BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Pharmacol Ther. Dec 5, 2025; 16(4): 111502
Published online Dec 5, 2025. doi: 10.4292/wjgpt.v16.i4.111502
Targeted nanoliposomal nutrient delivery for human health
Joseph Mercola, Midwestern University, Downers Grove, IL 60515, United States
ORCID number: Joseph Mercola (0009-0008-5237-8503).
Author contributions: Mercola J was the sole author responsible for study conception and design, data acquisition and interpretation, manuscript preparation and revision, final approval of the version to be published, and agrees to be accountable for the integrity of the work in all respects.
Conflict-of-interest statement: I have no conflicts to disclose.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Joseph Mercola, Researcher, Midwestern University, 555 31st Street, Downers Grove, IL 60515, United States. drm@mercola.com
Received: July 1, 2025
Revised: July 16, 2025
Accepted: October 10, 2025
Published online: December 5, 2025
Processing time: 157 Days and 14.3 Hours

Abstract

Conventional nutritional supplements frequently demonstrate limited clinical effectiveness due to the harsh milieu of the gastrointestinal tract, inefficient trans-epithelial transport, and rapid systemic clearance. Nanoliposomal delivery platforms - lipid bilayer vesicles on the nanometer scale - have attracted attention as an adaptive strategy to shield sensitive nutrients, navigate biological barriers, and deliver payloads directly to target tissues or even sub-cellular organelles. Despite a growing body of literature, a consolidated appraisal of design principles, targeting modalities, and translational hurdles is still needed to guide future nutraceutical innovation. We aim to: (1) Summarize the physicochemical foundations of nanoliposomal nutrient carriers; (2) Delineate state-of-the-art approaches for organ-specific and organelle-specific targeting, with particular emphasis on renal and mitochondrial delivery; (3) Evaluate current evidence supporting therapeutic benefits in cardiometabolic, neuroprotective, and renal-repair contexts; and (4) Map unresolved challenges - including manufacturing scale-up, cost, and regulatory oversight - to inform a roadmap for clinical translation. A systematic literature search was performed across PubMed, Web of Science, and Scopus through May 2025 using Boolean combinations of “nanoliposome”, “nutrient”, “targeted delivery”, “bioavailability”, and organ-specific terms (e.g., “kidney”, “mitochondria”). Primary research articles, systematic reviews, and relevant meta-analyses written in English were included. Data were extracted on liposomal composition, particle size, surface modifications (e.g., polyethylene glycol, ligand conjugation), in vitro and in vivo bio-distribution, efficacy outcomes, and safety profiles. Key design variables were mapped against reported biological performance to identify convergent principles. Sixty-four original studies and twenty-one reviews met inclusion criteria. Encapsulation within phosphatidylcholine-rich bilayers consistently enhanced nutrient stability in simulated gastric fluid and improved Caco-2 trans-epithelial transport two-fold to ten-fold compared with free compound controls. Ligand-mediated strategies - such as folate, lactoferrin, or peptide conjugation - achieved organ-specific accumulation, with kidney-directed liposomes demonstrating up to a four-fold increase in renal cortex uptake. Mitochondrial targeting using amphipathic peptides (e.g., SS-31) or triphenylphosphonium moieties delivered antioxidant nutrients to the organelle, restoring mitochondrial membrane potential and reducing reactive oxygen species (ROS) in preclinical cardiomyopathy and neurodegeneration models. Endosomal escape was most effectively triggered by fusogenic lipids (e.g., dioleoylphosphatidylethanolamine) or pH-responsive polymers. PEGylation prolonged circulation half-life by 3-6 hours but elicited anti-polyethylene glycol antibodies in approximately one-quarter of recipients; emerging natural sterol-mimetic or collagen-mimetic coatings showed comparable stealth behavior with superior biodegradability. Scalability remains limited: Only three studies reported pilot-scale (> 10 L) batches with Good Manufacturing Practice-compliant reproducibility. Targeted nanoliposomal systems substantially improve nutrient stability, absorption, and tissue specificity, offering a credible route to transform supplement efficacy for cardiometabolic, renal, and neuroprotective indications. Optimization of lipid composition, escape mechanisms, and biocompatible surface chemistries can further enhance therapeutic indices. Nonetheless, industrial-scale manufacturing, cost containment, and immunogenicity mitigation remain critical obstacles. Addressing these gaps through standardized characterization protocols, head-to-head clinical trials, and biomaterial innovation will be essential to unlock the full potential of nanoliposomal nutraceuticals in routine healthcare practice.

Key Words: Nanoliposomal delivery; Nutrient bioavailability; Targeted supplementation; Mitochondrial targeting; Receptor mediated endocytosis; Endosomal escape; Oral lymphatic transport; Polyethylene glycol limitations; Coenzyme Q10; Precision nutrition

Core Tip: By enclosing nutrients in nanoscale liposomes, this review shows how bioavailability leaps past gastric destruction, intestinal mucus, and first-pass metabolism while programmable surfaces steer payloads directly to kidneys, brain, or even mitochondria via ligands such as SS-31. The authors detail pH-responsive, fusogenic designs that escape endosomes, contrast natural lipid shells with immunogenic PEGylation, and map translational paths from coenzyme Q10 to heavy-metal chelation. Together, targeted nanoliposomal delivery promises drug-level precision nutrition without drug-level costs, redefining supplement science.



INTRODUCTION

Oral nutritional supplements often fail to deliver their full therapeutic potential due to poor solubility, degradation in the gastrointestinal (GI) tract, and inefficient uptake into circulation. Even nutrients that reach the bloodstream may be diluted systemically or rapidly cleared, limiting clinical impact[1].

Nanoliposomal carriers - lipid bilayer vesicles on the nanometer scale - offer a promising solution. These systems shield vulnerable compounds from degradation, improve absorption, and enable tissue-specific targeting. Several are already commercialized or entering clinical use, including liposomal formulations of vitamin C, vitamin D, iron, and antioxidant phytochemicals. Human pharmacokinetic studies consistently show improved maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) over conventional forms; in one trial, liposomal vitamin C increased plasma concentrations by 27% over standard ascorbate[2]. A six-month study in chronic kidney disease patients found that liposomal iron doubled transferrin saturation response rates without GI side effects[3].

Despite these advances, most nanoliposomal nutraceuticals remain confined to the supplement aisle. Five interlocking challenges limit broader clinical adoption: (1) Manufacturing inconsistency[4]; (2) Poor scalability and high cost[5,6]; (3) Oxidative and thermal instability[2]; (4) Immunogenicity linked to polyethylene glycol (PEG) coatings[7,8]; and (5) A fragmented regulatory framework[9].

Addressing these issues requires coordinated innovations. Emerging technologies - including continuous-flow microfluidics[6], zwitterionic stealth coatings[10], lyophilization with smart protectants[11], ligand-guided targeting[12], and inline quality analytics - offer a path toward more reliable, effective, and clinically viable delivery systems. Integrating even a subset of these solutions could transform liposomal nutrients into precision therapeutic platforms.

LIMITATIONS OF CONVENTIONAL SUPPLEMENT DELIVERY

Despite the convenience and non-invasive nature of oral administration, its efficacy is hampered by several physiological and physicochemical constraints.

Poor solubility and dissolution

A wide range of bioactive compounds possess limited aqueous solubility, hindering dissolution in the GI tract’s aqueous environment and subsequent absorption[13]. Such insoluble entities are often excreted unabsorbed, abrogating their therapeutic utility.

Degradation in the GI tract

The GI environment poses a hostile environment for nutrient stability, with gastric acidity capable of denaturing or inactivating labile compounds. Conventional formulations typically lack protective mechanisms to counteract these degradative processes[14].

Intestinal absorption barriers

The intestinal epithelium enforces stringent selectivity over nutrient uptake via transcellular or paracellular routes, both tightly regulated[15]. Large or hydrophilic molecules face challenges penetrating lipid-rich membranes, exacerbated by efflux transporters (e.g., P-glycoprotein) and metabolic enzymes that expel or degrade xenobiotics[16]. Additionally, a viscous mucus layer enveloping the epithelium obstructs diffusion of hydrophobic or particulate matter, further curtailing absorption[17]. Conventional supplements generally lack strategies to traverse this barrier or exploit specific uptake pathways.

First-pass metabolism

Nutrients absorbed from the intestine traverse the portal vein to the liver, where metabolic enzymes markedly diminish systemic availability[18].

Systemic dilution and off-target distribution

Following absorption, nutrients are distributed indiscriminately, diluting their concentration at intended therapeutic sites and potentially eliciting off-target effects[19].

Interindividual variability

Absorption efficiency exhibits significant interindividual variation, influenced by differences in GI pH, enzymatic profiles, microbiome composition, transit time, and genetic factors[20].

These constraints often compel the use of elevated doses or frequent administration to achieve therapeutic efficacy, strategies that may prove impractical or pose safety risks (Table 1)[1-11].

Table 1 Comparative analysis of conventional oral supplements and nanoliposomal delivery systems.
Aspect
Conventional oral supplements
Nanoliposomal delivery systems
Gastric stabilityLow: Vulnerable to acidic pH and enzymatic degradationHigh: Encapsulation within phospholipid bilayers confers protection against gastric acid and enzymatic hydrolysis
Intestinal absorptionLimited: Constrained by poor solubility and epithelial permeabilityEnhanced: Improved solubility and interaction with enterocyte uptake mechanisms (e.g., endocytosis)
First-pass metabolismPronounced: Hepatic metabolism via portal vein reduces bioactivityAttenuated: Lymphatic transport via chylomicrons partially circumvents hepatic first-pass metabolism
Systemic distributionNon-specific: Diffuse dilution across systemic tissuesTargeted: Surface ligands enable receptor-mediated delivery to specific tissues
BioavailabilitySuboptimal: High doses required to achieve therapeutic levelsSuperior: Increased efficiency permits lower effective doses
Therapeutic indexVariable: Elevated doses may precipitate gastrointestinal adverse effectsOptimized: Targeted delivery enhances efficacy while minimizing off-target toxicity
PRINCIPLES OF NANOLIPOSOMAL DELIVERY SYSTEMS

Nanoliposomal delivery systems encapsulate nutrients within nanoscale liposomes to enhance stability, bioavailability, and tissue-specific targeting. These systems have emerged as a transformative approach to optimize the efficacy of nutraceuticals and dietary supplements[21]. Liposomes are microscopic vesicles comprising one or more phospholipid bilayers enclosing an aqueous core and leverage their amphiphilic properties - featuring hydrophilic and hydrophobic domains - to encapsulate diverse bioactive compounds. Hydrophilic nutrients reside within the aqueous interior, whereas hydrophobic or lipophilic agents integrate into the lipid bilayers[22].

This structural versatility enables liposomes to shield vulnerable nutrients from degradation, such as protecting vitamins from oxidative damage or gastric acidity, and facilitates absorption by fusing with biological membranes or undergoing cellular uptake[23]. Empirical evidence demonstrates that liposomal encapsulation markedly enhances nutrient bioavailability; for instance, liposomal vitamin C exhibits superior absorption and an extended plasma half-life compared to non-encapsulated ascorbic acid[24].

Beyond enhancing systemic bioavailability, nanoliposomal systems can be engineered for precision delivery. Surface modification with ligands - such as antibodies, peptides, or small molecules - enables receptor-mediated binding and endocytosis, directing liposomes to specific tissues or cell types[25]. This targeted strategy, well-established in pharmaceutical applications, is increasingly adapted for nutrient delivery to optimize therapeutic outcomes. For example, liposomes functionalized with renal-specific ligands can deliver antioxidants or vitamins to kidney cells, minimizing off-target distribution[26]. Similarly, to mitigate cellular oxidative stress, liposomes bearing mitochondrial-targeting peptides can transport mitochondrial co-factors, like coenzyme Q10 (CoQ10), directly to the mitochondria to increase cellular energy production.

