nanotherapy, tumor microenvironment, gene modulation, microfluidics, microphysiological systems, translational nanomedicine, SDG 3

Citation: Peñaherrera-Pazmiño AB, Criollo M and Gonzalez-Pastor R (2025) Phytochemical nanoencapsulation and microfluidics drive gene and tumor microenvironment modulation. Front. Pharmacol. 16:1694752. doi: 10.3389/fphar.2025.1694752

Received: 28 August 2025; Accepted: 16 September 2025;
Published: 29 September 2025.

Edited by:

Momir Mikov, University of Novi Sad, Serbia

Reviewed by:

Sara Baldassari, University of Genoa, Italy

Copyright © 2025 Peñaherrera-Pazmiño, Criollo and Gonzalez-Pastor. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Rebeca Gonzalez-Pastor, cmViZWNhLmdvbnphbGV6QHV0ZS5lZHUuZWM=

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.