Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Progress in the design of ascorbic acid derivative-mediated drug delivery

Caroline Lamieac, Enas Elmowafy*b, Dalia A. Attiaa and Nahed D. Mortadab
aDepartment of Pharmaceutics and Pharmaceutical Technology, The British University in Egypt, Cairo, Egypt 11837
bDepartment of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Monazzamet Elwehda Elafrikeya Street, Abbaseyya, Cairo, Egypt 11566. E-mail: Enasmostafa@pharma.asu.edu.eg; Tel: (+202)01140380412
cDrug Discovery, Delivery and Patient Care (DDDPC), School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston Upon Thames, Surrey, KT1 2EE, UK

Received 30th May 2025 , Accepted 23rd September 2025

First published on 8th October 2025


Abstract

Antioxidant-based pharmaceutical products are currently experiencing a surge in popularity and satisfaction, demonstrating promising preclinical and clinical prospects. These products exert their beneficial effects by displaying protection against mischievous free radicals. One potent antioxidant is ascorbic acid (AA), which plays numerous crucial biochemical roles and is typically distinguished as a primary hydrophilic, non-enzymatic antioxidant in tissues. AA is a water-soluble essential antioxidant vitamin that can only be obtained from the diet. However, AA's instability, coupled with challenges related to its delivery, has presented formulation challenges for chemists. As a result, various stable hydrophilic and lipophilic derivatizations of AA have been devised. Capitalizing on their potential, delivery platforms, particularly nano-sized ones utilizing ascorbic acid derivatives, have been extensively investigated in recent years. Two such derivatives, namely, ascorbyl-6-palmitate (AP; a lipophilic derivative) and ascorbyl-2-glucoside (AA-2G; a hydrophilic derivative), have been extensively studied in previous works. Herein, the scientific data related to their utilization, either as a drug or as an integral component in delivery vehicles, and their pharmaceutical applications are evaluated.


1. Introduction

The term “antioxidant” is widely used and can be broadly depicted as an agent that considerably decreases or prevents the oxidation of oxidizable substrates; it is usually administered at a low concentration relative to that of the substrates.1,2 Generally, antioxidants exert their protective effects employing the following mechanisms: (i) scavenging of free radicals to neutralize and remove them, preventing oxidative damage; (ii) inactivation of peroxides and other reactive oxygen species, inhibiting their formation or neutralizing their reactivity; (iii) metal chelation, rendering pro-oxidant metal ions unavailable for harmful Fenton-type reactions; and (iv) quenching of subsequent lipid oxidation products, interrupting the chain reaction of lipid peroxidation. Through these diverse mechanisms, antioxidants play a pivotal part in defending biological systems from the deleterious effects of oxidative stress, preserving cellular integrity and function.3–6 Oxidative stress stems from an imbalance between the output of reactive free radicals and the body's ability to neutralize them, leading to potential cellular damage (Scheme 1).7–9
image file: d5ra03825a-s1.tif
Scheme 1 Balance between oxidative stress and antioxidant activity, highlighting the crucial role of AA and its derivatives. The left panel depicts the impact of oxidative stress, marked by an imbalance between the levels of reactive oxygen species (ROS) and antioxidants within the body. The central panel emphasizes the antioxidant properties of AA, explaining how it works to neutralize free radicals and mitigate oxidative damage. The right panel showcases various AA derivatives, including ascorbyl-6-palmitate and ascorbyl-2-glucoside, alongside innovative nano vesicular systems, such as aspasomes and glucospanlastics. Collectively, this figure underscores the importance of AA and its derivatives in combating oxidative stress and enhancing therapeutic efficacy through advanced delivery systems.

The antioxidant defense against oxidative stress can be done enzymatically and non-enzymatically. The primary enzymatic repairing systems include superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase.10,11 Conversely, the non-enzymatic systems include compounds that are able to scavenge the free radicals, such as vitamin C, carotenes, vitamin E, ferritin, selenium, melatonin, zinc, flavonoids, and cysteine.12–14 Current biomedicine theories suggest that oxidative stress is a key pathophysiological factor contributing to significant diseases related to stress and aging, such as cancer, lung disorders, and cardiovascular conditions.9,15–19

Vitamin C or L-ascorbic acid (L-AA) is a water-soluble, non-enzymatic antioxidant. It appears as a white or pale-yellow powder or powdery crystal with a slightly acidic taste. AA is freely soluble in water, slightly soluble in alcohol, and insoluble in chloroform and ether. The term “vitamin C” is used to refer AA in chemistry, which has a molecular weight of 176.13 Da and empirical formula of C6H8O6 (Fig. 1A). AA is a highly effective free radical scavenger in aqueous media, but it exhibits a relatively low antioxidant effect in hydrophobic phases.20 It has been identified as a potent skincare agent with considerable whitening and anti-wrinkle effects.21,22


image file: d5ra03825a-f1.tif
Fig. 1 The structure of some representative derivatives of ascorbic acid. (A) Ascorbic acid, (B) ascorbyl 6-palmitate, (C) ascorbyl 2,6-palmitate, (D) ascorbyl 6-octanoate, (E) ascorbyl 6-stearate, (F) ascorbyl 2-glucoside, (G) ascorbyl tetra-isopalmitate, (H) ascorbyl 2-O coumarate, and (I) ascorbyl 3-O coumarate.

Interestingly, AA is utilized as a drug or functional material in the pharmaceutical scene.23–27 However, the exploitation of AA as a drug is highly challenging due to its instability.24,28–32 Several strategies have been developed to overcome such challenges, including its incorporation in an assortment of delivery platforms. Instead, the synthesis of more stable AA derivatives with different chemical characteristics has been attempted, outperforming AA and showing better beneficial biological prospects.33–35 Beyond their non-derivatized types, both hydrophilic and lipophilic AA derivatives have been employed for the integration of novel multi-functional formulations in diverse pharmaceuticals. Clinically, such a perspective could highlight the relevance of such derivatives as therapeutics and as competent carriers, supporting nanotechnology and drug delivery in general.

Working on such considerations, the current review explores the exploitation and recent advances of AA derivatives in the pharmaceutical arena. Information was collected about the employed AA derivatives regarding their chemical nature and related features. Of note, two lipophilic and hydrophilic derivatives, namely ascorbyl 6-palmitate (AP) and ascorbyl 2-glucoside (AA-2G), were prioritized in the present review, due to their extensive utilization in drug-delivery systems. Specifically, an example of a greatly exploited nanoplatform: aspasomes (ASP) based on AP, was fully addressed, covering their delivery target and indication, composition, and efficacy.