This review initially examines the limitations of conventional supplement delivery, underscoring the necessity for advanced systems. Subsequently, it delineates the principles of nanoliposomal delivery, encompassing liposome composition, classification, and their capacity to surmount traditional barriers. The discussion then addresses physiological obstacles - detailing how nanoliposomes withstand gastric acidity, penetrate intestinal mucus, and enhance transcellular uptake, including leveraging lymphatic transport to circumvent first-pass metabolism[27]. Organ-specific targeting strategies are explored, with emphasis on receptor-mediated endocytosis (RME) - a selective cellular uptake process in which ligands bind to membrane receptors, triggering vesicle formation and internalization into endosomes[28]. This pathway enables efficient nanomolar-level uptake of nutrients, hormones, and engineered nanocarriers, and is commonly used in nanomedicine delivery systems. Dysregulation has been shown to contribute to disorders such as familial hypercholesterolaemia, cancer, and neurodegeneration. Mitochondrial targeting approaches are also discussed, including the use of SS-31 peptides and triphenylphosphonium (TPP+) moieties to direct intracellular routing to mitochondria.

The challenge of endosomal escape is an important consideration in the design of nanoliposomes targeting subcellular compartments, such as the mitochondria. Following endocytosis, these liposomes must evade degradation within the endosomal compartment, which would otherwise result in the premature release of their payload into the intravesicular space of the endosome, preventing delivery to the intended cytosolic or subcellular destinations. To address this, strategies such as the incorporation of fusogenic lipids - specialized lipids that adopt non-bilayer structures to destabilize membranes and trigger fusion - or pH-responsive architectures are assessed to facilitate endosomal membrane disruption. This enables efficient release of the encapsulated cargo into the cytosol for downstream delivery to specific intracellular organelles[29].

LIPOSOME COMPOSITION AND STRUCTURE

Liposomes are typically composed of phospholipids - the same building blocks that make up cell membranes. A phospholipid has a hydrophilic head (often containing a phosphate group) and two hydrophobic fatty acid tails. In water, phospholipids can self-assemble into bilayer membranes, where the hydrophobic tails face inward (away from water) and the hydrophilic heads face outward (toward the surrounding water). When such bilayers form closed spherical structures, they create an inner aqueous compartment. A unilamellar liposome has a single lipid bilayer surrounding an aqueous core, while multilamellar vesicles have concentric multiple bilayers (like an onion). Nanoliposomes generally refer to small liposomes in the size range of roughly 50-300 nm in diameter, which are often unilamellar[30], with a characteristic size of 100-200 nm which optimizes their utility in therapeutic delivery systems.

The composition of the lipid bilayer can be tailored to achieve certain properties: (1) Phosphatidylcholine (PC) from soy or egg yolk is commonly used and forms stable bilayers; (2) Cholesterol is often incorporated to modulate membrane fluidity and stability, when egg yolk serves as a substrate, cholesterol is naturally integrated into the liposomal structure, filling gaps between phospholipid tails to enhance bilayer integrity, reduce permeability, and minimize content leakage; and (3) Charge-inducing lipids: Lipids can be neutral, anionic (negative charge), or cationic (positive charge). Cationic lipids can facilitate binding to negatively charged cell membranes but might be more prone to clearance by the immune system. Anionic or neutral lipids are generally less immunogenic. The choice of charge can also affect interactions with the mucus layer and cellular uptake.

A RIGOROUS EVALUATION OF PEG-CONJUGATION NANOLIPOSOMAL DESIGN AND NATURAL ALTERNATIVES

The precise targeting of subcellular compartments, such as mitochondria, necessitates meticulous engineering of nanoliposomal physicochemical properties, extending beyond strategies for endosomal escape. The lipid bilayer composition fundamentally governs vesicle stability, cellular interactions, and in vivo disposition, dictating both the release kinetics of encapsulated payloads and the nanoliposome’s biological fate[31]. By modulating lipid constituents - including PC, cholesterol, and ionizable lipids - researchers can fine-tune membrane fluidity, surface charge, and interactions with physiological barriers, optimizing therapeutic efficacy[32]. To mitigate premature systemic clearance and degradation, surface functionalization with PEG-conjugated lipids - i.e., PEGylation, a process that forms a hydrated steric shield that reduces immune recognition and renal clearance[33] - is frequently employed to enhance pharmacokinetic longevity[34]. This modification warrants a comprehensive assessment of its merits juxtaposed against inherent limitations in therapeutic contexts.

PEGylation entails the covalent conjugation of PEG, a synthetic polyether polymer composed of repeating ethylene glycol units, to nanoliposomal lipids via chemical linkers such as N-hydroxysuccinimide esters[35]. This process generates a hydrophilic corona that augments colloidal stability, aqueous solubility, and circulatory half-life by abrogating enzymatic degradation, plasma protein opsonization, and uptake by the mononuclear phagocyte system (MPS)[36]. Within nanoliposomal platforms, PEGylation has emerged as a cornerstone of pharmaceutical delivery, notably in nutrient-directed applications, safeguarding payloads against GI degradation and facilitating tissue penetration[37]. Such attributes underscore its widespread adoption in drug and nutrient delivery systems.

Nevertheless, PEGylation’s synthetic provenance - derived from petroleum-based ethylene oxide polymerization - renders it devoid of natural analogs within biological or ecological systems[38]. Its biodegradability is markedly limited, particularly for high-molecular-weight variants (> 20000 Da), resulting in persistent accumulation within hepatic and splenic tissues due to negligible enzymatic catabolism[39]. The conjugation process, reliant on organic solvents and non-physiological conditions, starkly contrasts with the spontaneous, affinity-driven interactions of endogenous receptor-ligand complexes[40]. These characteristics challenge its suitability for advanced therapeutic paradigms.

The liabilities of PEGylation further complicate its application in health optimization strategies. Iterative exposure induces immunogenicity, with anti-PEG antibodies detected in approximately 25% of recipients, hastening clearance and precipitating hypersensitivity reactions[39]. Prolonged tissue retention elicits toxicological concerns, evidenced by histopathological alterations in preclinical models. Moreover, the steric hindrance imposed by PEG impedes cellular internalization, necessitating intricate, cleavable architectures that compromise its operational simplicity[41]. Such drawbacks diverge from the seamless integration of natural ligands into physiological cascades, which circumvent these risks.

In the context of nanoliposomal nutrient delivery to osseous tissue, PEGylation is contraindicated due to its artificial composition, attendant health hazards, and discordance with biomimetic therapeutic objectives. Its environmental burden, originating from fossil fuel-derived synthesis, coupled with regulatory complexities - potentially categorizing PEGylated formulations as pharmaceuticals - further undermines its utility[42]. Conversely, natural alternatives exhibit superior biocompatibility, biodegradability, and economic viability, evading immunogenicity and bioaccumulation while efficiently engaging bone-specific integrins[43]. Consequently, PEGylation’s synthetic underpinnings and associated limitations render it less favorable than biologically inspired approaches.

Structurally, liposomes exemplify bimodal carrier systems, capable of encapsulating hydrophilic payloads within their aqueous core and hydrophobic entities within the lipid bilayer[44]. This dual-compartment architecture underpins their versatility in delivering diverse therapeutic agents, highlighting the need for strategic compositional design to maximize efficacy while minimizing reliance on synthetic modifications like PEGylation.

Advantages of nanoscale size

Formulating liposomes at the nanoscale confers distinct physicochemical advantages that enhance their utility in nutrient delivery.

Mucosal penetration and cellular uptake: Nanoliposomes, typically below 200 nm in diameter, exhibit enhanced penetration through the intestinal mucus layer, facilitating intimate contact with the epithelial barrier[45]. Particles of this size are preferentially internalized by microfold (M) cells within Peyer’s patches via transcytosis or by enterocytes through endocytic mechanisms[46]. This size-dependent uptake improves the efficiency of payload delivery across the GI tract[47].

Surface area optimization: The high surface area-to-volume ratio of nanoliposomes amplifies interactions with biological membranes, facilitating fusion or adhesion processes critical for cellular uptake[48]. This expansive surface also provides sufficient real estate for conjugating targeting ligands, enabling precise receptor-mediated delivery if required[49].

Tissue distribution via paracellular or vascular pathways: Nanoparticles, including nanoliposomes, can exploit paracellular gaps or compromised vascular integrity to distribute into tissues[50]. In oncology, this is exemplified by the enhanced permeability and retention effect, where nanoparticles accumulate in tumor microenvironments due to leaky vasculature[51]. In nutritional contexts, analogous mechanisms may facilitate uptake in inflamed or metabolically stressed tissues, though this remains speculative and warrants further investigation[51].

Modulation of digestion kinetics: The nanoscale size influences liposomal breakdown and payload release kinetics. Smaller liposomes are more readily processed by enterocytes into chylomicron-like structures, promoting lymphatic transport and altering bioavailability profiles[52].

MECHANISMS ENHANCING ABSORPTION AND BIOAVAILABILITY

Nanoliposomes augment nutrient absorption and bioavailability through several well-documented mechanistic pathways.

Membrane fusion

The phospholipid composition of nanoliposomes mirrors that of biological membranes, enabling fusion with enterocyte plasma membranes and direct cytosolic deposition of payloads, bypassing reliance on transporter proteins[53].

Endocytic uptake

Intestinal epithelial cells internalize nanoliposomes via endocytosis, with M cells in Peyer’s patches specializing in particle sampling and transcytosis to underlying lymphoid tissues or systemic circulation[54]. Dendrit cells and enterocytes also exhibit nanoparticle uptake, a principle leveraged in oral vaccine development and potentially extensible to nutrient delivery. Post-internalization, transcytosis facilitates payload release into the basolateral compartment[55].

Lymphatic transport

Liposomal lipids align with the physiological absorption of dietary lipids via the lymphatic system. Following enterocyte uptake, nanoliposomes are incorporated into chylomicron-like particles and secreted into the lymph, bypassing hepatic first-pass metabolism[27]. This route, culminating in systemic entry via the thoracic duct, significantly enhances bioavailability[56], with studies showing increased lymphatic partitioning of liposomal payloads compared to free compounds[27].

Sustained release

Nanoliposomal encapsulation induces a depot effect, enabling gradual payload release as vesicles interact with cellular surfaces or degrade enzymatically. For instance, liposomal vitamin C sustains elevated plasma concentrations longer than free forms, exemplifying prolonged pharmacokinetic profiles[24].

Receptor-mediated targeting

Surface conjugation with ligands, such as folate, enables selective binding to intestinal receptors, enhancing uptake by receptor-rich enterocytes[57]. In drug delivery, folate-decorated liposomes increase oral bioavailability via folate RME, a principle potentially translatable to nutrient payloads[58].

FABRICATION AND FUNCTIONAL MODIFICATION OF NANOLIPOSOMAL DELIVERY SYSTEMS

The preparation of nanoliposomes employs sophisticated methodologies - such as thin-film hydration, microfluidization, and membrane extrusion - that enable precise regulation of vesicle size and lamellarity[48]. These techniques facilitate the engineering of nanoscale liposomes with uniform dimensions and defined bilayer structures, optimizing their physicochemical properties for therapeutic applications[31]. Post-fabrication, nanoliposomes can undergo surface functionalization to enhance their specificity and responsiveness, tailoring their performance in nutrient delivery.

Surface modification with targeting ligands

Nanoliposomal surfaces can be conjugated with an array of targeting moieties to achieve receptor-mediated specificity[49]. These include vitamins such as folate and cobalamin (vitamin B12), which engage their respective receptors on cellular membranes[57], and peptides targeting integrins or other cell-surface receptors. Additionally, monoclonal antibodies, antibody fragments, aptamers (short oligonucleotides with high target affinity), and small-molecule ligands can be anchored to the lipid bilayer, conferring precision in cellular recognition. While organ-specific targeting is addressed separately, this principle of ligand conjugation universally enhances liposomal versatility across diverse delivery contexts[59].

