Polymer lipid hybrid nanoparticles for phytochemical delivery: challenges, progress, and future prospects

Introduction

Phytochemicals are naturally occurring compounds found in plants (from the Greek word “phyton” meaning plant) . These bioactive compounds are responsible for the color, flavor, and aroma of plants, but more importantly, they contribute to the plant’s defense mechanisms against pathogens, pests, and environmental stresses. Beyond their role in plant biology, phytochemicals have gained significant attention for their potential health benefits in humans . Although they are not essential nutrients like vitamins and minerals, phytochemicals play a crucial role in maintaining health and preventing disease. These compounds are broadly categorized into several classes, including alkaloids, flavonoids, phenolic acids, terpenoids, and glycosides, each with distinct chemical structures and biological activities .

Phytochemicals have captured the interest of the scientific community and the pharmaceutical industry alike because of their extensive therapeutic potential. They function as antioxidants, anti-inflammatory agents, anticancer compounds, and antimicrobials, offering a natural and multifaceted arsenal for combating a wide array of diseases . Despite their promising bioactivities, the clinical application of phytochemicals is often limited by several inherent drawbacks such as poor water solubility, low bioavailability, rapid metabolism, and instability under physiological conditions . These challenges necessitate the development of advanced delivery systems to harness the full potential of phytochemicals in therapeutic applications.

Polymer lipid hybrid nanoparticles (PLHNPs) represent an innovative class of delivery vehicles that combines the beneficial properties of both polymeric and lipid-based nanoparticles, thus offering a creative approach to enhance the delivery and efficacy of drugs/phytochemicals. The architecture of PLHNPs typically consists of a lipid core or shell enveloped by a polymer matrix, which can vary in its configuration depending on the specific requirements of the delivery system . The lipid components, often phospholipids, cholesterol, and surfactants are integral for solubilizing lipophilic drugs. The polymer component, which can include materials such as polyethylene glycol (PEG), poly(lactic-co-glycolic acid) (PLGA), polycaprolactone (PCL), and chitosan (CHS), provides structural integrity, controlled release properties, and protection against premature degradation . This hybrid structure improves the encapsulation efficiency of phytochemicals/drugs and enhances their solubility, stability, and bioavailability. Additionally, the physicochemical characteristics of PLHNPs can be tailored by varying the concentrations of polymers and lipids .

PLHNPs can address various challenges associated with phytochemical delivery. The benefits of PLHNPs include their small particle size, high encapsulation efficiency, enhanced stability, and improved dissolution in harsh gastrointestinal (GI) fluids. Following oral administration, PLHNPs demonstrate superior intestinal absorption and bioavailability, attributed to their enhanced stability and dissolution rate in GI fluids. Further, PLHNPs overcome limitations such as rapid metabolism and limited bioavailability by encapsulating phytochemicals within the hybrid matrices. This encapsulation enhances the stability of phytochemicals, extends their circulation time within the body, and enhances their therapeutic effectiveness . Additionally, PLHNPs possess the capability to encapsulate and co-deliver two drugs with distinct physicochemical characteristics to a targeted site and show synergistic therapeutic efficacy. In PLHNPs, the lipophilic drugs are encapsulated within the polymeric core, while hydrophilic drugs are entrapped in the lipid shell. PLHNPs demonstrate relatively greater loading capacity for lipophilic compounds than other nanoparticle systems .

Moreover, the surface modification of PLHNPs with targeting ligands, such as antibodies, peptides, or aptamers, has been explored to improve the selective delivery of drugs/phytochemicals to specific tissues or cells. A site-specific targeting approach enhances the therapeutic efficacy of phytochemicals and reduces systemic toxicity. In addition to enhancing solubility and targeting, PLHNPs offer controlled release properties that are crucial for maintaining therapeutic drug levels over extended periods of time . By adjusting the polymer composition and lipid matrix, researchers can fine-tune the release kinetics of phytochemicals, ensuring sustained therapeutic effects. This controlled release mechanism is particularly advantageous for chronic conditions requiring long-term treatment, such as cancer, cardiovascular diseases, and neurodegenerative disorders .

This review aims to discuss the ability of PLHNPs to improve the therapeutic delivery of phytochemicals for biomedical applications. In this review, we discuss the obstacles in the conventional delivery of phytochemicals, types of PLHNPs, different phytochemical-loaded PLHNPs for improved phytochemical delivery, challenges in clinical translation of PLHNPs, and future perspectives.