2. Beneficial effects and roles of ascorbic acid in the body

AA is a crucial nutrient that performs an assortment of physiological tasks. It participates in the repair and maintenance of diverse tissues throughout the body. Additionally, AA is involved in the enzymatic production of certain neurotransmitters, highlighting its importance in supporting neurological function.36–41 As a potent antioxidant, mitigating oxidative stress, which is the key driver of inflammation, AA exhibits strong anti-inflammatory properties through several mechanisms.42–47 Recently, it has been identified that AA may be a beneficial adjunct therapy for inflammatory bowel diseases due to its anti-inflammatory properties, immune support, and potential to promote gut health.48,49

In the context of cutaneous physiology, AA is valuable for the biosynthesis and retention of collagen, a critical structural protein that provides strength, elasticity, and integrity to the dermal layer of the skin. Beyond its structural and antioxidant functions, AA also exhibits profound modulatory effects on key cellular signaling pathways governing skin cell growth and differentiation. Intriguingly, emerging research has pinpointed the potential of AA in the context of skin cancer, demonstrating its ability to reduce the viability and invasive potential of certain cutaneous malignant cells.50–52 Several dermatological clinical trials have investigated the role of AA in the remediation of skin diseases, such as atopic dermatitis,53 herpes zoster,54 malignant melanoma,55 as adjuvant therapy in acne,56 allergic contact dermatitis,57 and psoriasis.58 AA offers a multitude of additional benefits for cutaneous health and function. As a photoprotective agent, it can shield the skin from the deleterious influences of UVA and UVB radiation, mitigating the harmful impacts of sun exposure.59,60 The vitamin also promotes neocollagenesis, stimulating the synthesis of new collagen fibers to support dermal structure and elasticity.61,62 Moreover, AA exhibits the power to suppress melanogenesis, making it a valuable ingredient in skin lightening and depigmentation applications.63–65 It has also been studied for its curative competence in the management and control of various inflammatory skin ailments46,66 (Fig. 2).


image file: d5ra03825a-f2.tif
Fig. 2 Diagram representing the role of ascorbic acid in the skin. In the context of cutaneous physiology, AA is substantial for the following roles: (i) biosynthesis of collagen, supporting dermal structure and elasticity, (ii) suppression of melanogenesis, benefiting skin lightening, whitening and depigmentation applications, (iii) exfoliation, acting as mild exfoliator due to its acidity (iv) photoprotection, shielding the skin from the deleterious influences of UVA and UVB radiation upon sun exposure, (v) anti-inflammatory potential, possessing curative competence in the management and control of various inflammatory skin ailments, and (vi) countering skin cancer and aging via mitigating oxidative stress. This figure was created with BioRender.

AA has been demonstrated to have direct anti-tumor potentials, with the ability to induce apoptosis (programmed cell death) and suppress the proliferation of an assortment of cancer cell types.67–71 AA can also exert indirect anti-cancer impacts by enhancing the mission of the immune system, promoting the differentiation, proliferation, and activation of various immune cell types.72,73 AA can increase the cytotoxicity and cytokine production of natural killer (NK) cells, being crucial for the immune-mediated termination of cancerous cells.74–76

Collectively, the multifaceted benefits of AA, ranging from photoprotection and collagen synthesis to melanogenesis suppression and potential anti-cancer applications, solidify its status as a versatile and indispensable nutrient for maintaining optimal body health and function.

3. Ascorbic acid utilization in delivery systems

Importantly, benefiting from its anti-oxidant potential, AA has been exploited as a drug and incorporated in delivery systems to tackle its instability. Indeed, AA is highly susceptible to degradation in aqueous environments, particularly at elevated pH levels, in the presence of oxygen, and when exposed to metal ions, often leading to a noticeable color change in AA-containing formulations.28,77–79

To overcome these stability challenges and harness the benefits of AA, researchers have incorporated it into various delivery approaches aimed at protecting the vitamin from the factors that contribute to its degradation.80–82 As a consequence, AA is formulated in an assortment of delivery modes for various administration routes, specifically the topical route.83–88 Interestingly, the incorporation of AA along with various drugs and bioactive compounds such as oxaliplatin and olaparib into nanoplatforms has emerged as a promising strategy to boost their therapeutic performance across different applications.89–91

On the other hand, lately, AA has been utilized as a functional component in delivery systems. It is exploited as a reducing agent in the synthesis of metallic nanoparticles, including silver, gold, selenium, iron oxide and copper ones.26,92–96 AA is also employed to modify the surfaces of different NPs, imparting desirable merits to the surface-modified ones, such as titanium dioxide NPs.26 In cosmetic-directed research, for modulating skin hyperpigmentation and whitening, AA has been incorporated in various nanoplatforms, such as nanoemulsions,97 and liposomes98 and ethyl cellulose NPs in hydroxypropyl methyl gels.99

4. Exploitation of ascorbic acid derivatives in the pharmaceutical arena

As aforementioned, one of the greatest challenges in the exploitation of AA is maintaining its stability. The high aqueous solubility and inherent instability of AA, particularly in the presence of factors, such as oxygen, pH changes, and metal ions, can limit its effective administration and formulation. To address this challenge, lately, researchers have developed striking strategies to limit the degradation processes that affect AA, including the use of stable derivatives, which exhibit enhanced stability compared to the parent compound.100–102 To address the stability challenges of AA, researchers have synthesized a wide range of stable hydrophilic and lipophilic derivatives of the vitamin. The preponderance of these derivatives over AA has been verified, concerning their stability and functionality (Fig. 3).
image file: d5ra03825a-f3.tif
Fig. 3 Comprehensive overview highlights the significance of ascorbic acid derivatives in pharmaceutical and drug delivery applications. The central circle identifies ascorbic acid as the primary component, emphasizing its inherent instability and high aqueous solubility. Surrounding this core are AA derivatives designed to enhance the stability and efficacy of ascorbic acid. The outer ring illustrates the therapeutic potential of these derivatives, detailing their roles as antioxidants, antiviral agents, anti-inflammatory compounds, anti-cancer agents, and antimicrobial substances. Additionally, these derivatives serve as functional excipients, such as solubilizers, emulsifiers, and stabilizers, and play a crucial role in the formation of nanoparticles, including asposomes and glucospanlastics. This figure was created with BioRender.