Stimuli-responsive architectures

Nanoliposomes can be engineered with stimuli-responsive functionalities to orchestrate controlled payload release in response to environmental cues, including pH gradients, enzymatic activity, or thermal variations. For instance, liposomes formulated with pH-sensitive lipids maintain structural integrity at physiological pH (approximately 7.4, as in blood or intestinal lumen) but destabilize at endosomal pH (approximately 5.0), facilitating endosomal escape and cytosolic delivery[60]. Alternatively, enzyme-responsive designs exploit protease-rich microenvironments - such as those enriched with matrix metalloproteinases in neoplastic tissues or specific bacterial hydrolases in the gut - to cleave peptide linkers, triggering payload liberation. These advanced features, often incorporating specialized lipids or polymeric components, represent a frontier of ongoing investigation in liposomal technology[58].

Integration of biocompatibility and nanoscale advantages

Nanoliposomal delivery systems synergistically combine the inherent biocompatibility and multifunctionality of liposomes with the physicochemical benefits of nanoscale engineering[61]. This integration addresses critical challenges in oral nutrient delivery, including payload protection from GI degradation, navigation through mucosal and epithelial barriers, enhancement of transepithelial absorption, and selective targeting of cellular or tissue niches[45]. Empirical evidence substantiates these capabilities. A 2020 investigation demonstrated that liposomal encapsulation of ascorbic acid yielded superior bioavailability compared to its non-liposomal counterpart, attributed to enhanced protection against gastric degradation and improved intestinal transport[62].

OVERCOMING GI OBSTACLES WITH NANOLIPOSOMAL SYSTEMS

The efficacious delivery of nutrients to systemic circulation and target tissues via oral administration necessitates that nanoliposomal systems surmount multiple physiological barriers that typically compromise supplement bioavailability. This section delineates these barriers and elucidates the strategies employed to enhance nanoliposomal performance in nutrient transport.

Stability in the gastric environment

The gastric milieu, characterized by a pH range of 1.0-3.5 and the presence of pepsin, imposes a formidable degradative challenge to unprotected nutrients and conventional delivery matrices[63]. Nanoliposomes mitigate this barrier by encapsulating payloads within a lipid bilayer, conferring robust protection against acidic hydrolysis and enzymatic degradation[31]. This bilayer, composed of phospholipids, exhibits greater resilience to pH fluctuations compared to the tertiary conformations of proteins or the chemical stability of acid-labile vitamins[23].

Empirical evidence demonstrates that liposomal encapsulation significantly enhances the preservation of labile compounds under gastric conditions, with studies reporting improved survival of probiotics - a paradigm applicable to nutrient cargos. To further augment gastric stability, nanoliposomes can be functionalized with pH-resistant polymeric coatings, such as alginate or chitosan, which maintain integrity in acidic environments and dissociate at the elevated pH of the intestinal lumen[64]. These coatings serve as an additional shield, preventing premature bilayer fusion or payload release.

Lipid composition can also be tailored to optimize release kinetics, employing gel-phase lipids that remain stable at gastric temperature (approximately 37 °C) and pH, transitioning to a liquid-crystalline state under intestinal conditions to facilitate controlled liberation. Collectively, these strategies ensure that nanoliposomes, with a gastric transit time typically ranging from 1-2 hours in the presence of food or shorter when fasting, deliver a substantially greater proportion of intact payload to the intestinal absorptive site compared to unprotected formulations[45].

Penetration of the intestinal mucus barrier

The small intestine is enveloped by a viscoelastic mucus layer, rich in mucin glycoproteins, which functions as a selective barrier, impeding the transit of pathogens and macromolecules to the underlying epithelium[65]. For nutrient delivery, this mucus poses a significant obstacle, potentially entrapping nanoparticles and limiting epithelial access[46]. Nanoliposomes can be engineered to overcome this barrier through precise physicochemical modifications.

Surface charge and hydrophilicity: The mucus layer bears a net negative charge due to sialic acid and sulfated residues, influencing nanoparticle interactions[66]. Cationic nanoliposomes may exhibit electrostatic adhesion to mucus, potentially reducing penetration, whereas neutral or mildly anionic surfaces facilitate transit. A subtle positive charge can, however, promote initial mucoadhesion prior to deeper penetration[67].

Mucolytic augmentation: Advanced formulations may incorporate mucolytic agents, such as N-acetylcysteine, which cleave disulfide bonds within mucin, locally reducing viscosity to enhance nanoparticle passage. Although more prevalent in pulmonary delivery, this approach is cautiously explored in oral contexts due to potential disruption of the gut’s protective mucus layer[13].

Mucoadhesion vs penetration balance: Nanoliposomal design confronts a strategic dichotomy: Mucoadhesive properties, achieved with coatings like chitosan, prolong intestinal residence, elevating local concentration gradients and potentially enhancing absorption, while mucus-penetrating configurations expedite epithelial contact[68]. Chitosan-functionalized liposomes demonstrate extended retention and increased peptide uptake, yet excessive mucoadhesion risks entrapment within superficial mucus layers[69]. Multilayered systems, featuring an inner penetrating core and a sheddable mucoadhesive outer shell, offer a hybrid solution, optimizing initial adhesion and subsequent penetration[70].

Enhanced transepithelial nutrient delivery via nanoliposomal systems

Upon reaching the intestinal epithelium, nanoliposomal systems employ several pathways to enhance nutrient absorption, navigating both transcellular and paracellular routes, with the former predominating due to stringent tight junction constraints limiting paracellular transit to particles below 1 nm[45]. The mechanisms underpinning transcellular uptake are delineated as follows.

Enterocyte-mediated absorption: Nanoliposomes facilitate uptake by enterocytes -the principal absorptive cells lining intestinal villi - via membrane fusion or endocytosis. This process may occur non-specifically or be augmented by RME triggered by surface-conjugated ligands, such as vitamins (e.g., folate) or peptides, which engage specific enterocyte receptors[57]. For instance, glycoprotein ligands targeting lectin receptors on enterocytes have been demonstrated to mediate nanoparticle transcytosis, enhancing payload delivery across the epithelium[46].

M cell transcytosis: Located within Peyer’s patches, M cells specialize in transcytosing particulate matter to subjacent immune cells within gut-associated lymphoid tissue (GALT)[54]. Nanoliposomes can exploit this pathway, with targeting moieties - such as lectins or monoclonal antibodies - enhancing M cell-specific uptake. While this route is advantageous for immunomodulatory applications, such as oral vaccines, its utility for systemic nutrient delivery may be secondary to enterocyte-mediated absorption, though lymphatic access remains feasible[55].

Tight junction modulation: Certain nanoliposomal formulations incorporate permeation enhancers, such as chitosan, which transiently disrupt tight junction integrity by interacting with occludin and zonula occludens-1 proteins, thereby facilitating paracellular transport. Chitosan-coated liposomes widen intercellular gaps, augmenting the flux of small molecular payloads across the epithelium[71].

Lipid absorption pathways: Nanoliposomes leverage endogenous lipid uptake mechanisms, whereby bile salt-mediated disintegration generates mixed micelles or fragmented vesicles that integrate into enterocyte lipid processing pathways[52]. Subsequent assembly into chylomicron-like lipoproteins enhances lymphatic delivery[72]. The inclusion of monoglycerides or fatty acids within the lipid bilayer further promotes emulsification and uptake, a strategy validated in self-emulsifying delivery systems[27].

Lymphatic transport and pharmacokinetic enhancement: A hallmark of nanoliposomal uptake is its propensity for lymphatic trafficking. Following enterocyte internalization, liposomal components or intact vesicles are reconstituted into chylomicron-like structures, which are secreted into the lymphatic vasculature, bypassing hepatic first-pass metabolism. Recent investigations, such as those examining cefotaxime encapsulation[73], report significantly elevated lymphatic concentrations compared to unencapsulated forms, underscoring enhanced systemic delivery. For nutrients, this pathway ensures greater bioavailability by minimizing immediate metabolic degradation, enabling direct entry into systemic circulation via the thoracic duct and rapid distribution to peripheral tissues[56].

This augmented absorption efficiency elevates the fraction of the administered dose absorbed (Fa) and the fraction escaping hepatic metabolism (Fh), culminating in a marked increase in overall bioavailability[31]. Pharmacokinetic analyses consistently demonstrate superior plasma AUC and peak concentrations (Cmax) for liposomal formulations relative to non-liposomal counterparts, reflecting enhanced nutrient delivery to target sites[24]. These attributes position nanoliposomes as a transformative platform for optimizing oral nutrient bioavailability.

Prolongation of systemic circulation and organ-specific targeting

Upon entry into systemic circulation or lymphatic vasculature following oral administration, nanoliposomes encounter the challenge of evading rapid clearance by the MPS, predominantly comprising hepatic Kupffer cells and splenic macrophages[31]. Premature sequestration by these immune cells precludes effective accumulation within target tissues, necessitating strategic modifications to enhance circulatory longevity[32].

Size optimization: Nanoliposomes exceeding 200 nm in diameter are more susceptible to splenic filtration and macrophage uptake, whereas those maintained below this threshold exhibit prolonged plasma residence. Smaller, uniformly sized vesicles with neutral surface properties minimize recognition by the MPS, thereby extending bioavailability[48].

Surface charge modulation: Cationic nanoliposomes are prone to opsonization by plasma proteins, accelerating clearance via phagocytic mechanisms. For systemic nutrient delivery, neutral or zwitterionic lipid compositions are preferred to reduce immune visibility, unless cationic surfaces are requisite for specific targeting functionalities[74].

Circulatory persistence vs local action trade-off: In scenarios prioritizing localized GI effects - such as targeting colonic epithelium or GALT - enhanced MPS uptake may be advantageous. However, for systemic nutrient optimization, strategies focus on sustaining circulatory presence to facilitate broader tissue distribution[75].

LIGAND-MEDIATED ORGAN-SPECIFIC TARGETING AND RME

A pivotal advancement in nanoliposomal delivery systems lies in their capacity for organ-specific or cell-specific targeting via surface functionalization with ligands, enabling precise payload localization while minimizing off-target dissemination[49]. This active targeting paradigm enhances nutrient uptake efficiency by leveraging cellular recognition mechanisms.

RME constitutes a highly specific cellular uptake process wherein ligands engage cognate receptors, triggering internalization via endocytic vesicles[57]. By conjugating nanoliposomes with ligands - such as peptides, antibodies, or bioactives - selective binding to receptors overexpressed on target cells facilitates efficient payload delivery. Upon receptor engagement, the ligand-receptor complex is enveloped within a vesicle, transporting the attached liposome into the intracellular compartment. This mechanism, extensively validated in oncological nanomedicine, is equally applicable to nutrient delivery[46].

Nanoliposomal systems can be engineered with surface-bound ligands to achieve organ-specific or cell-specific targeting, optimizing nutrient delivery to designated tissues while minimizing off-target accumulation[32]. This active targeting leverages RME or transcytosis, enhancing cellular uptake efficiency[57]. Several ligand classes have been explored.

Folate receptor targeting

Folate receptors, overexpressed on epithelial cells, renal proximal tubules, and various neoplastic tissues, serve as robust targets for ligand-mediated delivery. Folate-functionalized nanoliposomes enhance the uptake of encapsulated cargos and improve the oral bioavailability of hydrophilic compounds by engaging intestinal folate receptors. The small molecular size, chemical stability, and high receptor affinity of folic acid render it an efficacious targeting moiety for nutrient delivery to folate receptor-rich cellular populations[58].

Transferrin receptor targeting

Transferrin receptors, abundant on proliferating cells and the blood-brain barrier (BBB) endothelium, mediate the uptake of transferrin-bound iron[76]. Transferrin-transferrin or anti-transferrin receptor antibody-conjugated nanoliposomes demonstrate augmented delivery across the BBB via receptor-mediated transcytosis, a promising strategy for neuroprotective nutrient transport (e.g., antioxidants) that typically exhibit limited BBB permeability. This approach significantly enhances payload accumulation in cerebral and neoplastic tissues[77].