Review Obstacles in the conventional delivery of phytochemicals

The conventional delivery of phytochemicals faces numerous challenges that limit its clinical application and therapeutic efficacy. These challenges arise from the physicochemical characteristics of phytochemicals, as well as from the physiological barriers they encounter in the body. One major challenge is poor water solubility. Many phytochemicals are hydrophobic and show poor water solubility, which significantly restricts their absorption in the gastrointestinal tract (GIT) when administered orally. This low solubility leads to low bioavailability, resulting in sub-therapeutic levels of the phytochemical at the target site . Another significant issue is low bioavailability. Bioavailability refers to the proportion of an administered substance that enters the bloodstream and is available for therapeutic action. Phytochemicals often exhibit low bioavailability due to poor solubility, rapid metabolism, and degradation in the physiological fluids. This necessitates higher doses to achieve therapeutic effects, which may increase the risk of side effects and toxicity . Chemical instability is also a critical challenge. Phytochemicals can be chemically unstable and degrade under physiological conditions, such as varying pH levels, temperature, and enzymatic activity. Degradation reduces the effective concentration of the phytochemical, diminishing its therapeutic potential . Rapid metabolism and clearance further complicate phytochemical delivery. Phytochemicals are often rapidly metabolized by liver enzymes and cleared from the body through renal or biliary excretion, resulting in short plasma half-lives and requiring frequent dosing to maintain effective therapeutic levels. This rapid clearance reduces the duration of action, making it challenging to achieve sustained therapeutic effects . Poor permeability and penetration are additional obstacles. Phytochemicals may have difficulties crossing biological membranes, such as the intestinal epithelium or the blood–brain barrier, because of their molecular size, polarity, or lipophilicity. Poor permeability limits the ability of phytochemicals to reach intracellular or central nervous system targets, reducing therapeutic efficacy in tissues that are difficult to access. Variability in absorption is another significant issue. The absorption of phytochemicals can be influenced by various factors, including food intake, gut microbiota, and individual genetic differences. This variability leads to inconsistent therapeutic outcomes among different individuals, and food–drug interactions can further complicate dosing regimens and efficacy . Further, conventional delivery methods often lack specificity, resulting in the distribution of phytochemicals throughout the body rather than targeting specific tissues or cells. Non-specific distribution increases the risk of off-target effects and systemic toxicity, reducing the concentration of the phytochemical at the desired site of action and decreasing therapeutic effectiveness . In addition, conventional delivery systems often cannot provide controlled or sustained release of phytochemicals, leading to fluctuating plasma levels. These fluctuations can result in suboptimal therapeutic effects and increased side effects. Lack of controlled release is particularly problematic for chronic conditions that require consistent drug exposure over extended periods of time . Therefore, there is a need for innovative delivery systems that improve solubility, stability, bioavailability, and targeted delivery, while also providing controlled release. PLHNPs represent a promising solution to many of these challenges, offering a versatile platform to maximize the therapeutic potential of phytochemicals.

Overview of PLHNPs for phytochemical delivery

PLHNPs represent an innovative solution for the effective delivery of phytochemicals by harnessing the combined advantages of both polymeric and lipidic nanoparticles. PLHNPs represent higher encapsulation efficiency, ensuring that a higher proportion of the phytochemical payload is successfully encapsulated within the hybrid matrix of nanoparticles . This improved encapsulation leads to enhanced bioavailability, meaning more of the phytochemical can reach the systemic circulation, resulting in greater therapeutic efficacy. Structurally, PLHNPs maintain their integrity through a unique hybrid architecture. They typically consist of a lipid core or shell surrounded by a polymer matrix. The lipid components, including phospholipids, cholesterol, and surfactants, play a crucial role in solubilizing lipophilic phytochemicals and facilitating interactions with biological membranes. The polymers provide structural stability, controlled release properties, and protection against premature degradation . PLHNPs address various challenges associated with phytochemical delivery. They overcome limitations such as poor solubility, rapid metabolism, and limited bioavailability by encapsulating phytochemicals within lipid–polymer hybrid matrices. This encapsulation enhances the stability of phytochemicals, prolongs their circulation time in the body, and enhances their therapeutic effectiveness . Additionally, surface engineering of PLHNPs with different ligands facilitates specific delivery of drug/phytochemicals to desired tissues or cells, reduces their adverse effects, and improves their therapeutic efficacy . The development of PLHNPs for phytochemical delivery holds significant promise across various biomedical applications. PLHNPs can be utilized in cancer therapy, cardiovascular disease management, neurodegenerative disorder treatment, and other areas of medicine where phytochemicals show therapeutic potential.