The structural alteration of the AA ring in position 2, 3, 5 or 6 to produce different AA derivatives not only improves its stability but also maintains its antioxidant activity. Representative examples of such AA derivatives are shown in Fig. 1. One example of a water-soluble AA derivative, ascorbyl-2-glucoside (AA-2G) or 2-O-glucopyranosyl-AA, with an added glucose moiety bound to AA, was synthesized and revealed to prevent AA oxidative degradation.103,104 Other lipophilic AA derivatives have also been synthesized via the esterification of AA with various acids. Ascorbyl 6-octanoate is formed by the esterification of AA with the 6-carbon saturated fatty acid, octanoic acid (also known as caprylic acid). Ascorbyl-2,6-dipalmitate105 and ascorbyl-6-palmitate are fat-soluble esters of AA and palmitic acid, possessing good skin penetration and antioxidant activities.106,107 3-O-Ethyl-L-ascorbyl-6-ferulate is formed by the esterification of AA at the C-3 position with ferulic acid (a phenolic compound in plant cell walls possessing antioxidant potential). 3-O-Ethyl-L-ascorbyl-6-palmitate is formed by the esterification of palmitic acid at the C-6 position, and there is an ethyl group attached at the C-3 position of the AA molecule. It was found that the encapsulation of these novel AA esters within lignin-based nanoparticles resulted in a boost to the antioxidant power and in the prevention of the degradation of AA compounds.108

Ascorbyl-6-stearate, in which the stearic acid is attached to the C-6 hydroxyl group of the AA molecule, was found to be similar to ascorbyl 6-palmitate and verified to have anti-tumor activity.109 Ascorbyl 2-O-coumarate and ascorbyl 3-O-coumarate are formed via the esterification of the C-2 or C-3 hydroxyl group of the AA molecule and coumaric acid.22 Ascorbyl tetraisopalmitate is a liquid form of AA. Four isopalmitic acid molecules are attached to the AA at different hydroxyl groups (likely the 2-, 3-, 5-, and 6-positions). The multiple esterification steps enhance the lipophilicity and stability of AA.22

Indeed, the newly designed AA derivatives have secured a substantial place in pharmaceutical research, breaking new ground in the pharmaceutical industry and redirecting toward integrative approaches, which necessitate collaboration among the chemical, pharmaceutical, and pharmacology fields, to elucidate their full potential. AA derivatives offer a promising therapeutic framework for enriching the drug-delivery arena.

AA derivative-based therapies could possess a wide-ranging leverage on diversified ailments affected by oxidative stress. AA derivatives can provide a therapeutic avenue for the treatment of oxidative stress comorbidities.

Earlier work has demonstrated that AA-2G can inhibit Helicobacter pylori-induced apoptosis in epithelial cells of the stomach through a mitochondrial pathway.110,111 Similarly, in the small intestine, the derivative promotes expansion of Muribaculaceae and modulates Paenibacillus populations, suggesting a comprehensive approach to restoring gut ecosystem balance.112 Beyond direct microbiota modulation, AA-2G has been shown to ameliorate high fructose-induced neuroinflammation through gut microbiota and leaky gut mechanisms, demonstrating the compound's ability to influence the gut–brain axis and prevent diet-induced metabolic dysfunction.

Additionally, the derivative 2-O-β-D-glucopyranosyl-L-AA has been shown to exert immunomodulatory effects and positively influence gut microbiota composition.110,113 Studies indicate that this compound can alleviate conditions, such as colitis and neuroinflammation linked to high fructose intake, suggesting its role in maintaining gut integrity and function.

Considerably, some AA derivatives have been well-documented to exert a complex and multifaceted role in the regulation of melanogenesis, demonstrating both stimulatory and inhibitory impacts related to the specific compound and experimental conditions. The study found that various AA derivatives, such as AA-2G and 3-O-ethyl L-AA, were able to inhibit the autoxidation of L-DOPA in vitro. Additionally, magnesium L-ascorbyl 2-phosphate and L-cysteine were shown to attenuate the cytotoxicity induced by high concentrations of L-DOPA. Interestingly, while magnesium L-ascorbyl 2-phosphate and AA-2G increased the intracellular levels of melanin and the darkly pigmented cell count, the cysteine derivative L-cysteinamide exhibited the opposite effect, decreasing both the melanin levels and the count of pigmented cells.114,115

One of the novel AA derivatives is the 1,2,3-triazole L-AA (L-ASA) conjugate, which was synthesized and evaluated in one study.116 This compound features a p-bromophenyl substituent on the 1,2,3-triazole moiety, connected to the L-ASA core via a hydroxyethylene linker. In the antiproliferative evaluation, compound 4e demonstrated selective cytotoxicity against breast adenocarcinoma MCF-7 cells, exhibiting an IC50 value of 6.72 μM. Importantly, L-ASA did not inhibit the growth of foreskin fibroblasts, indicating its selective antiproliferative power. Such promising results highlight that the potent growth-inhibition of 4e on the MCF-7 breast cancer cells may involve the modulation of the HIF-1 signaling pathway.

Hydrophilic AA derivatives also demonstrate improved intracellular uptake compared to AA, as the modified compounds can more readily permeate the cell membrane and accumulate within the cells.117 Interestingly, the topical application of AA glucosides is well-documented to constantly release the active form of AA through enzymatic hydrolysis in the skin.118 In general, developing multifunctional hybrid materials via the covalent combination of two substances with featured biological properties can provide additional advantages compared to the individual components.119

Hydrophobic derivatives excel at preventing diverse harmful effects induced by X-ray radiation. AP, demonstrating high free radical-scavenging abilities, more effectively suppressed X-ray-induced increases in intracellular ROS levels and lipid peroxidation in keratinocytes, and it significantly prevented X-ray-induced apoptosis. The studies presented here highlight the superior antioxidant and protective properties of lipophilic VC derivatives. The lipophilic AP exhibits enhanced activities compared to AA. AP has demonstrated higher free radical-scavenging abilities, more effective suppression of X-ray-induced oxidative damage, and better prevention of radiation-induced cell death in human cells, likely due to its improved lipophilicity.120 It potently inhibits the activity of 5-lipoxygenase, an enzyme participating in inflammatory pathways, more than 15-lipoxygenase, and this inhibitory effect is stronger than those of known synthetic drugs. Correspondingly, enzyme kinetics and molecular docking analyses have revealed AP as a non-competitive, reversible 5-lipoxygenase inhibitor, highlighting the importance of its lipophilic nature.121 The enhanced lipophilicity conferred by the palmitate moiety likely contributes to AP's improved protective effects compared to the more hydrophilic AA, suggesting that the lipophilic VC derivative could be a promising countermeasure against radiation-induced oxidative damage.