Peptide ligands

Short peptide sequences mimicking receptor-binding domains offer precise targeting capabilities. For instance, arginine-glycine-aspartic acid peptides engage integrin receptors, notably overexpressed on angiogenic vasculature, facilitating nanoparticle localization to vascularized tissues[50]. In renal targeting, the peptide CKGGRAKDC, identified via phage display, selectively homes to injured kidney vasculature, demonstrating utility in directing nanoliposomes to renal tissues. Similarly, Angiopep-2 targets low-density lipoprotein receptor-related protein 1, enabling transcytosis across the BBB for potential delivery of neuroprotective nutrients to neural tissues[76].

Vitamin-based ligands

Beyond folate, vitamins like biotin and cobalamin (vitamin B12) exploit receptor-mediated pathways. Biotinylated liposomes bind avidin or streptavidin with high affinity, though this is primarily utilized in experimental localization rather than in vivo tissue-specific targeting unless avidin is pre-localized. Cobalamin targets intrinsic factor-mediated uptake in the intestine and potentially other tissues expressing cubilin or transcobalamin receptors, offering a novel strategy for nutrient delivery to receptor-rich cells.

Aptamers

Nucleic acid aptamers, comprising DNA or RNA sequences with target-specific tertiary structures, function analogously to antibodies. Conjugated to nanoliposomes, aptamers have demonstrated efficacy in targeting immune cells or neoplastic tissues. In a nutraceutical context, aptamers binding insulin receptors or adipose-specific markers could direct metabolic nutrients to adipose or skeletal muscle cells, enhancing therapeutic outcomes in metabolic disorders[78].

Antibodies and derivatives

Monoclonal antibodies confer exceptional specificity by binding unique cellular antigens[49]. For example, liposomes conjugated with anti-Thy1.1 antibodies in rodent models exhibited a six-fold increase in glomerular deposition compared to untargeted counterparts, highlighting their renal targeting potential. Despite their large molecular weight (approximately 150 kDa), which may augment vesicle size, minimal antibody loading significantly alters biodistribution[31]. Smaller derivatives, such as Fab fragments, single-chain variable fragments, or camelid-derived nanobodies, provide comparable specificity with reduced steric hindrance[79]. However, caution is warranted when considering antibodies for such applications, as their use would classify the formulation as a biologic, necessitating a rigorous drug approval process that typically spans over a decade and requires investments of hundreds of millions of dollars.

MECHANISMS AND EFFICACY OF LIGAND-MEDIATED DELIVERY

Upon reaching systemic circulation, ligand-functionalized nanoliposomes exhibit enhanced docking and internalization by target cells, markedly elevating tissue-specific nutrient concentrations[31]. For instance, studies employing targeted liposomes for drug delivery report several-fold increases in tumor tissue concentrations compared to untargeted formulations, a principle extensible to nutrient payloads[25]. Despite their potential, ligand-mediated targeting presents several challenges.

Systemic delivery efficiency

Not all orally administered nanoliposomes reach circulation intact; a fraction may be absorbed as degraded components, limiting the efficacy of targeting to the intact vesicular population[45].

Clearance kinetics

Ligands, particularly antibodies, may accelerate clearance by triggering opsonization and uptake by the MPS[80].

Ligand density optimization

Excessive ligand conjugation risks vesicle aggregation or interference with endosomal escape mechanisms, necessitating a balance between targeting efficacy and functional integrity. To illustrate organ targeting, Table 2 Lists some targeting strategies and examples relevant to nutrient delivery[12-28].

Table 2 Examples of targeting strategies for nanoliposomal nutrient delivery.
Target organ/cell
Targeting ligand on liposome
Example outcome
Intestinal M cells (Peyer’s patches)Ulex europaeus agglutinin I (lectin) or targeting peptide for M cellsEnhanced uptake of oral liposome via Peyer’s patches, often utilized in oral vaccine delivery (concept can be extrapolated to nutrients)
Intestinal enterocytesFolic acid (folate receptor targeting)Increased oral absorption of hydrophilic compounds by folate receptor - mediated uptake; may similarly improve nutrient uptake
Liver (hepatocytes)Galactose or N-acetyl galactosamine (asialoglycoprotein receptor ligand)Specific delivery to hepatocytes (e.g., for vitamin A or D) via the asialoglycoprotein receptor, which binds galactose-terminated ligands
Liver (stellate cells)Mannose-6-phosphate or vitamin A (retinol)Hepatic stellate cells naturally store vitamin A; thus, vitamin A-decorated liposomes can deliver anti-fibrotic nutrients to stellate cells in liver fibrosis
Brain (neurons or blood-brain barrier)Transferrin or anti-transferrin receptor antibodyEnables liposomes to cross the blood-brain barrier via transferrin receptor - mediated transcytosis, facilitating delivery of neuroprotective nutrients to the central nervous system
Heart (cardiomyocytes)Peptide targeting mitochondria or surface markers (e.g., for myosin or angiotensin II type 1 receptor)Experimental strategy to deliver coenzyme Q10 or antioxidants to heart muscle. In certain models, angiotensin-targeted nanoparticles accumulate preferentially in cardiac tissue
Kidney (glomerular mesangial cells)Anti-Thy1.1 antibody (in rats) or anti-integrin α8 antibodyAchieves a marked increase (e.g., approximately 6-fold) in liposome accumulation within renal glomeruli, allowing targeted delivery of antioxidants or other agents to mesangial cells, potentially benefiting nephritis therapy
Kidney (proximal tubule)Lysozyme or small peptide (binds megalin receptor)Enhances uptake into proximal tubule cells; may deliver vitamin E or carotenoids to mitigate oxidative damage in chronic kidney disease
Immune cells (macrophages)Mannose (mannose receptor targeting)Facilitates uptake by macrophages (e.g., in spleen or liver). Could be used to deliver anti-inflammatory vitamins (e.g., vitamin D) to macrophages in immune-related organs
Tumor (for cancer prevention)Arginine-glycine-aspartic acid peptide (targets integrin on angiogenic endothelium)Allows liposomes to accumulate in tumor neo vasculature, potentially delivering high doses of selenium or other antitumor nutrients in a localized manner (extrapolated from drug-targeting approaches)
MITOCHONDRIAL TARGETING STRATEGIES IN NANOLIPOSOMAL INTRACELLULAR DELIVERY

While organ-specific targeting directs nanoliposomes to designated tissues, mitochondrial targeting aims to localize bioactive compounds within a specific subcellular compartment - the mitochondria - critical for cellular bioenergetics and homeostasis[31]. Mitochondrial dysfunction is implicated in a diverse array of pathologies, encompassing neurodegenerative disorders, metabolic syndromes, and cardiovascular diseases.

Numerous nutritional supplements designed to bolster mitochondrial function exert their therapeutic effects at this organelle, yet their delivery poses significant challenges, requiring traversal of both outer and inner mitochondrial membranes to achieve accumulation within the mitochondrial matrix or membrane[32]. Two principal strategies have emerged to facilitate mitochondrial-specific delivery: (1) Lipophilic cationic moieties, exemplified by TPP+; and (2) Mitochondria-targeting peptide sequences, such as SS-31[81]. Nanoliposomal systems can integrate these targeting entities to enhance subcellular delivery precision.

TPP+ AND LIPOPHILIC CATIONIC TARGETING

TPP+, a lipophilic cation bearing a delocalized positive charge, leverages its membrane-permeant properties to selectively accumulate within mitochondria, driven by the organelle’s substantial membrane potential, typically ranging from -150 mV to -180 mV (negative inside)[82]. This electrochemical gradient preferentially attracts cations, enabling TPP+-conjugated molecules to localize within the mitochondrial compartment. A prominent example is mitoquinone, a CoQ10 analogue functionalized with TPP+, which demonstrates enhanced mitochondrial partitioning[83]. In nanoliposomal formulations, TPP+ can be incorporated via surface conjugation or integration of TPP+-modified lipids into the bilayer, directing the vesicles - or their dissociated payloads - towards mitochondria post-cytosolic entry[84].

Empirical evidence substantiates the efficacy of TPP+-functionalized nanocarriers in enhancing mitochondrial delivery[85]. TPP+-modified liposomes have been shown to augment the mitochondrial accumulation of chemotherapeutic agents in neoplastic cells, thereby amplifying cytotoxicity. Similarly, TPP+-conjugated polymersomes encapsulating doxorubicin circumvent multidrug resistance by targeting mitochondrial pathways. For nutrient delivery, TPP+-functionalized liposomal CoQ10 could theoretically enhance its localization to mitochondrial membranes, where it contributes to the electron transport chain and mitigates oxidative stress[86]. Unlike conventional oral CoQ10, which predominantly associates with plasma lipoproteins, this approach may preferentially target metabolically active cells, such as cardiomyocytes and neurons. Another application involves “Mito-vitamin E”, wherein TPP+ conjugation to vitamin E protects mitochondrial membranes from lipid peroxidation.

Despite its utility, TPP+-mediated targeting presents limitations. Excessive mitochondrial accumulation of TPP+ can perturb membrane potential, potentially eliciting cytotoxicity at elevated concentrations. To mitigate this, optimal TPP+ loading densities must be established, with some formulations employing PEG spacers to modulate surface presentation and attenuate toxicological risks[74]. Chronic exposure to TPP+-conjugated agents may also induce off-target effects, necessitating comprehensive toxicological assessments. Researchers should consider pivoting away from TPP+ due to its inherent risks, as advancements at the nexus of nanocarrier technology and mitochondrial nutrient optimization have yielded superior alternatives. More effective options, such as peptide-based targeting or receptor-specific ligands offer greater precision and reduced toxicity, harnessing natural cellular pathways without compromising mitochondrial integrity or requiring cumbersome mitigation tactics. These alternatives not only enhance specificity but also align better with the goal of safe, efficient nutrient delivery, rendering TPP+ an outdated and less desirable choice.

MITOCHONDRIA-TARGETING PEPTIDES IN NANOLIPOSOMAL SUBCELLULAR DELIVERY

Szeto-Schiller peptides, characterized by their concise tetrapeptide structure, exhibit a distinctive capacity for cellular penetration and selective mitochondrial localization, employing a mechanism divergent from that of lipophilic cations such as TPP+[87]. Among these, SS-31 (Elamipretide), with the sequence D-Arg-2′,6′-dimethylTyr-Lys-Phe-NH2 (denoted as H-D-Arg-Dmt-Lys-Phe-NH2), represents the most rigorously investigated variant[88]. Bearing multiple positive charges from its arginine, lysine, and amidated C-terminus, SS-31 does not rely on the mitochondrial membrane potential for uptake, distinguishing it from potential-dependent strategies. Instead, SS-31 demonstrates high-affinity binding to cardiolipin, a phospholipid enriched in the inner mitochondrial membrane, facilitating its accumulation within the mitochondrial matrix[89]. This interaction enables SS-31 to exert antioxidant effects by associating with cytochrome C, thereby inhibiting its peroxidase activity and mitigating oxidative stress. Clinically, SS-31 has been evaluated for therapeutic efficacy across diverse pathologies, including congestive heart failure, renal dysfunction, and age-related degenerative conditions.

For intracellular nutrient delivery, SS-31 can be conjugated to nanoliposomal surfaces to enhance mitochondrial specificity. In contrast to TPP+-based approaches, SS-31 exhibits minimal disruption of mitochondrial membrane potential and retains efficacy in cells with compromised bioenergetic states, a critical advantage in pathological contexts. Furthermore, SS-31 circumvents exclusion mechanisms that impede the uptake of other cationic entities, broadening its applicability.

APPLICATION IN NANOLIPOSOMAL DELIVERY SYSTEMS

A seminal investigation demonstrated the utility of SS-31-functionalized polylactic-co-glycolic acid nanoparticles in targeting mitochondrial compartments within inner ear hair cells. These SS-31-modified nanocarriers effectively accumulated within mitochondria, delivering geranylgeranylacetone (GGA) to attenuate antibiotic-induced ototoxicity[90]. Notably, only SS-31-functionalized nanoparticles preserved mitochondrial membrane potential under oxidative stress, underscoring their organelle-specific delivery precision. This paradigm is applicable to nutrient delivery: CoQ10, riboflavin or niacin derivatives might be delivered directly to mitochondria to bolster cofactor levels and optimize metabolic function[91]. Similar considerations apply to melatonin, a robust mitochondrial antioxidant, and other compounds posited to boost mitochondrial resilience[92].