Types of PLHNPs

Generally, PLHNPs are classified based on the arrangement of polymers and lipids within the hybrid system. In the hybrid structures, polymers enhance overall particle stability and modulate the release of encapsulated drugs from the hybrid matrix. However, lipids provide more space for drug encapsulation and biocompatibility of the system. Therefore, the advancement in the LPHNPs yields better and prolonged therapeutic efficacy. Different types and structural advantages of PLHNPs are illustrated in Figure 1 and are discussed in detail as follows.

[2190-4286-15-118-1]

Figure 1: Different types of PLHNPs. Figure 1 was created in BioRender. Rizwanullah, M. (2024) BioRender.com/c14a571. This content is not subject to CC BY 4.0.

Polymer core–lipid shell hybrid nanoparticles

As the name suggests, polymer core–lipid shell hybrid nanoparticles are composed of a polymer core that is covered by mono/bilayers of a lipoidal shell. The polymeric core significantly enhances the stability of the outer lipoidal shell. The biodegradable polymeric core with a stable outer lipoidal shell makes these PLHNPs an excellent nanocarrier for therapeutic drug delivery and the treatment of various diseases. The amphiphilicity of biodegradable polymers and lipids promotes the encapsulation of both lipophilic as well as hydrophilic chemotherapeutic drugs within the hybrid system. During the development of LPHNPs, different physicochemical characteristics such as size, loading capacity, charge, solubility, release, and colloidal stability can be modulated by modification in the polymer/lipid ratio .

Monolithic PLHNPs

Monolithic PLHNPs are the simplest among PLHNPs; they are simply mixed nanosystems of polymer/copolymer and lipids with the help of surfactants. In this system, the lipids are scattered in a polymeric/copolymeric matrix . Monolithic PLHNP systems are very similar to colloidal polymeric nanocarriers. In these nanocarriers, phospholipids help to form a carrier-like structure, which is an integral part of the system. In addition, the modification of lipoidal layers with a PEG chain provides flexibility to the nanocarrier. The ratio of the polymer and lipid can easily be adjusted to modulate the physicochemical characteristics of the nanocarrier and can reduce systemic toxicity .

Core–shell type hollow PLHNPs

The core–shell type hollow PLHNPs comprise an inner hollow positively charged lipidic core, a polymeric layer in the middle, and an outer PEG lipoidal layer . The inner hollow core of the system is filled with water/or buffer. Because of the positive charge, the lipids in the inner core encapsulate the drug more efficiently compared to PLHNPs with a polymeric core. In addition, because of the outer lipoidal PEG layer, these nanocarriers escape the uptake by macrophages and enhance the stability of the biological fluids . During the development of these nanocarriers, the concentration of cationic lipids for the inner core, density of the PEG chain on the outer layer, and molecular weight of the polymers are adjusted to modulate their physicochemical characteristics .

Polymer-caged liposomes

As the name suggests, the structural arrangement of these nanocarriers involves the surface coating of liposomes with biodegradable polymers/copolymers. The surface modification not only imparts surface functionality to the nanocarrier but also enhances its therapeutic efficacy by site-specific targeting and controlled release of the encapsulated drugs. Among all PLHNPs, these hybrid nanocarriers show the highest stability in the biological fluids and stimuli-responsive release of encapsulated drugs. In addition, the polymeric cage protects the drug from the harsh environment, and encapsulated drugs can be released under specific biological conditions. Further, the polymer coating provides better colloidal stability, sustained drug release, and high loading capacity to the hybrid nanocarriers .

Cell membrane-camouflaged PLHNPs

PLHNPs have been coated with cell membranes (e.g., erythrocytes) to develop membrane-camouflaged PLHNPs. These hybrid nanocarriers are also called biomimetic hybrid nanocarriers because their surface chemistry mimics natural cell membranes . The PLHNPs are coated with cell membranes via the extrusion technique. The coating of PLHNPs with red blood cells yields a natural vehicle for drug delivery, and these nanocarriers can easily escape the uptake by macrophages. In this system, the drugs are encapsulated in the lipophilic polymeric core, and the lipids in the outer natural membrane enhance the sustained release of drugs. With the development of these hybrid nanocarriers, the biological barriers in therapeutic drug delivery can be easily overcome. These hybrid nanocarriers show prolonged half-life and stability in biological systems, thereby enhancing therapeutic efficacy .