5. Characteristic methodologies of ascorbic acid derivatives as functional components of delivery systems

As therapeutic entities, a series of structural derivatives of AA bearing different moieties have been synthesized using diverse chemical approaches, offering strikingly advanced scaffolds mostly for the design of antioxidant, anti-inflammatory and anticancer molecules. Indeed, continued characterization and evaluation seem necessary to investigate their benefits via an assortment of preliminary and screening tests.

In order to demonstrate their stability and hence supremacy over AA, the stability of the fabricated derivatives in aqueous solution was tested by dissolving them in a suitable buffer and storing at 37 °C for a specified period, followed by measuring the three obtained concentrations and comparing them with the initial concentration.122–125

Importantly, the structural and conformational properties of the synthesized derivatives were elucidated to identify their chemical entities using 1H and 13C NMR.126,127 Other solid-state depiction tests, such as DSC, FTIR and X-ray diffraction, were also carried out. Considerably, structure–activity relationship analysis was performed to demonstrate the impact of derivatization and substitution on their bioactive potencies.

In vitro screening of proliferation across different cancer cell lines, employing the MTT test, was carried out to demonstrate the anticancer activity of some promising derivatives. For example, the anticancer potential of the 2,3-di-O-aryl/alkyl sulfonate derivatives of L-AA on breast and lung cancer cells was investigated, showing noticeable cytotoxicity and selectivity for cancer cells.128

Another instance involves evaluating the antiproliferative effectiveness of 1,2,3-triazolyl-L-AA derivatives on various malignant tumor cell lines, including cervical carcinoma (HeLa), breast adenocarcinoma (MCF-7), colorectal carcinoma (HCT-116), lung adenocarcinoma (A549), ductal pancreatic adenocarcinoma (CFPAC-1), hepatocellular carcinoma (HepG2), colorectal adenocarcinoma, and metastatic cells (SW620). The findings indicated that some synthesized derivatives exhibited selective antiproliferative activity against HeLa, HCT-116, and MCF-7 cells, as well as A549 cells, which appeared to be associated with the nature of their substitutions. Additionally, an Annexin V assay was done to assess the type of cell death, particularly apoptosis, associated with the antiproliferative effects. The study measured the percentages of apoptotic cells, along with necrosis, to emphasize the significant inhibition of cell growth.116

An antioxidant potential test was also done using the commonly employed reagent, DPPH, involving incubating the tested compounds with this reagent and monitoring its reduction through measuring the resultant absorbance as well as that of DPPH alone.116 The values of IC50 (concentration causing 50% of the absorbance) and the antiradical efficiency (1000-fold inverse of IC50) were determined as indicators of the free radical-scavenging activity and compared with those for AA.129 Interestingly, the results attained in earlier studies pinpointed the formation of AA derivatives that can act as potent radical scavengers based on the substitution pattern relative to AA.130,131 It is to be noted that other radicals, such as ABTS and galvinoxyl, were also utilized in assays of the radical-scavenging activity.132

Specifically, the investigation of the binding interactions with different targets to demonstrate their mechanism of action and specificity and the prediction of their pharmacokinetic features were done utilizing in silico molecular docking simulations and ADME profiling, respectively. Investigating the binding interactions between non-pancreatic secretory phospholipase A2 and the halo 6-fatty acid esters of L-AA, as well as the 6-fatty acid esters of L-AA, which demonstrate significant interactions with secretory phospholipase A2.133 A high binding affinity of 6-amino-6-deoxy-L-AA (D1) and N-methyl-6-amino-6-deoxy-L-AA with human peroxiredoxin was also verified, and the docking score was found to be comparable to that for AA.131 Interestingly, when assessing its binding with polymorphonuclear lymphocyte 5-LOX and soybean 15-LOX, the lipophilic derivative, AA 6-palmitate, showed greater MolDock and Rerank scores than AA yet comparable scores to the reference inhibitors, pinpointing its capability to inhibit lipid peroxidation (sLOX).121

Other tests have also been conducted to verify that they fulfill the intended purpose for their manufacture, such as testing the effect of alkylglyceryl-L-AA derivatives on the melanogenesis of B16 melanoma 4A5 cells and normal melanocytes.125 The antiviral potential of some AA derivatives against various viruses, including Herpes simplex virus and Influenza viruses, was also tested.134,135

6. Selected examples of lipophilic and hydrophilic ascorbic acid derivatives

Based on their relevance in the pharmaceutical field, two AA derivatives of different natures will be discussed in this review: ascorbyl 6-palmitate (AP) as a hydrophobic derivative and ascorbyl 2-glucoside (AA-2G) as a hydrophilic derivative.

6.1. Ascorbyl 6-palmitate “AP”

Ascorbyl palmitate (AP) is a lipophilic, stable ester derivative of AA. It is an odorless white-to-yellowish powder with a slight citrus-like aroma. AP has a molecular weight of 414.53 Da.121,136 As shown in Fig. 1B, the hydroxyl group in position 6 is combined with the alkyl chain of palmitic acid to produce ascorbyl-6-palmitate.

AP is a well-recognized skin-whitening agent that enhances skin elasticity by facilitating collagen production. As a lipophilic compound, it also assists in skin penetration.137,138 Its antioxidant property stems from the formation of hydrogen peroxide, and it presents an effect on the apoptotic pathways by inducing pro-oxidant damage, which cannot be that cancerous cells cannot adapt to or resist.139 From skin-delivery perspectives, AP is added to topical preparations to safeguard against oxidative changes in dermal components and as an anti-inflammatory agent.140,141 The augmented antioxidant activity may be due to the hydrophobic palmitate chain of the AP, which has the ability to more easily penetrate cells and better preserve lipids and other cellular components from peroxidation induced by free radicals.142,143

Given the excellent preservation of AA's antioxidant properties, AP has shown effectiveness in reducing cellular reactive oxygen species levels after exposure to ultraviolet light, as stated earlier by Meves et al.136 This makes AP a valuable active oxygen scavenger that can protect the skin against oxidative damage.144 Owing to its lipophilic nature, AP has exhibited remarkable in vitro skin retention and penetration, particularly in the epidermis, as verified by in vivo skin-delivery investigations.145

Interestingly, AP has been thoroughly reported to preserve the anti-cancer properties of AA, functioning as an inhibitor of DNA replication and cell growth in different cancer cells, including skin cancer.146,147 D'Souza et al. investigated AP anticancer activity, incorporating AP in liposome nanoparticles and evaluating its toxicity against various cell lines (Human ovarian carcinoma A2780, renal adenocarcinoma ACHN, breast tumor cells MCF7 and BT20 and mouse renal carcinoma line RAG). It was revealed that the cell death process was due to the generation of oxygen reactive species.148