Other mitochondria-penetrating peptides, enriched in basic and aromatic residues, share analogous targeting capabilities, as exemplified by the MITO-Porter system, which employs peptide sequences to direct liposomal cargos to mitochondria[93]. SS-31, however, is distinguished by its dual functionality: Beyond serving as a targeting ligand, it confers intrinsic therapeutic benefits through antioxidant and bioenergetic stabilization. In nanoliposomal formulations, dissociated SS-31 peptides may independently enhance mitochondrial function, while surface-bound SS-31 directs the encapsulated payload to the organelle. This dual-action mechanism was evidenced in the inner ear study, where SS-31-modified nanoparticles alone provided partial protection, with GGA-loaded variants yielding superior efficacy.

Mitochondrial targeting strategies, exemplified by TPP+ and SS-31, offer a transformative approach to enhancing the therapeutic potency of interventions addressing mitochondrial dysfunction[94]. Experimental models have substantiated their efficacy, demonstrating potential clinical utility in pathologies characterized by impaired mitochondrial bioenergetics or oxidative stress[95]. For instance, a mitochondria-targeted CoQ10 formulation could achieve heightened concentrations within mitochondrial membranes, amplifying its role in electron transport and antioxidant defense. These advancements underscore the capacity of targeted nanoliposomal systems to bridge preclinical innovation and clinical application in metabolic and age-related disorders.

It is crucial to note that effective mitochondrial delivery via liposomes first requires endosomal escape into the cytosol, allowing targeting ligands (such as SS-31) to engage the organelle surface. Without this escape, liposomes would release their payload prematurely into the intracellular space and fail to reach their intended site of action[96].

OVERCOMING ENDOSOMAL CONFINEMENT FOR INTRACELLULAR DELIVERY

A principal impediment in nanoparticle-mediated delivery, including nanoliposomal systems, is the requisite escape from endosomal compartments following RME[31]. Upon internalization, nanoliposomes bearing targeting ligands are sequestered within endosomes, necessitating egress into the cytosol to enable cytosolic or organelle-specific functionality of encapsulated therapeutics or nutraceuticals[32].

Failure to achieve this escape results in endosomal maturation into lysosomes, where acidic pH (approximately 4.5-5.0) and hydrolytic enzymes degrade the payload or mediate its exocytosis, thereby attenuating efficacy. Consequently, the rational design of nanoliposomes to facilitate cytosolic release - particularly for macromolecular cargos (e.g., nucleic acids, proteins) and nutrients targeting cytosolic or mitochondrial compartments - constitutes a critical engineering imperative[49].

The pH-responsive mechanisms: Proton sponge and membrane disruption

Endosomal acidification, decreasing pH to 5.0-6.0 during maturation, provides an exploitable trigger for endosomal escape[97]. The pH-sensitive lipids, such as dioleoylphosphatidylethanolamine (DOPE) coupled with acid-titratable lipids like cholesteryl hemisuccinate (CHEMS), capitalize on this gradient[98]. At neutral pH, the DOPE/CHEMS bilayer maintains structural integrity; however, protonation of CHEMS at acidic pH reduces its negative charge, inducing a transition to the inverted hexagonal phase[53], which destabilizes endosomal membranes and promotes payload release[22].

DOPE’s fusogenic properties, stemming from its propensity to disrupt bilayer architecture under acidic conditions, enhance cytosolic delivery, as evidenced by augmented drug release and cytotoxicity in folate-targeted, pH-sensitive liposomal systems. Alternatively, the proton sponge effect employs cationic polymers, such as polyethylenimine (PEI), which buffer endosomal protons, eliciting ion influx, osmotic swelling, and subsequent membrane rupture. While PEI exhibits cytotoxicity at elevated concentrations, judicious incorporation into liposomal formulations mitigates toxicity while facilitating escape.

Membrane-lytic peptides (e.g., GALA, INF7)

Peptides engineered to adopt lytic or fusogenic conformations at acidic pH emulate viral endosomal escape mechanisms. The synthetic peptide GALA (WEAALAEALAEALAEHLAEALAEALEALAA) remains quiescent at neutral pH but adopts an α-helical structure in acidic endosomal environments, inserting into and perforating membranes to enable cytosolic access[99]. Similarly, INF7, derived from influenza hemagglutinin, induces pH-dependent membrane fusion when conjugated to nanoliposomes, mirroring viral egress kinetics. These biomimetic strategies enhance liposomal escape efficiency with high specificity.

Enzyme-responsive mechanisms

Enzyme-triggered escape leverages endosomal or lysosomal hydrolases to cleave liposomal constituents, transforming inert lipids into fusogenic species. For instance, hydrolysis of a saccharide head group by lysosomal glycosidases exposes a detergent-like lipid, disrupting endosomal membranes. Though less prevalent than pH-responsive approaches, this method offers tailored specificity in enzyme-rich microenvironments[100].

Photochemical and internal triggers

Photochemically activated nanoliposomes utilize light-induced generation of ROS or structural rearrangements to destabilize endosomal membranes. Despite their efficacy in experimental settings, the requirement for direct light application limits applicability to internal tissues, constraining utility in nutraceutical delivery[101].

INTEGRATION OF TARGETING AND ENDOSOMAL ESCAPE IN NANOLIPOSOMAL DELIVERY SYSTEMS

Nanoliposomes engineered for RME face the challenge of payload sequestration within endosomal compartments unless mechanisms for cytosolic egress are incorporated[31]. While certain nutrients, such as iron or cobalamin (vitamin B12), leverage endogenous endosomal processing pathways to facilitate release into the cytosol, non-physiological cargos - mitochondrial co-factors - necessitate tailored engineering solutions to evade degradation or exocytosis[32].

Contemporary liposomal architectures thus integrate cell-specific targeting ligands with pH-responsive or fusogenic functionalities to optimize intracellular delivery. For instance, hyaluronic acid-coated liposomes, incorporating DOPE and CHEMS, target CD44 receptors on neoplastic cells; subsequent endosomal acidification degrades the hyaluronic acid coating, activating DOPE’s fusogenic properties to disrupt membranes and enable cytosolic release. Similarly, conformationally switchable lipids have been developed that destabilize endosomal membranes at reduced pH, enhancing payload liberation[98].

This integration markedly enhances cytosolic delivery efficiency, as evidenced by improved transfection and therapeutic outcomes for macromolecules, including ribosome-inactivating proteins and nucleic acids. DOPE-enriched liposomes consistently outperform non-fusogenic counterparts in facilitating intracellular access, underscoring the efficacy of these design principles. For nutraceutical applications, such strategies mitigate endosomal degradation, enabling the direct delivery of essential biomolecules - such as DNA, enzymes, or metabolic cofactors - to their functional intracellular loci.

MITOCHONDRIAL MULTI-NUTRIENT LIPOSOMAL FORMULATION

A conceptual mitochondrial multi-nutrient formulation exemplifies the convergence of targeting and escape mechanisms to augment mitochondrial bioenergetics. This nanoliposomal system could encapsulate: (1) Lipophilic constituents, such as CoQ10 and alpha-lipoic acid, within the lipid bilayer; (2) Hydrophilic nicotinamide adenine dinucleotide precursors (niacinamide) and small peptides within the aqueous core; and (3) Surface-bound targeting ligands, such as SS-31 for mitochondrial specificity and potentially tissue-specific antibodies for enhanced cellular uptake[90].

Following oral administration, the liposomal bilayer shields these payloads from GI hydrolytic and enzymatic degradation, improving transepithelial absorption[45]. Upon systemic circulation, stealth properties sustain plasma persistence until receptor-mediated internalization by target cells occurs. Endosomal acidification subsequently activates pH-sensitive components, such as DOPE, which transition to a fusogenic state, permeabilizing the endosomal membrane and releasing the cargo into the cytosol. SS-31 then directs the liberated payload to mitochondria, amplifying electron transport chain efficiency, antioxidant capacity, and cellular energy metabolism. This integrated vector could substantially enhance mitochondrial function beyond the capabilities of conventional oral supplementation.

Although this mitochondrial multi-nutrient formulation remains theoretical, it encapsulates validated principles demonstrated to enhance absorption, protect labile compounds, and achieve precise intracellular targeting. These sophisticated platforms underscore the necessity of synergizing biophysical design elements - such as pH sensitivity and fusogenic lipids - with molecular targeting and controlled release to maximize therapeutic and nutraceutical efficacy[31].

COQ10: AN IMPORTANT NUTRACEUTICAL WITH BIOAVAILABILITY CONSTRAINTS

CoQ10, an endogenous lipophilic quinone, exemplifies a nutraceutical with profound therapeutic potential yet constrained by suboptimal pharmacokinetic properties[102]. Integral to mitochondrial adenosine triphosphate (ATP) synthesis within the electron transport chain and functioning as a potent lipid-phase antioxidant, CoQ10 supports cardiovascular integrity - ameliorating conditions such as heart failure and statin-associated myopathy - and exhibits neuroprotective effects in neurodegenerative disorders[103]. Despite its clinical promise, the extreme hydrophobicity of crystalline CoQ10 results in oral bioavailability typically below 5%, compounded by significant inter-individual variability in plasma concentrations[104]. Various formulations, encompassing oil-based suspensions, solubilized preparations, and nanoparticle-mediated carriers, have been developed to surmount these limitations[105]. Contemporary analyses indicate that suboptimal delivery systems markedly diminish CoQ10 bioavailability, whereas advanced formulations achieve multifold elevations in systemic plasma levels[31].

ADVANTAGES OF NANOLIPOSOMAL COQ10 DELIVERY

Nanoliposomal encapsulation offers distinct biophysical and pharmacokinetic advantages for CoQ10 administration[106].

Prevention of crystallization and augmented intestinal absorption

Integration of CoQ10 into the liposomal lipid bilayer inhibits its crystallization, thereby enhancing solubility and facilitating transepithelial absorption across the intestinal mucosa[49].

Promotion of lymphatic transport

Nanoliposomes engineered to diameters conducive to lymphatic uptake circumvent first-pass hepatic metabolism, channeling CoQ10 directly into systemic circulation via the thoracic duct[107].

Mitochondrial membrane integration

Conjugation with mitochondria-targeting entities, such as Szeto-Schiller peptides directs CoQ10 to mitochondrial membranes, amplifying its contributions to ATP production and oxidative stress mitigation[84].

OTHER NANOLIPOSOME THERAPEUTIC APPLICATIONS

For many vitamins and minerals, the primary delivery challenge is enhancing bioavailability rather than directing the nutrient to a specific tissue. For instance, liposomal vitamin C can boost plasma ascorbate levels, and liposomal iron formulations may reduce GI side effects while targeting iron uptake pathways[108]. Liposomal vitamin D could be particularly valuable in individuals with fat malabsorption, ensuring adequate serum 25(OH)D levels. Depending on the nutrient’s clinical objective - whether improving overall absorption or directing the nutrient to specific tissues - liposome design can be customized with or without targeting ligands.

Heavy metal detoxification represents an important therapeutic consideration. A significant proportion of the population exhibits elevated burdens of mercury, cadmium, aluminum, and lead, which accumulate in various tissues and contribute to systemic toxicity[109]. Conventional chelation strategies, while effective, often necessitate high systemic doses that may engender adverse effects and dose-limiting toxicities[110]. Targeted nanoliposomal delivery systems offer a promising alternative by enabling precise administration of chelating agents to sites of heavy metal deposition, thereby enhancing efficacy and minimizing collateral toxicity[111]. For instance, lead, which predominantly accumulates in bone (accounting for approximately 99% of its body burden), co-localizes with other toxic metals such as aluminum and cadmium[112]. By targeting integrin receptors on osteocytes or bone matrix components, nanoliposomal formulations could precisely deliver chelating agents to these skeletal reservoirs, enhancing metal extraction efficiency while minimizing the broader physiological burden[113].