Surface modification of PLHNPs

Surface modification of PLHNPs involves the functionalization of the outer surface of nanoparticles with specific molecules or ligands to impart desired properties or functionalities. This process plays a crucial role in enhancing the targeting, stability, and therapeutic efficacy of PLHNPs for drug/phytochemical delivery. Surface modification of PLHNPs begins with the preparation of the nanoparticles themselves. PLHNPs are typically synthesized using techniques such as solvent evaporation, nanoprecipitation, double emulsion, or solvent injection, resulting in the formation of nanoparticles with a lipid–polymer hybrid structure . Once the PLHNPs are synthesized, their surface can be modified through various strategies. Figure 2 illustrates a comparison between conventional PLHNPs and surface-modified PLHNPs.

[2190-4286-15-118-2]

Figure 2: (A) Conventional PLHNPs and (B) surface-functionalized PLHNPs. Figure 2 was created in BioRender. Rizwanullah, M. (2024) BioRender.com/b56n651. This content is not subject to CC BY 4.0.

The most common approach is the attachment of targeting ligands onto the PLHNPs’ surface. These ligands can include antibodies, peptides, aptamers, or small molecules that specifically bind to receptors overexpressed on the surface of target cells or tissues. The conjugation of targeting ligands to the surface of PLHNPs enables specific delivery of drug/phytochemicals to desired sites within the body, such as tumor cells in cancer therapy. This targeted delivery can increase the therapeutic efficacy of drugs while reducing side effects by minimizing off-target effects . For instance, Garg et al. fabricated fucose ligand-decorated PLHNPs for the co-delivery of methotrexate and aceclofenac to achieve targeted and synergistic therapeutic efficacy against breast cancer (BC) . The scheme for the development of these ligand-decorated PLHNPs is depicted in Figure 3. The findings suggested that the targeted PLHNPs significantly improved uptake in BC cells by receptor-mediated endocytosis when compared with non-targeted PLHNPs. A pharmacodynamic study of targeted PLHNPs in DMBA-induced BC-bearing female BALB/c mice showed that the targeted PLHNPs yielded significantly enhanced and synergistic therapeutic efficacy and showed much better tumor inhibition and improved survival rate than the non-targeted PLHNPs.

[2190-4286-15-118-3]

Figure 3: (A) Schematic illustrating the synthesis of DSPE-PEG(2000)-NH2-fucose; Step 1: oxidation of ʟ-fucose, step 2: formation of Schiff’s base, and step 3: reduction to a secondary amine. (B) Development of PLHNPs with fucose-decorated phospholipids. Figure 3 was reprinted with permission from . Copyright 2017 American Chemical Society. This content is not subject to CC BY 4.0.

Another surface modification strategy involves the addition of stealth coatings, such as PEG, to the nanoparticle surface. A schematic representation for the development of PEGylated PLHNPs is depicted in Figure 4A. PEGylation involves attaching PEG chains to the surface of the nanoparticles. This modification provides several advantages. PEGylation increases the stability of the PLHNPs in biological fluids by preventing aggregation and reducing protein adsorption. It also extends the circulation time of nanoparticles in the bloodstream by reducing immune system recognition and clearance. This leads to improved bioavailability and allows for sustained release of the encapsulated therapeutic agents . The advantages of PEGylated PLHNPs include enhanced biocompatibility and reduced immunogenicity. The PEG layer creates a hydrophilic barrier around the nanoparticles, which minimizes the interaction with blood components and immune cells. This reduces the risk of immune reactions and increases the half-life of the nanoparticles in the body. Additionally, PEGylation can improve the solubility of hydrophobic drugs, facilitating their delivery. The hybrid structure of these nanoparticles combines the benefits of both polymeric and lipid-based systems, offering controlled drug release and efficient encapsulation of various therapeutic agents. Overall, PEGylated PLHNPs offer a versatile and effective platform for various therapeutic applications .

[2190-4286-15-118-4]

Figure 4: (A) Schematic illustration of the development of PEGylated PLHNPs. Figure 4A was created in BioRender. Rizwanullah, M. (2024) BioRender.com/p31e321. This content is not subject to CC BY 4.0.; (B) Scheme for the development of ligand-decorated redox-sensitive PLHNPs. Figure 4B was adapted from (© 2019 J. Wang et al., published by Elsevier Masson SAS, distributed under the terms of the Creative Commons Attribution Noncommercial Noderivatives 4.0 International License, https://creativecommons.org/licenses/by-nc-nd/4.0/). This content is not subject to CC BY 4.0.