Abdel-hady et al. explored a new application of AP in tissue engineering due to its osteogenic (bone-forming) effect. The combination of AP and strontium polyphosphate nanoparticles within the fiber mats exhibited synergistic effects, further enhancing the osteogenic potential of the biomaterial. The presence of AP was found to enhance the osteogenic differentiation of human mesenchymal stem cells that were seeded on the fiber mats. This is a novel observation highlighting the ability of AP to stimulate bone-forming cell behavior.149

AP has been revealed as a potent antimicrobial agent with superior antibacterial activity against Helicobacter pylori compared to AA. In contrast to AA, which paradoxically increased H. pylori survival under aerobic conditions, AP exhibited strong inhibitory effects on the bacteria under both aerobic and microaerophilic conditions.150

Furthermore, AP forms lamellar vesicles in water (called ASP), in addition to cholesterol and diacetyl phosphate, to strengthen the bilayer and stabilize the formulation.151 ASP vesicles are considered a new generation of antioxidant nano-vesicles having a hydrophilic core and lipophilic shell that can incorporate both hydrophilic and lipophilic drugs. Additionally, studies have shown that ASP vesicles have greater bilayer stability than liposomes, owing to their content of AP that achieves better skin penetrability than AA.138,152 Table 1 displays earlier works highlighting different applications of AP in the pharmaceutical and medical fields, including as a therapeutic agent, an auxiliary amphiphilic molecule in delivery platforms, and the main component of newly developed aspasomal vesicles.

Table 1 Different applications of AP in the pharmaceutical and medical fields, highlighting the versatile uses of AP in enhancing delivery, providing stability, and offering therapeutic benefits across various medical fields
Role/function of AP Delivery system/dosage form Status of investigation Application of AP Reference
Active constituent In vitro (assessed the recovery of the ascorbate radical from neural tissues susceptible to the hypoxic stimulus “cerebral cortex and carotid body”, following the administration of AP by gavage) Carrier delivery of ascorbates into “bio-membranes” neural tissues due to its lipophilicity 153
Microemulsion In vitro (determination of the effective concentrations of AP for inhibiting free radicals that were formed in porcine skin, using electron paramagnetic resonance (ESR) spectroscopy and the spin trapping approach) Skin protection against UVB irradiation using microemulsion as a carrier system 154
Cream, gel, and oil/water emulsion (O/W) In vitro (determination of skin hydrating effect in comparison to vitamin E) Skin moisturizing potential effect 155
Solid lipid nanoparticles and nanostructured lipidic carriers in hydrogels In vitro (PS, PDI determination, DSC), ex vivo (determination of penetration ability using excised human skin), in vivo (moisturizing effect testing on female Caucasian volunteers) Skin moisturizing and penetration abilities of AP have been evaluated when the compound is entrapped within solid lipid nanoparticles and when incorporated into a nanostructured lipid carrier hydrogel formulation 156
Semisolid nanostructured lipid carrier In vitro (physicochemical evaluation and release study) Antioxidant potential and permeation enhancement 157
Polyethylene glycol phosphatidyl ethanolamine micelles In vitro (physicochemical profiling and cytotoxicity assay) and in vivo (female Balb/c mice “induction of murine mammary carcinoma)” AP has demonstrated anti-cancer effectiveness against cancer cell lines, as verified by the assigned in vitro and in vivo studies 158
Liposomes In vitro (physicochemical characterization, cytotoxicity, antioxidant and antimicrobial activity) AP exhibits enhanced stability and safety, as well as antimicrobial and antioxidant activities 159
Excipient (negatively charged lipid) Lipid nanoparticles (composed of phosphatidylcholine and AP) In vitro (physical, morphological characterization and cytotoxicity assay) and in vivo (Balb/c female mice via the induction of orthotopic breast cancer models) Surface-charge modifier to enhance targeting in the induced tumor and exhibit superior tumor inhibitory potentials 160
Main component of bilayer vesicle formation ASP In vitro (physical characterization, DSC, release study, transdermal permeation, DPPH assay) and ex vivo (transdermal permeation) Ability to form a bilayer vesicular system called (ASP) 140
Hydrogel In vitro (evaluation of gel properties, including rheology, drug loading, cytotoxicity, and drug release, using an intestinal model) Hydrogel formation for rectal delivery (local, intestinal delivery of macromolecules) 161
Glossypol incorporated in AP coagel for topical formulation In vitro (evaluation of the immobilization effect of coagels on sperm motility) Coagel formation that was able to immobilize gossypol 162
Auxiliary amphiphilic molecule AP in chondroitin sulfate/chitosan nanoparticles In vitro (physical characterization, DSC, FTIR, release study, DPPH assay) Suspension for arthritis treatment via the intra-articular route 163
In vivo (male rats weighing 180–225 g)


Another derivative of AP that can form micelles in aqueous media is trisodium L-ascorbyl 2-phosphate 6-palmitate. The conjugation of trisodium salt having a phosphate group with palmitate can enhance the hydrophilicity and the chemical stability of the compound. Inoue et al. were the first to explore this derivative of AA. Their study highlighted the benefits of incorporating the trisodium L-ascorbyl 2-phosphate 6-palmitate AA derivative into a nanocarrier-based drug-delivery system for skincare applications.164

Considerably, a crucial point is that different alkyl chains can be used to form ester bonds with AA, and modifying the alkyl chain component can be used as a way to improve the surface activity of the resulting AA ester compound. For example, ascorbyl dipalmitate (ADP) is a modified AA compound that contains two palmitate chains: one at the 6 position and another at the 2 position, differentiating it from the simpler ascorbyl 6-palmitate derivative. In 2010, Moribe et al. combined the AA derivative, ascorbyl dipalmitate (ADP), with the surfactant distearoyl phosphatidyl ethanolamine–polyethylene glycol 2000 (DSPE–PEG) in molar ratios of 1[thin space (1/6-em)]:[thin space (1/6-em)]1 and 2[thin space (1/6-em)]:[thin space (1/6-em)]1 to formulate stable nanoparticles capable of encapsulating hydrophobic drugs (amphotericin B). The inclusion of ascorbyl dipalmitate was believed to contribute to the stabilization of the nanoparticle structure and provide antioxidant properties to the overall drug-delivery system.165