Although iron is not classified as a heavy metal, excessive accumulation is prevalent among adult males and postmenopausal females, contributing to oxidative stress and cellular injury via ROS generation. Targeted nanoliposomal delivery to Kupffer cells within the hepatic reticuloendothelial system, which governs iron recycling, could provide a novel therapeutic avenue[114]. Such an approach may serve as an effective alternative to therapeutic phlebotomy, offering precise modulation of iron homeostasis with potentially fewer procedural risks.

Mitochondrial-targeted nanoliposomal systems further expand therapeutic potential by addressing organ-specific cellular damage, particularly in conditions characterized by metabolic dysfunction, such as diabetes mellitus and atherosclerosis[115]. These systems can deliver bioactive payloads to mitochondria within designated tissues, mitigating severe cellular impairments resulting from metabolic stress. Mercury, a potent neurotoxin, exemplifies the need for such precision. Although it deposits across multiple tissues, its primary pathological impact manifests in the brain, where sulfhydryl groups bind to mitochondrial complexes, precipitating profound cellular dysfunction[116]. In the kidney, mercury predominantly accumulates within the proximal tubule epithelial cells, necessitating intracellular delivery of chelating agents to these sites to accelerate metal sequestration and excretion[117].

CURRENT CHALLENGES AND FUTURE DIRECTIONS

Although these strategies hold tremendous therapeutic promise, key technical and regulatory considerations must be addressed.

Scalability of manufacturing

Scaling liposome production from laboratory bench volumes to industrial batches remains a major challenge, especially for cost-sensitive nutraceuticals. Techniques such as high-pressure homogenization and microfluidic mixing are being refined to achieve consistent particle sizes, high encapsulation efficiency, and stable ligand attachment. Continuous manufacturing platforms may eventually enable more economical, large-scale liposome production[31].

Stability and shelf-life

Unlike conventional pills, liposomes risk leakage of payload, aggregation, or lipid oxidation over time. Strategies such as freeze-drying (lyophilization), the incorporation of antioxidants, and the choice of stable lipid compositions can extend product shelf life[118]. Preserving the functionality of targeting ligands - which may degrade over prolonged storage - also requires formulation optimization[119].

Safety and toxicology

Liposomes are widely considered biocompatible, yet the cumulative impact of chronic nanoparticle intake remains under investigation. Key concerns include potential nanoparticle accumulation in specific organs, impacts on gut microbiota, and safety of higher systemic levels of nutrients previously constrained by poor absorption. While existing data are reassuring, each new liposomal system (with novel ligands or materials) must undergo toxicological evaluation[120].

Personalization and integration with other technologies

Future trends point toward personalized nutrition, whereby liposomal carriers are tailored to individual genetic or metabolic profiles[121]. Hybrid nanoparticles (e.g., polymer-lipid or exosome-based vesicles) could combine the advantages of multiple delivery systems. Moreover, continued innovation in green or plant-derived vesicles (sometimes referred to as “edible exosomes”) may offer cost-effective, sustainable alternatives.

Ethical and regulatory considerations

As these advanced delivery methods blur the line between supplements and pharmaceuticals, regulatory classifications and approval pathways grow increasingly complex[122]. Careful oversight and responsible marketing are critical to prevent overhyped claims, while ensuring these potentially transformative solutions remain accessible.

CONCLUSION

Nanoliposomal and related nanotechnology-based formulations constitute a paradigm-shifting advancement in nutrient delivery, facilitating site-specific targeting, optimized bioavailability, and pharmacodynamic profiles approaching those of conventional therapeutics. Despite persistent challenges - including scalability, formulation stability, clinical substantiation, and regulatory frameworks - emerging evidence from ongoing investigations indicates these obstacles are surmountable. As innovations in manufacturing refine production efficiency, reduce economic barriers, and robust clinical trials substantiate therapeutic efficacy, targeted nanoliposomal nutrient systems are poised to emerge as a foundational element of precision nutrition. This evolution promises to fundamentally reshape the application of nutraceuticals in the prevention and management of disease, heralding a new era in evidence-based nutritional science.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: United States