Surface modification of PLHNPs can also involve the incorporation of stimuli-responsive moieties onto the nanoparticle surface. These moieties enable the nanoparticles to respond to specific stimuli, such as pH changes, temperature shifts, or enzyme activity, thereby triggering controlled drug release at the target site . The advantages of stimuli-responsive nanoparticles include targeted and controlled drug delivery. By responding to specific stimuli, these nanoparticles can release their drug payload at the desired site of action, such as a tumor or inflamed tissue, while minimizing drug release in healthy tissues. This targeted release improves therapeutic outcomes and reduces side effects. Additionally, the ability to fine-tune the release profile based on external or internal stimuli allows for customized treatment regimens . The hybrid structure of PLHNPs offers the combined benefits of both polymeric and lipid-based carriers, providing stability, biocompatibility, and efficient drug encapsulation. Applications of stimuli-responsive PLHNPs are vast and impactful. In cancer therapy, they are used to deliver chemotherapeutic agents specifically to tumor sites, where the acidic microenvironment or specific enzymes can trigger drug release . For instance, Wang et al. fabricated transferrin ligand-decorated redox-sensitive PLHNPs to achieve improved therapeutic efficacy of afatinib against non-small cell lung cancer . The complete scheme for the development of ligand-decorated redox-sensitive PLHNPs is represented in Figure 4B. The developed targeted PLHNPs exhibit excellent physicochemical characteristics and showed GSH-triggered drug release and significantly improved cytotoxicity against PC-9 cells in comparison to the non-targeted PLHNPs. In vivo studies suggested that the developed redox-sensitive targeted PLHNPs represented a much higher accumulation of the drug in the tumor due to targetability and higher drug release at tumor pH. Consequently, the redox-sensitive targeted PLHNPs showed much better therapeutic efficacy compared to non-targeted and non-redox-sensitive PLHNPs. Overall, surface modification of PLHNPs is a versatile strategy for enhancing nanoparticles’ targeting, stability, and therapeutic efficacy for drug/phytochemical delivery.

PLHNPs for the delivery of single phytochemicals

PLHNPs represent a versatile and effective platform for the delivery of single phytochemicals, offering enhanced stability, bioavailability, and targeted delivery. As previously discussed, many phytochemicals are lipophilic. Figure 5 illustrates the chemical structures of the various phytochemicals examined in this paper. After oral administration, phytochemicals exhibit poor solubility in the GI milieu and limited absorption from the GIT, resulting in low oral bioavailability and, consequently, reduced bioactivity. Recent advances regarding PLHNPs encapsulating single phytochemicals to overcome the limitations and better management of different diseases are discussed below. Pharmaceutical attributes with major outcomes are summarized in Table 1 at the end of this section.

[2190-4286-15-118-5]

Figure 5: Image illustrating the chemical structures of different phytochemicals reviewed in this paper for therapeutic delivery with PLHNPs for the treatment of different diseases.

Table 1: Summary of pharmaceutical properties with major findings of different phytochemical encapsulated PLHNPs.