In 2021, the same research group investigated the high loading capacity of ascorbyl 2,6-dipalmitate in newly developed nanoparticles. They successfully obtained ADP nanoparticles when the molar ratio of ADP to DSPE–PEG (a lipid-based stabilizer) was between 5[thin space (1/6-em)]:[thin space (1/6-em)]1 and 20[thin space (1/6-em)]:[thin space (1/6-em)]1, opposing earlier works stating that molar ratios greater than 3[thin space (1/6-em)]:[thin space (1/6-em)]1 fail to form nanoparticles. Interestingly, the proposed nanoparticles were morphologically rod-shaped, possessing a size around 100 nm. According to the accomplished work, the critical ADP to DSPE–PEG molar ratio was determined to be less than 33[thin space (1/6-em)]:[thin space (1/6-em)]1. Based on previous studies, these rod-shaped ADP nanoparticles could serve as an efficient administration vehicle enclosing high-dose AA for optimal tumor-targeting in cancer therapy, showing their superior in vitro and in vivo behavior compared to spherical particles.166

Concerning the combination of palmitate-based AA esters, Plaza-Oliver et al. investigated the use of both ascorbyl 2-palmitate and ascorbyl 2,6-dipalmitate in the development of nanoemulsions containing α-tocopherol for oral delivery. The study examined the effect of the intestinal protein corona on the mucodiffusion of these types of drug-delivery systems under simulated intestinal conditions.167 When the nanoemulsions were incubated in simulated intestinal fluid, an “intestinal protein corona” (I-PC) was generated on the colloidal surface. The researchers found that the formation of this I-PC influenced the possible interaction between the proposed nanoemulsions and the intestinal mucus barrier, displacing the nanoemulsions from an “immobile-hindered” population to a mobile “diffusive” population. The study highlighted the critical impact of this I-PC on the substantial mucodiffusion properties of the formed nanoemulsions within the intestinal mucus layer, which is an important consideration for the design of promising oral nano-platforms.105

In a study published in 2021, Sonkaew et al. utilized the environmentally friendly supercritical CO2 technique to produce ascorbyl 2,6-dipalmitate (ADP) nanoparticles. The researchers found that the resulting nanoparticles containing ADP exhibited potent antioxidant activities, and these beneficial properties were maintained even after the nanoparticles were incorporated into cellulose-based packaging films.168

6.1.1. Aspasomes: an example of AP-based nanocarriers. Nano-dermatological solutions utilizing lipid-based nanocarriers, particularly safe and biocompatible nano-sized vesicular ones, could offer promising merits in dermal-oriented applications.169–172 These advantages include the facile consistency of a protective topical film on the surface of dermal layers, enhancement of skin hydration and occlusion to improve barrier function, boosting of the penetrability and deposition of payloads within the various dermal strata, and enhancement of solubilization and bioavailability of hydrophobic drugs to enable more effective delivery to the target site.169,173–176 Compared to non-lipid nanocarriers, these lipid-based systems exhibit superior performance in terms of topical formulation development, skin delivery, and optimization of the pharmacokinetic profiles of lipophilic compounds for dermatological applications.177

Of these, ASP vesicles represent a novel class of multilayered nanovesicles, with AP serving as the primary and extensively studied component of the vesicle bilayer. AP, a hydrophobic derivative of AA, has obtained FDA approval and is commonly utilized as an inactive ingredient in numerous skincare formulations.178 In the fabrication of ASP, AP is consolidated with cholesterol and charged lipids for optimal drug association and entrapment, as demonstrated in several studies.140,151,179 ASP vesicles were initially proposed in 2004 by Gopinath et al., who explored the potential of the amphiphilic AP to form bilayered vesicles using cholesterol as a stabilizing agent and dicetyl phosphate to induce charge, aiming to encapsulate the hydrophilic drug azidothymidine.140 Fig. 4 demonstrates the schematic diagram of the assembly of the main components of ASP vesicles.


image file: d5ra03825a-f4.tif
Fig. 4 Schematic diagram of the assembly of the main components of aspasomes vesicles. For the formation of aspasomes, the amphiphilic ascorbyl palmitate (AP) is consolidated with phospholipid to form bilayered vesicles. Cholesterol is added as a stabilizing agent. This figure was created with BioRender.

ASP vesicles have demonstrated several advantageous features for topical drug delivery. These include enhanced permeation through the stratum corneum, safety, and biocompatibility, as well as the use of low drug amounts while achieving therapeutically relevant concentrations in the targeted skin areas, thereby reducing the potential for systemic side effects.180,181 AP serves a dual function in ASP, performing as both a lipid bilayer-producing agent and a stabilizer, all while exhibiting its inherent antioxidant activity. The role of oxidative stress and imbalance in the antioxidant–oxidant equilibrium is well-recognized in the pathogenesis of assorted skin ailments.182 The antioxidant capacity of ASP can play a crucial part in mitigating the excessive inflammation associated with worsened skin conditions by neutralizing the involved ROS.183

Interestingly, ASP vesicles have been verified to possess superb antioxidant and dermal permeation enhancement attributes, which have enabled their use in the treatment of conditions affecting the skin, such as androgenic alopecia, acne, and psoriasis.137,179,184 Owing to the capacity of ASP vesicles to be dermally retained, the payload persistence in the target region can be anticipated and potentiated, as stated earlier.137,138 Assortments of scientific studies were conducted to incorporate various hydrophilic and lipophilic drugs in these vesicles, as demonstrated in Table 2.