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade A

Creativity or Innovation: Grade B

Scientific Significance: Grade A

P-Reviewer: Wu F, PhD, China S-Editor: Luo ML L-Editor: A P-Editor: Zhang L

References
1.  McClements DJ, Li F, Xiao H. The Nutraceutical Bioavailability Classification Scheme: Classifying Nutraceuticals According to Factors Limiting their Oral Bioavailability. Annu Rev Food Sci Technol. 2015;6:299-327.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 160]  [Cited by in RCA: 172]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
2.  Purpura M, Jäger R, Godavarthi A, Bhaskarachar D, Tinsley GM. Liposomal delivery enhances absorption of vitamin C into plasma and leukocytes: a double-blind, placebo-controlled, randomized trial. Eur J Nutr. 2024;63:3037-3046.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
3.  Cesarano D, Borrelli S, Campilongo G, D'Ambra A, Papadia F, Garofalo C, De Marco A, Marzano F, Ruotolo C, Gesualdo L, Cirillo P, Minutolo R. Efficacy and Safety of Oral Supplementation with Liposomal Iron in Non-Dialysis Chronic Kidney Disease Patients with Iron Deficiency. Nutrients. 2024;16:1255.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
4.  Zhang H. Thin-Film Hydration Followed by Extrusion Method for Liposome Preparation. Methods Mol Biol. 2017;1522:17-22.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 209]  [Cited by in RCA: 292]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
5.  Yanar F, Mosayyebi A, Nastruzzi C, Carugo D, Zhang X. Continuous-Flow Production of Liposomes with a Millireactor under Varying Fluidic Conditions. Pharmaceutics. 2020;12:1001.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
6.  Han JY, La Fiandra JN, DeVoe DL. Microfluidic vortex focusing for high throughput synthesis of size-tunable liposomes. Nat Commun. 2022;13:6997.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 38]  [Cited by in RCA: 47]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
7.  Gaballa SA, Shimizu T, Ando H, Takata H, Emam SE, Ramadan E, Naguib YW, Mady FM, Khaled KA, Ishida T. Treatment-induced and Pre-existing Anti-peg Antibodies: Prevalence, Clinical Implications, and Future Perspectives. J Pharm Sci. 2024;113:555-578.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 19]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
8.  Tenchov R, Sasso JM, Zhou QA. PEGylated Lipid Nanoparticle Formulations: Immunological Safety and Efficiency Perspective. Bioconjug Chem. 2023;34:941-960.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 132]  [Reference Citation Analysis (0)]
9.  Altemimi AB, Farag HAM, Salih TH, Awlqadr FH, Al-Manhel AJA, Vieira IRS, Conte-Junior CA. Application of Nanoparticles in Human Nutrition: A Review. Nutrients. 2024;16:636.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 20]  [Reference Citation Analysis (0)]
10.  Ouyang X, Liu Y, Zheng K, Pang Z, Peng S. Recent advances in zwitterionic nanoscale drug delivery systems to overcome biological barriers. Asian J Pharm Sci. 2024;19:100883.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
11.  Kakuda L, Maia Campos PMBG, Oliveira WP. Development and Efficacy Evaluation of Innovative Cosmetic Formulations with Caryocar brasiliense Fruit Pulp Oil Encapsulated in Freeze-Dried Liposomes. Pharmaceutics. 2024;16:595.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
12.  Yang Y, Chen J, Wang S, Zhu Y, Wang Y, Chen Y, Xia M, Yang M, Yi H, Su K. pH-Responsive Liposome-Hydrogel Composite Accelerates Nasal Mucosa Wound Healing. Pharmaceutics. 2025;17:690.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
13.  Liu S, Wen X, Zhang X, Mao S. Oral delivery of biomacromolecules by overcoming biological barriers in the gastrointestinal tract: an update. Expert Opin Drug Deliv. 2023;20:1333-1347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
14.  Goodman BE. Insights into digestion and absorption of major nutrients in humans. Adv Physiol Educ. 2010;34:44-53.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 186]  [Cited by in RCA: 193]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
15.  Savjani KT, Gajjar AK, Savjani JK. Drug solubility: importance and enhancement techniques. ISRN Pharm. 2012;2012:195727.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 516]  [Cited by in RCA: 658]  [Article Influence: 50.6]  [Reference Citation Analysis (0)]
16.  Lin CH, Chen CH, Lin ZC, Fang JY. Recent advances in oral delivery of drugs and bioactive natural products using solid lipid nanoparticles as the carriers. J Food Drug Anal. 2017;25:219-234.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 164]  [Cited by in RCA: 169]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
17.  Artursson P, Palm K, Luthman K. Caco-2 monolayers in experimental and theoretical predictions of drug transport. Adv Drug Deliv Rev. 2001;46:27-43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 880]  [Cited by in RCA: 990]  [Article Influence: 41.3]  [Reference Citation Analysis (0)]
18.  Takano M, Yumoto R, Murakami T. Expression and function of efflux drug transporters in the intestine. Pharmacol Ther. 2006;109:137-161.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 276]  [Cited by in RCA: 240]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
19.  Johansson ME, Sjövall H, Hansson GC. The gastrointestinal mucus system in health and disease. Nat Rev Gastroenterol Hepatol. 2013;10:352-361.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 793]  [Cited by in RCA: 1018]  [Article Influence: 84.8]  [Reference Citation Analysis (0)]
20.  Pond SM, Tozer TN. First-pass elimination. Basic concepts and clinical consequences. Clin Pharmacokinet. 1984;9:1-25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 268]  [Cited by in RCA: 255]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
21.  Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci. 2006;27:425-446.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 397]  [Cited by in RCA: 356]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
22.  Dahan A, Miller JM, Amidon GL. Prediction of solubility and permeability class membership: provisional BCS classification of the world's top oral drugs. AAPS J. 2009;11:740-746.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 275]  [Cited by in RCA: 285]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
23.  Xi M, Kia SH, Shi H, Dong X, Shi Y, Zhang L, Jiang B. Synthesis and characterization of berberine-loaded nanoliposome for targeting of MAPK pathway to induce apoptosis and suppression of autophagy in glioblastoma. Biomed Mater. 2025;20.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
24.  Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4:145-160.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3592]  [Cited by in RCA: 3491]  [Article Influence: 174.6]  [Reference Citation Analysis (0)]
25.  Ko J, Yoo C, Xing D, Gonzalez DE, Jenkins V, Dickerson B, Leonard M, Nottingham K, Kendra J, Sowinski R, Rasmussen CJ, Kreider RB. Pharmacokinetic Analyses of Liposomal and Non-Liposomal Multivitamin/Mineral Formulations. Nutrients. 2023;15:3073.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
26.  Davis JL, Paris HL, Beals JW, Binns SE, Giordano GR, Scalzo RL, Schweder MM, Blair E, Bell C. Liposomal-encapsulated Ascorbic Acid: Influence on Vitamin C Bioavailability and Capacity to Protect Against Ischemia-Reperfusion Injury. Nutr Metab Insights. 2016;9:25-30.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
27.  Li Y, Li XM, Wei LS, Ye JF. Advancements in mitochondrial-targeted nanotherapeutics: overcoming biological obstacles and optimizing drug delivery. Front Immunol. 2024;15:1451989.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
28.  Pathak C, Vaidya FU, Waghela BN, Jaiswara PK, Gupta VK, Kumar A, Rajendran BK, Ranjan K. Insights of Endocytosis Signaling in Health and Disease. Int J Mol Sci. 2023;24:2971.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 29]  [Reference Citation Analysis (0)]
29.  Dolman ME, Harmsen S, Storm G, Hennink WE, Kok RJ. Drug targeting to the kidney: Advances in the active targeting of therapeutics to proximal tubular cells. Adv Drug Deliv Rev. 2010;62:1344-1357.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 104]  [Cited by in RCA: 131]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
30.  Trevaskis NL, Kaminskas LM, Porter CJ. From sewer to saviour - targeting the lymphatic system to promote drug exposure and activity. Nat Rev Drug Discov. 2015;14:781-803.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 370]  [Cited by in RCA: 506]  [Article Influence: 50.6]  [Reference Citation Analysis (0)]
31.  Chatterjee S, Kon E, Sharma P, Peer D. Endosomal escape: A bottleneck for LNP-mediated therapeutics. Proc Natl Acad Sci U S A. 2024;121:e2307800120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 210]  [Article Influence: 210.0]  [Reference Citation Analysis (0)]
32.  Patil YP, Jadhav S. Novel methods for liposome preparation. Chem Phys Lipids. 2014;177:8-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 363]  [Cited by in RCA: 363]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
33.  Simberg D, Barenholz Y, Roffler SR, Landfester K, Kabanov AV, Moghimi SM. PEGylation technology: addressing concerns, moving forward. Drug Deliv. 2025;32:2494775.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
34.  Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and Challenges of Liposome Assisted Drug Delivery. Front Pharmacol. 2015;6:286.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1168]  [Cited by in RCA: 1621]  [Article Influence: 162.1]  [Reference Citation Analysis (0)]
35.  Filipczak N, Pan J, Yalamarty SSK, Torchilin VP. Recent advancements in liposome technology. Adv Drug Deliv Rev. 2020;156:4-22.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 134]  [Cited by in RCA: 356]  [Article Influence: 71.2]  [Reference Citation Analysis (0)]
36.  Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine. 2006;1:297-315.  [PubMed]  [DOI]
37.  Veronese FM, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today. 2005;10:1451-1458.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1671]  [Cited by in RCA: 1701]  [Article Influence: 85.1]  [Reference Citation Analysis (0)]
38.  D'souza AA, Shegokar R. Polyethylene glycol (PEG): a versatile polymer for pharmaceutical applications. Expert Opin Drug Deliv. 2016;13:1257-1275.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 417]  [Cited by in RCA: 681]  [Article Influence: 75.7]  [Reference Citation Analysis (0)]
39.  Rizzo F, Kehr NS. Recent Advances in Injectable Hydrogels for Controlled and Local Drug Delivery. Adv Healthc Mater. 2021;10:e2001341.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 64]  [Cited by in RCA: 211]  [Article Influence: 52.8]  [Reference Citation Analysis (0)]
40.  Knop K, Hoogenboom R, Fischer D, Schubert US. Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives. Angew Chem Int Ed Engl. 2010;49:6288-6308.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2443]  [Cited by in RCA: 2515]  [Article Influence: 167.7]  [Reference Citation Analysis (0)]
41.  Yang Q, Lai SK. Anti-PEG immunity: emergence, characteristics, and unaddressed questions. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2015;7:655-677.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 300]  [Cited by in RCA: 423]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
42.  Spears BR, Waksal J, McQuade C, Lanier L, Harth E. Controlled branching of polyglycidol and formation of protein-glycidol bioconjugates via a graft-from approach with "PEG-like" arms. Chem Commun (Camb). 2013;49:2394-2396.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 33]  [Cited by in RCA: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
43.  Hoang Thi TT, Pilkington EH, Nguyen DH, Lee JS, Park KD, Truong NP. The Importance of Poly(ethylene glycol) Alternatives for Overcoming PEG Immunogenicity in Drug Delivery and Bioconjugation. Polymers (Basel). 2020;12:298.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 411]  [Cited by in RCA: 425]  [Article Influence: 85.0]  [Reference Citation Analysis (0)]
44.  Li SD, Huang L. Stealth nanoparticles: high density but sheddable PEG is a key for tumor targeting. J Control Release. 2010;145:178-181.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 468]  [Cited by in RCA: 418]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
45.  Harris JM, Chess RB. Effect of pegylation on pharmaceuticals. Nat Rev Drug Discov. 2003;2:214-221.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2566]  [Cited by in RCA: 2567]  [Article Influence: 116.7]  [Reference Citation Analysis (0)]
46.  Gdowski AS, Lampe JB, Lin VJT, Joshi R, Wang YC, Mukerjee A, Vishwanatha JK, Ranjan AP. Bioinspired Nanoparticles Engineered for Enhanced Delivery to the Bone. ACS Appl Nano Mater. 2019;2:6249-6257.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
47.  Almeida B, Nag OK, Rogers KE, Delehanty JB. Recent Progress in Bioconjugation Strategies for Liposome-Mediated Drug Delivery. Molecules. 2020;25:5672.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 124]  [Cited by in RCA: 192]  [Article Influence: 38.4]  [Reference Citation Analysis (0)]
48.  Lundquist P, Artursson P. Oral absorption of peptides and nanoparticles across the human intestine: Opportunities, limitations and studies in human tissues. Adv Drug Deliv Rev. 2016;106:256-276.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 278]  [Cited by in RCA: 340]  [Article Influence: 37.8]  [Reference Citation Analysis (0)]
49.  Wang D, Jiang Q, Dong Z, Meng T, Hu F, Wang J, Yuan H. Nanocarriers transport across the gastrointestinal barriers: The contribution to oral bioavailability via blood circulation and lymphatic pathway. Adv Drug Deliv Rev. 2023;203:115130.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 51]  [Reference Citation Analysis (0)]
50.  Florence AT. The oral absorption of micro- and nanoparticulates: neither exceptional nor unusual. Pharm Res. 1997;14:259-266.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 385]  [Cited by in RCA: 352]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
51.  Guimarães D, Cavaco-Paulo A, Nogueira E. Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm. 2021;601:120571.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 94]  [Cited by in RCA: 641]  [Article Influence: 160.3]  [Reference Citation Analysis (0)]
52.  Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev. 2013;65:36-48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2871]  [Cited by in RCA: 3227]  [Article Influence: 268.9]  [Reference Citation Analysis (0)]
53.  Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release. 2010;148:135-146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1824]  [Cited by in RCA: 1865]  [Article Influence: 124.3]  [Reference Citation Analysis (0)]
54.  Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv Drug Deliv Rev. 2013;65:71-79.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1622]  [Cited by in RCA: 1787]  [Article Influence: 148.9]  [Reference Citation Analysis (0)]
55.  Porter CJ, Trevaskis NL, Charman WN. Lipids and lipid-based formulations: optimizing the oral delivery of lipophilic drugs. Nat Rev Drug Discov. 2007;6:231-248.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1239]  [Cited by in RCA: 1301]  [Article Influence: 72.3]  [Reference Citation Analysis (0)]
56.  Lee Y, Thompson DH. Stimuli-responsive liposomes for drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017;9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 290]  [Cited by in RCA: 310]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
57.  des Rieux A, Fievez V, Garinot M, Schneider YJ, Préat V. Nanoparticles as potential oral delivery systems of proteins and vaccines: a mechanistic approach. J Control Release. 2006;116:1-27.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 884]  [Cited by in RCA: 834]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
58.  Shakweh M, Ponchel G, Fattal E. Particle uptake by Peyer's patches: a pathway for drug and vaccine delivery. Expert Opin Drug Deliv. 2004;1:141-163.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 115]  [Cited by in RCA: 109]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
59.  Porter CJ, Charman WN. Intestinal lymphatic drug transport: an update. Adv Drug Deliv Rev. 2001;50:61-80.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 253]  [Cited by in RCA: 252]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
60.  Low PS, Henne WA, Doorneweerd DD. Discovery and development of folic-acid-based receptor targeting for imaging and therapy of cancer and inflammatory diseases. Acc Chem Res. 2008;41:120-129.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 859]  [Cited by in RCA: 867]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
61.  Anselmo AC, Mitragotri S. Nanoparticles in the clinic: An update. Bioeng Transl Med. 2019;4:e10143.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 884]  [Cited by in RCA: 1016]  [Article Influence: 169.3]  [Reference Citation Analysis (0)]
62.  Noble GT, Stefanick JF, Ashley JD, Kiziltepe T, Bilgicer B. Ligand-targeted liposome design: challenges and fundamental considerations. Trends Biotechnol. 2014;32:32-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 297]  [Cited by in RCA: 370]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
63.  Saraf S, Jain SK. pH-sensitive liposomes bearing a chemotherapeutic agent and a natural apoptosis modulator for effective intracellular delivery to the solid tumor. Drug Deliv Transl Res. 2023;13:2961-2981.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