Encapsulated Phytochemical PLHNPs type Pharmaceutical attributes Ref.   PS (nm) PDI ZP (mV) EE (%) Major outcomes curcumin polymer core–lipid shell 184 – −29.3 53.2 Outstanding pharmaceutical attributes with excellent hemocompatibility. Notably higher cellular uptake in MCF-7 cells and a substantial decrease in the IC50 value compared to the free drug curcumin monolithic 216.6 ± 4.7 0.205 ± 0.02 −0.23 ± 0.12 96.0 ± 0.6   Surface modification with RGD peptide significantly improves the cellular uptake in HUVEC cells. Significantly higher anticancer therapeutic potential against B16 cells than the pure compound. Much higher in vivo antitumor activity in B16 tumor-bearing female BALB/c mice on intraperitoneal injection than the pure compound.   quercetin polymer core–lipid shell 339 ± 1.6 0.1 ± 0.02 −32.6 ± 0.51 78 ± 5.5   The developed PLHNPs effectively adhere to the scalp and show significantly higher follicular uptake. Significantly higher regrowth of hair in SD rats bearing testosterone-induced alopecia.   quercetin monolithic 110.6 0.237 −31.9 96.22   Enhanced cellular accumulation and antiproliferative activity against P388 cells. 3.75-fold increased bioavailability after oral ingestion in SD rats. Superior therapeutic efficacy in DBA/2 mice bearing P388 cells-induced ascitic leukemia.   thymoquinone monolithic 179.63 ± 4.77 0.21 ± 0.01 +26.52 ± 2.21 85.49 ± 3.73   Outstanding mucoadhesive properties and stability in GI environments. Higher intestinal permeation and 4.74-fold increased bioavailability after oral administration compared to free compound. Markedly higher cytotoxicity against MCF-7 and MDA-MB-231 cells.   thymoquinone monolithic <350 <0.3 >−19 88.42 ± 2.58   Higher skin permeation and skin retention compared to the conventional formulation and non-irritant. Markedly enhanced antiproliferative activity against MCF-7 and MBD-MB-231 cells compared to the native compound.   resveratrol polymer core–lipid shell 375 ± 13 – −22 ± 1.6 76 ± 4.2   Better stability, biocompatibility, and controlled release profile. Markedly enhanced antiproliferative activity against MCF-7 cancer cells compared to the native compound.   resveratrol polymer-caged liposome 212–225 0.122–0.172 4.15–14.77 74–77   Excellent mucoadhesive characteristics and sustained drug release profiles. Stronger anti-oxidative and anti-inflammatory activity compared to the native compound.   apigenin monolithic 125.73 ± 5.57 0.18 ± 0.02 −26.71 ± 1.93 77.43 ± 3.62   Exceptional colloidal stability and controlled drug release properties. Almost halved the IC50 value against MCF-7 and MDA-MB-231 cells.   apigenin monolithic 234.8 ± 12.28 0.11 ± 0.04 ‒5.15 ± 0.70 55.18 ± 3.61   Enhanced apoptosis and cell cycle arrest against HCT-116 cells. Much higher cytotoxicity compared to the native compound.   isoliquiritigenin polymer core–lipid shell 73.24 ± 1.83 – −17.21 ± 0.90 96.75 ± 1.41   Better stability in the harsh GI environment. Superior therapeutic efficacy against BC in both in vitro and in vivo studies. Higher absorption and 3.8-fold increased oral bioavailability.   isoliquiritigenin polymer core–lipid shell 137.2 ± 2.6 – −34.21 ± 1.23 90.8 ± 1.5   Modification of PLHNPs with iRGD significantly enhances their cellular internalization and antiproliferative activity against MDA-MB-231 cells. Significantly enhanced tumor-growth inhibition potential in a 4T1-bearing mouse xenograft.   hydroxycamptothecin polymer core–lipid shell 249.8 ± 24.4 0.289 −25.6 ± 3.8 65.93 ± 0.52   Surface functionalization and PEG-conjugation did not modulate the drug release profiles. Ligand decoration with RGD-peptide significantly improved the cellular internalization potential and antiproliferative activity against MDA-MB-435s cells.   hydroxycamptothecin polymer core–lipid shell 226.4 0.236 – –   Much higher dose- and time-dependent cytotoxicity against both MCF-7 and HepG2 cells. 3-fold increased bioavailability after intravenous administration in SD rats compared to native compounds. Excellent therapeutic efficacy without severe side effects in murine LLC-GFP-luc lung cancer-bearing Kunming mice.   psoralen polymer core–lipid shell 93.44 ± 2.39 0.257 ± 0.02 −27.63 ± 0.31 76.9   Excellent pharmaceutical attributes with controlled drug release characteristics. Higher tumor inhibition rate in MCF-7 tumor-bearing BALB/c female mice than native compound.   ursolic acid polymer core–lipid shell 145.1 ± 2.6 0.1 ± 0.01 −42 ± 1.2 –   Excellent serum stability and long-term stability. Much higher cellular internalization and cytotoxicity against AsPC-1 and BxPC-3 cells.   ursolic acid polymer-caged liposome 135.4 ± 0.636 <0.3 +7.8 94.3   Better stability and controlled release characteristics. Much higher accumulation in tumor and tumor growth inhibition in U14 tumor-bearing female CD-1 mice.   baicalin monolithic 184.3 0.177 −19.8 90.12   Stronger cytotoxic potential against HCT-116 cells than free drug. Rapid absorption and almost 2-fold improved bioavailability after oral intake in male Albino rats.   silymarin polymer core–lipid shell 286.5 ± 23.8 0.23 ± 0.008 45.3 ± 8.9

留言 (0)

沒有登入
gif