Table 2 Examples of aspasomes prepared with various drugsa
Drug Drug class (indication) Route of administration Composition Status of investigation Main findings References
a PS: particle size, PDI: polydispersity index, EE: entrapment efficiency, ZP: zeta potential, *SGOT (serum glutamic oxaloacetic transaminase), *SGPT (serum glutamic pyruvic transaminase), and *TNF-α and *IL-1β are the major serum parameters for the assessment of rheumatoid arthritis in rats; *hemi-MASI (the hemi melasma area and severity index score).
Azido-thymidine (AZT) Antiretroviral medication (HIV/AIDS) Transdermal -Ascorbyl-6-palmitate In vitro -The presence of cholesterol in ASP was found to not significantly affect the vesicular size, surface charge, and drug entrapment 140
-Cholesterol -The AZT release from ASP was found to undergo a substantial change upon changing the assigned cholesterol proportion
-Dicetyl phosphate -The ASP formulation containing 45 mol% cholesterol showed the highest latency or slowest release rate of the encapsulated drug compared to other cholesterol compositions tested
  -ASP had much higher antioxidant potency than AA and improved AZT transdermal permeation
Methotrexate (MTX) Anti-rheumatic (arthritis) Topical -Ascorbyl 6-palmitate In vitro & in vivo -The optimized ASP formulation exhibited a smooth surface morphology with a particle size of 386.8 nm, appreciable drug loading of 19.41%, a negative surface charge, controlled methotrexate release over a period of 24 hours and a steady permeation rate 152
-Cholesterol -The increase in the cholesterol content was revealed to boost the loading capacity of the ASP
-1,2-Dimyristoyl-sn-glycero-3-phosphocholine (DMPC) -The in vitro drug release followed Higuchi's model and Fickian kinetics
  -The absence of skin irritation of the prepared hydrogel-loaded ASP formulation was confirmed
  -Transdermal application of a methotrexate-loaded ASP hydrogel formulation in arthritic rats for 12 days led to significant improvements in comparison to the arthritic control. Specifically, the treatment exhibited a 21.25% reduction in rat paw diameter, a 40.43% decrease in SGOT, a 54.75% decrease in SGPT, a 33.99% reduction in TNFα, a 34.79% reduction in IL-1β, an 84.41% reduction in cartilage damage, an 82.37% reduction in inflammation, an 84.38% reduction in pannus formation, and an 80.52% reduction in bone resorption
  -ASP loaded with MTX had a beneficial dual effect against conditions related to inflammation
Melatonin Natural antioxidant hormone (hair growth in androgenic alopecia (AGA)) Topical -Ascorbyl 6-palmitate In vitro, ex vivo & clinical assessment -The PS increased with increasing AP concentration 184
-Dicetyl phosphate (DCP) -The DPPH assay revealed the preservation of AP antioxidant potential
-Cholesterol -The ex vivo permeation study verified the capability of the prepared ASP to improve topical drug permeation across all skin strata, as compared to melatonin solution: a 1.34-fold greater permeation in the stratum corneum, a 2.1-fold greater permeation in the epidermis, and a 2.7-fold greater permeation in the dermis
  -Melatonin ASP showed clinically promising results in the treatment of AGA compared to the melatonin solution. The ASP-based treatment led to an increase in hair thickness and density and a decrease in hair loss, as evidenced by the obtained photographic observations of most of the treated patients
Magnesium ascorbyl phosphate (MAP) Antioxidant derivative of AA (melasma) Transdermal -Ascorbyl 6-palmitate In vitro, ex vivo & clinical assessment -The amounts of cholesterol and MAP were found to significantly increase the EE% 138
-Cholesterol -Enhanced permeation ASP over drug solution was demonstrated over the drug solution
-Lecithin -MAP aspasomal cream performed better than the gel, exhibiting the greatest improvement in the treatment of melasma, as indicated by the hemi-MASI score. Notably, 35% of sufferers rated the ASP cream as an excellent treatment for melasma, with no observed side effects
Tizanidine (TZN) Skeletal muscle relaxant Transdermal -Ascorbyl 6-palmitate In vitro, ex vivo & in vivo -The optimized aspasomal formulations showed high TZN entrapment%, small size and high TZN release 185
-Cholesterol -The prepared ASP were revealed to be non-irritating and safe for topical skin application
-Span 60 -Ex vivo permeation studies demonstrated that the ASP-based formulation resulted in a 4.4-fold boost in the steady-state flux compared to the free drug
  -The pharmacokinetic study revealed that the optimized ASP led to a 3.4-fold enhancement in the drug bioavailability relative to the oral tablets
Idebenone/Naproxen Idebenone: antioxidant Topical -Ascorbyl 6-palmitate In vitro & in vivo -The ASP formulation with the best physicochemical properties was composed of AP, cholesterol, and DMPA or DMPG at a lipid molar ratio of 35[thin space (1/6-em)]:[thin space (1/6-em)]55[thin space (1/6-em)]:[thin space (1/6-em)]10, and it was prepared by subjecting the mixture to 50% amplitude 186
Naproxen: anti-inflammatory (skin inflammatory pathologies) -Cholesterol -The combination of the loaded idebenone and naproxen was found to not significantly impact the physicochemical properties of the ASP vesicles or their thermostability
  -Negatively charged phospholipids: DMPA & DMPG -The release studies revealed that the biphasic kinetic profile of the attained ASP, with a controlled release of both the loaded drugs (idebenone and naproxen) for up to 24 hours
    -The in vivo study showed that the ASP-based formulation was able to decrease chemically induced skin erythema more effectively than a commercially available naproxen gel, with the effects observed after just 1 hour of application
    -The ASP demonstrated a potential synergy between the loaded idebenone and naproxen and AP, self-assembled within the vesicular bilayer, when applied topically
Mometasone furoate Psoriasis Topical -AP In vitro, ex vivo & in vivo -The optimized formula showed increased drug-loading capacity compared to the conventional gel formulation, and enhanced permeability and controlled drug release through the skin barrier 145
-Cholesterol -The in vivo animal model demonstrated superior therapeutic efficacy compared to the conventional gel formulation, with improved anti-inflammatory and anti-psoriatic effects, leading to a significant improvement in the clinical symptoms of psoriasis
-Dicetyl phosphate  
Itraconazole (ITZ) Anti-tumorigenic for skin cancer Topical -AP-cholesterol In vitro, ex vivo & in vivo -The optimized ASP exhibited desirable characteristics, including a nano-size of 67.83 ± 6.16 nm, a negative surface charge of −79.40 ± 2.23 mV, over 95% entrapment of ITZ, and high colloidal stability 187
-Epikuron® TM 200 -The presence of AP in the formulation provided considerable antioxidant power and enhanced the cytotoxicity of ITZ against skin cancer cells “A431 cells; IC50 = 5.3 ± 0.27 μg mL−1
  -The aspasomal cream formulation demonstrated improved skin penetrability and enhanced in vivo anticancer competence, resulting in a substantial reduction of “62.68%” in tumor weight compared to the control


Expanding on previous research, aspasomal dispersions were effectively created using the rotary-evaporation–sonication approach. This method has been noted for its ability to produce vesicles with outstanding drug-incorporation efficiency.179 This technique involves creating a thin film that promotes thorough and effective hydration of the vesicles, allowing for the encapsulation of substantial quantities of drugs, as illustrated in Fig. 5.


image file: d5ra03825a-f5.tif
Fig. 5 Method of aspasomal dispersion preparation “thin film hydration method”. In the thin film hydration method, the main components of aspasomes (ascorbyl palmitate, phospholipid and cholesterol) are dissolved in a mixture of chloroform[thin space (1/6-em)]:[thin space (1/6-em)]ethanol (2[thin space (1/6-em)]:[thin space (1/6-em)]1). Organic solvent evaporation is performed in a rotary evaporator at 60 °C and 120 rpm. The formed dry thin film is hydrated with phosphate buffer saline (PBS; pH 7.4). This figure was created with BioRender.