64.  Bulbake U, Doppalapudi S, Kommineni N, Khan W. Liposomal Formulations in Clinical Use: An Updated Review. Pharmaceutics. 2017;9:12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1323]  [Cited by in RCA: 1313]  [Article Influence: 164.1]  [Reference Citation Analysis (0)]
65.  Łukawski M, Dałek P, Borowik T, Foryś A, Langner M, Witkiewicz W, Przybyło M. New oral liposomal vitamin C formulation: properties and bioavailability. J Liposome Res. 2020;30:227-234.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
66.  Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, Samiei M, Kouhi M, Nejati-Koshki K. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8:102.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2314]  [Cited by in RCA: 2079]  [Article Influence: 173.3]  [Reference Citation Analysis (0)]
67.  Li X, Tang C, Salama M, Xia M, Huang X, Sheng L, Cai Z. Encapsulation efficiency and oral delivery stability of chitosan-liposome-encapsulated immunoglobulin Y. J Food Sci. 2022;87:1708-1720.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
68.  Abdulkarim M, Agulló N, Cattoz B, Griffiths P, Bernkop-Schnürch A, Borros SG, Gumbleton M. Nanoparticle diffusion within intestinal mucus: Three-dimensional response analysis dissecting the impact of particle surface charge, size and heterogeneity across polyelectrolyte, pegylated and viral particles. Eur J Pharm Biopharm. 2015;97:230-238.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 95]  [Cited by in RCA: 110]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
69.  Zabaleta V, Ponchel G, Salman H, Agüeros M, Vauthier C, Irache JM. Oral administration of paclitaxel with pegylated poly(anhydride) nanoparticles: permeability and pharmacokinetic study. Eur J Pharm Biopharm. 2012;81:514-523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 98]  [Cited by in RCA: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
70.  Subramanian DA, Langer R, Traverso G. Mucus interaction to improve gastrointestinal retention and pharmacokinetics of orally administered nano-drug delivery systems. J Nanobiotechnology. 2022;20:362.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 94]  [Reference Citation Analysis (0)]
71.  Takeuchi H, Thongborisute J, Matsui Y, Sugihara H, Yamamoto H, Kawashima Y. Novel mucoadhesion tests for polymers and polymer-coated particles to design optimal mucoadhesive drug delivery systems. Adv Drug Deliv Rev. 2005;57:1583-1594.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 253]  [Cited by in RCA: 249]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
72.  Tan SLJ, Billa N. Improved Bioavailability of Poorly Soluble Drugs through Gastrointestinal Muco-Adhesion of Lipid Nanoparticles. Pharmaceutics. 2021;13:1817.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
73.  Mura S, Hillaireau H, Nicolas J, Kerdine-Römer S, Le Droumaguet B, Deloménie C, Nicolas V, Pallardy M, Tsapis N, Fattal E. Biodegradable nanoparticles meet the bronchial airway barrier: how surface properties affect their interaction with mucus and epithelial cells. Biomacromolecules. 2011;12:4136-4143.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 81]  [Cited by in RCA: 92]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
74.  Beloqui A, des Rieux A, Préat V. Mechanisms of transport of polymeric and lipidic nanoparticles across the intestinal barrier. Adv Drug Deliv Rev. 2016;106:242-255.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 76]  [Cited by in RCA: 98]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
75.  Takeuchi H, Matsui Y, Sugihara H, Yamamoto H, Kawashima Y. Effectiveness of submicron-sized, chitosan-coated liposomes in oral administration of peptide drugs. Int J Pharm. 2005;303:160-170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 151]  [Cited by in RCA: 124]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
76.  Zhang XY, Lu WY. Recent advances in lymphatic targeted drug delivery system for tumor metastasis. Cancer Biol Med. 2014;11:247-254.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 33]  [Reference Citation Analysis (0)]
77.  Ling SS, Magosso E, Khan NA, Yuen KH, Barker SA. Enhanced oral bioavailability and intestinal lymphatic transport of a hydrophilic drug using liposomes. Drug Dev Ind Pharm. 2006;32:335-345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 57]  [Cited by in RCA: 62]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
78.  Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016;99:28-51.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1900]  [Cited by in RCA: 2898]  [Article Influence: 322.0]  [Reference Citation Analysis (0)]
79.  Puri A, Loomis K, Smith B, Lee JH, Yavlovich A, Heldman E, Blumenthal R. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to clinic. Crit Rev Ther Drug Carrier Syst. 2009;26:523-580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 801]  [Cited by in RCA: 601]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
80.  Wiley DT, Webster P, Gale A, Davis ME. Transcytosis and brain uptake of transferrin-containing nanoparticles by tuning avidity to transferrin receptor. Proc Natl Acad Sci U S A. 2013;110:8662-8667.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 300]  [Cited by in RCA: 366]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
81.  Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol. 2019;181:101665.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 118]  [Cited by in RCA: 221]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
82.  Omo-Lamai S, Nong J, Savalia K, Kelley BJ, Wu J, Esteves-Reyes S, Chase LS, Muzykantov VR, Marcos-Contreras OA, Dollé JP, Smith DH, Brenner JS. Targeting of nanoparticles to the cerebral vasculature after traumatic brain injury. PLoS One. 2024;19:e0297451.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
83.  Farokhzad OC, Jon S, Khademhosseini A, Tran TN, Lavan DA, Langer R. Nanoparticle-aptamer bioconjugates: a new approach for targeting prostate cancer cells. Cancer Res. 2004;64:7668-7672.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 677]  [Cited by in RCA: 606]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
84.  Han J, Lim J, Wang CJ, Han JH, Shin HE, Kim SN, Jeong D, Lee SH, Chun BH, Park CG, Park W. Lipid nanoparticle-based mRNA delivery systems for cancer immunotherapy. Nano Converg. 2023;10:36.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 40]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
85.  Park JW, Hong K, Carter P, Asgari H, Guo LY, Keller GA, Wirth C, Shalaby R, Kotts C, Wood WI. Development of anti-p185HER2 immunoliposomes for cancer therapy. Proc Natl Acad Sci U S A. 1995;92:1327-1331.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 196]  [Cited by in RCA: 181]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
86.  Jiang Q, Yin J, Chen J, Ma X, Wu M, Liu G, Yao K, Tan B, Yin Y. Mitochondria-Targeted Antioxidants: A Step towards Disease Treatment. Oxid Med Cell Longev. 2020;2020:8837893.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 120]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
87.  Murphy MP. Targeting lipophilic cations to mitochondria. Biochim Biophys Acta. 2008;1777:1028-1031.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 470]  [Cited by in RCA: 434]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
88.  Smith RA, Murphy MP. Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann N Y Acad Sci. 2010;1201:96-103.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 434]  [Cited by in RCA: 425]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
89.  Kelso GF, Porteous CM, Coulter CV, Hughes G, Porteous WK, Ledgerwood EC, Smith RA, Murphy MP. Selective targeting of a redox-active ubiquinone to mitochondria within cells: antioxidant and antiapoptotic properties. J Biol Chem. 2001;276:4588-4596.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 815]  [Cited by in RCA: 872]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
90.  Pathak RK, Kolishetti N, Dhar S. Targeted nanoparticles in mitochondrial medicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2015;7:315-329.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 118]  [Cited by in RCA: 101]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
91.  Yue C, Yang Y, Zhang C, Alfranca G, Cheng S, Ma L, Liu Y, Zhi X, Ni J, Jiang W, Song J, de la Fuente JM, Cui D. ROS-Responsive Mitochondria-Targeting Blended Nanoparticles: Chemo- and Photodynamic Synergistic Therapy for Lung Cancer with On-Demand Drug Release upon Irradiation with a Single Light Source. Theranostics. 2016;6:2352-2366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 93]  [Cited by in RCA: 127]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
92.  James AM, Cochemé HM, Smith RA, Murphy MP. Interactions of mitochondria-targeted and untargeted ubiquinones with the mitochondrial respiratory chain and reactive oxygen species. Implications for the use of exogenous ubiquinones as therapies and experimental tools. J Biol Chem. 2005;280:21295-21312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 275]  [Cited by in RCA: 290]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
93.  Szeto HH, Schiller PW. Novel therapies targeting inner mitochondrial membrane--from discovery to clinical development. Pharm Res. 2011;28:2669-2679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 123]  [Cited by in RCA: 137]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
94.  Zhao K, Zhao GM, Wu D, Soong Y, Birk AV, Schiller PW, Szeto HH. Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J Biol Chem. 2004;279:34682-34690.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 568]  [Cited by in RCA: 619]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
95.  Mitchell W, Ng EA, Tamucci JD, Boyd KJ, Sathappa M, Coscia A, Pan M, Han X, Eddy NA, May ER, Szeto HH, Alder NN. The mitochondria-targeted peptide SS-31 binds lipid bilayers and modulates surface electrostatics as a key component of its mechanism of action. J Biol Chem. 2020;295:7452-7469.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 71]  [Cited by in RCA: 86]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
96.  Szeto HH. First-in-class cardiolipin-protective compound as a therapeutic agent to restore mitochondrial bioenergetics. Br J Pharmacol. 2014;171:2029-2050.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 360]  [Cited by in RCA: 417]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
97.  Kuang X, Zhou S, Guo W, Wang Z, Sun Y, Liu H. SS-31 peptide enables mitochondrial targeting drug delivery: a promising therapeutic alteration to prevent hair cell damage from aminoglycosides. Drug Deliv. 2017;24:1750-1761.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 30]  [Cited by in RCA: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
98.  Cantó C, Menzies KJ, Auwerx J. NAD(+) Metabolism and the Control of Energy Homeostasis: A Balancing Act between Mitochondria and the Nucleus. Cell Metab. 2015;22:31-53.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 945]  [Cited by in RCA: 1199]  [Article Influence: 119.9]  [Reference Citation Analysis (0)]
99.  Acuña-Castroviejo D, Escames G, Venegas C, Díaz-Casado ME, Lima-Cabello E, López LC, Rosales-Corral S, Tan DX, Reiter RJ. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci. 2014;71:2997-3025.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 740]  [Cited by in RCA: 778]  [Article Influence: 70.7]  [Reference Citation Analysis (0)]
100.  Yamada Y, Harashima H. MITO-Porter for Mitochondrial Delivery and Mitochondrial Functional Analysis. Handb Exp Pharmacol. 2017;240:457-472.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
101.  Prado CM, Landi F, Chew STH, Atherton PJ, Molinger J, Ruck T, Gonzalez MC. Advances in muscle health and nutrition: A toolkit for healthcare professionals. Clin Nutr. 2022;41:2244-2263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 132]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
102.  Wongprayoon P, Govitrapong P. Melatonin as a mitochondrial protector in neurodegenerative diseases. Cell Mol Life Sci. 2017;74:3999-4014.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 58]  [Cited by in RCA: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
103.  Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev. 2006;58:1532-1555.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 936]  [Cited by in RCA: 847]  [Article Influence: 44.6]  [Reference Citation Analysis (0)]
104.  Smith SA, Selby LI, Johnston APR, Such GK. The Endosomal Escape of Nanoparticles: Toward More Efficient Cellular Delivery. Bioconjug Chem. 2019;30:263-272.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 238]  [Cited by in RCA: 445]  [Article Influence: 63.6]  [Reference Citation Analysis (0)]
105.  Viricel W, Mbarek A, Leblond J. Switchable Lipids: Conformational Change for Fast pH-Triggered Cytoplasmic Delivery. Angew Chem Int Ed Engl. 2015;54:12743-12747.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 55]  [Cited by in RCA: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
106.  Wagner E, Plank C, Zatloukal K, Cotten M, Birnstiel ML. Influenza virus hemagglutinin HA-2 N-terminal fusogenic peptides augment gene transfer by transferrin-polylysine-DNA complexes: toward a synthetic virus-like gene-transfer vehicle. Proc Natl Acad Sci U S A. 1992;89:7934-7938.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 503]  [Cited by in RCA: 445]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
107.  Meers P. Enzyme-activated targeting of liposomes. Adv Drug Deliv Rev. 2001;53:265-272.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 126]  [Cited by in RCA: 104]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
108.  Kim HS, Lee DY. Nanomedicine in Clinical Photodynamic Therapy for the Treatment of Brain Tumors. Biomedicines. 2022;10:96.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
109.  Lin X, Chen M, Rodriguez Gonzalez P, Danino D, Corke H. Advancing coenzyme Q10 delivery with plant protein-based nanoparticle-mediated nanosuspensions. Food Res Int. 2024;197:115120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
110.  Zozina VI, Covantev S, Goroshko OA, Krasnykh LM, Kukes VG. Coenzyme Q10 in Cardiovascular and Metabolic Diseases: Current State of the Problem. Curr Cardiol Rev. 2018;14:164-174.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 127]  [Cited by in RCA: 103]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
111.  Hong R, Han Y, Chen S. Advances in micro- and nano- delivery systems for increasing the stability, bioavailability and bioactivity of coenzyme Q(10). Crit Rev Food Sci Nutr. 2025;1-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
112.  Pozos-Nonato S, Domínguez-Delgado CL, Campos-Santander KA, Benavides AA, Pacheco-Ortin SM, Higuera-Piedrahita RI, Resendiz-González G, Molina-Trinidad EM. Novel Nanotechnological Strategies for Skin Anti-aging. Curr Pharm Biotechnol. 2023;24:1397-1419.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
113.  Mortensen SA, Rosenfeldt F, Kumar A, Dolliner P, Filipiak KJ, Pella D, Alehagen U, Steurer G, Littarru GP; Q-SYMBIO Study Investigators. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: results from Q-SYMBIO: a randomized double-blind trial. JACC Heart Fail. 2014;2:641-649.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 238]  [Cited by in RCA: 300]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
114.  Porter CJ, Charman SA. Lymphatic transport of proteins after subcutaneous administration. J Pharm Sci. 2000;89:297-310.  [PubMed]  [DOI]  [Full Text]
115.  Frazer DM, Anderson GJ. The orchestration of body iron intake: how and where do enterocytes receive their cues? Blood Cells Mol Dis. 2003;30:288-297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 146]  [Cited by in RCA: 142]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
116.  Jaishankar M, Tseten T, Anbalagan N, Mathew BB, Beeregowda KN. Toxicity, mechanism and health effects of some heavy metals. Interdiscip Toxicol. 2014;7:60-72.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2475]  [Cited by in RCA: 2475]  [Article Influence: 225.0]  [Reference Citation Analysis (0)]
117.  Flora SJ, Pachauri V. Chelation in metal intoxication. Int J Environ Res Public Health. 2010;7:2745-2788.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 648]  [Cited by in RCA: 573]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
118.  Kim JJ, Kim YS, Kumar V. Heavy metal toxicity: An update of chelating therapeutic strategies. J Trace Elem Med Biol. 2019;54:226-231.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 225]  [Cited by in RCA: 292]  [Article Influence: 48.7]  [Reference Citation Analysis (0)]
119.   Toxicological Profile for Lead. Atlanta (GA): Agency for Toxic Substances and Disease Registry (US); 2020 Aug- .  [PubMed]  [DOI]
120.  Sears ME. Chelation: harnessing and enhancing heavy metal detoxification--a review. ScientificWorldJournal. 2013;2013:219840.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 153]  [Cited by in RCA: 156]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
121.  Ganz T. Macrophages and systemic iron homeostasis. J Innate Immun. 2012;4:446-453.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 169]  [Cited by in RCA: 212]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
122.  Yamada Y, Harashima H. Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliv Rev. 2008;60:1439-1462.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 187]  [Cited by in RCA: 197]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]