Specifically, for lipid-based vesicles generated using thin-film hydration technique, formulation and processing parameters have been well documented to greatly affect the physical characteristics of the produced nanosystems. Formulation design and optimization are crucial steps for boosting the therapeutic competence of vesicles.

Of note, referring to literature, ASP-based topical products (creams or gels) enclosing diverse drugs were evaluated clinically as a competent remedy for androgenic alopecia, melasma, fungal infections (candidiasis and tinea), acne and skin aging.180,188–192

6.2. Ascorbyl 2-glucoside “AA-2G”

Ascorbyl-2-glucoside (AA-2G) has emerged as one of the most investigated vitamin C derivatives and has garnered significant concern due to its excellent thermal stability and oxidation resistance.193,194 AA-2G is a hydrophilic derivative of AA. It appears as a white or yellowish-white powder or crystalline powder. AA-2G is colorless and has a molecular weight of 338.265 Da, with a log[thin space (1/6-em)]P value of less than −2, indicating its hydrophilic nature.195 AA-2G is produced by a reaction between glucose and AA. As illustrated in Fig. 1F, the hydroxyl group at the second position of the AA molecule interacts with glucose to form AA-2G. This connection enhances the molecule's stability, safeguarding it from possible degradation due to exposure to high temperatures and metal ions and varying pH levels. Furthermore, this specific binding site contributes to the degradation process and directly affects the antioxidant activity of AA-2G.196

When applied topically, ascorbyl-2-glucoside (AA-2G) is reported to undergo hydrolysis in the presence of a cellular α-glucosidase enzyme, which then converts it to L-AA within the skin.197–200 In addition to its topical uses, AA-2G is utilized as a food additive and as a functional excipient in cosmetic preparations. AA-2G is utilized as a whitening agent due to its ability to reduce melanin production, and in the formulation of lipid-soluble vitamins.114,195,199,201

AA-2G can directly provide significant, concentration-dependent preservation against oxidative stress-prompted cell death in human dermal fibroblasts, without requiring conversion to AA. Experiments confirmed that the prophylactic impacts of AA-2G are inherent to the compound, rather than resulting from its hydrolysis to release AA. Hanada et al. suggest that AA-2G may have practical utility as a beneficial antioxidant agent even before it is converted into the active AA form.202

Lately, in the drug-delivery arena, AA-2G has been identified as a solubilizer for poorly soluble drugs. In light of this, Inoue et al. reported that co-grinding of AA-2G with the poorly water-soluble drug, clarithromycin, yielded considerable drug solubilization, and strikingly, nanoparticle formation was observed when using the drug in a molar ratio of 2[thin space (1/6-em)]:[thin space (1/6-em)]1.203 This substance has been documented not only as a solubilizer but also as a promising auxiliary material in nanoparticle design.204 On the other hand, due to its antioxidant power, it has been utilized as a drug, as reported by Lin et al. (2016). In their investigation, AA-2G was enclosed in a microemulsion. The findings indicated that the AA-2G microemulsion possessed higher penetrability in comparison to the commercially available emulsions.205 Recently, AA-2G has been investigated as a prodrug derivative of AA in commercial products, demonstrating a stronger antioxidative and hence a promising protective leverage. Notably, using AA-2G at a much lower concentration, reaching 1.8%, produced comparable results to those achieved with a higher AA concentration (15%).206

The incorporation of AA-2G into innovative nanoparticles has emerged as an attractive strategy to potentiate the therapeutic competence of this compound. In a recent study, researchers developed solid-in-oil nanodispersions encapsulating either AA-2G or AA to improve their stability and optimize their delivery for corneal wound healing applications. The nanodispersion formulations, with an average size of 200–300 nm, exhibited high encapsulation efficiencies, good colloidal stability, and appropriate rheological properties for topical ocular administration. In vitro evaluation using human corneal epithelial cells demonstrated that both the AA-2G and AA nanodispersions significantly enhanced cellular proliferation, migration, and wound closure, compared to the free compounds. Importantly, the AA-2G nanodispersion showed superior performance over the AA nanodispersion in promoting corneal wound healing, which was attributed to the better stability and bioavailability of the AA-2G form in the ocular environment. Promising in vivo results in a rabbit corneal wound model were obtained, where the nanodispersion treatments accelerated wound closure and improved histological markers of wound healing.193

Recent research has developed innovative vesicles called glucospanlastics, which incorporate ascorbyl-2-glucoside (AA-2G) as an integral component to enhance their antioxidant and therapeutic properties in the delivery of itraconazole.207 Table 3 demonstrates earlier works highlighting different applications of AA-2G in the drug delivery arena.

Table 3 Different applications of AA-2G in the pharmaceutical and medical fields
Role/function of AA-2G Delivery system/dosage form Status of investigation Application of AA-2G Reference
Active constituent In vitro (DPPH assay and skin permeation) Skin antioxidant effect and capability of skin permeation 199
In vitro (evaluation of neurite-forming cells using neural precursor cell line “PC12 cells”) Regulation of the redox state in the neural cells. Neural differentiation and growth of neural network-forming cells 208
In vivo (cisplatin-induced nephrotoxicity in mice) Protection against oxidative renal damage induced by cisplatin 209
Gel In vitro (B16 melanoma cells and 3D human skin) and in vivo (clinical assessment) Inhibition of melanin formation and treatment of gingival melanin pigmentation 210
In vivo (exposure of mice to gamma radiation) Radio protection (antioxidant potential, powerful haemopoietic stimulation and gastrointestinal protection) 211
In vitro (sperm motility and lipid peroxidation) and in vivo (clinical) Human sperm motility preservation after cryopreservation and sperm recovery 212
In vivo (isolation of hepatocytes from rats) Creation of DNA synthesis and proliferation of rat hepatocytes 213
Cosmetic lotion as a drug In vitro (scavenging of free radicals) Providing stability and antioxidant power 200
In vitro (oxidative cell death protection assay using human fibroblast cells) Protection of the cells from free radical-induced cytotoxicity 214
In vivo (determination of liver regeneration and function in 70% hepatectomized rats) Powerful stimulation of normal hepatocyte proliferation and reestablishment of liver function 215
In vitro (MTT assay on colon-26 cells) and in vivo (assessment of antitumor activity in colon-26 xenograft mice) Cancer treatment (inhibitor of tumor growth for infusion therapy) 216
In vitro (investigation of reactive oxygen species and potential of mitochondrial membrane in gastric adenocarcinoma cells) Protection against H. pylori infection in epithelial cells of the stomach via reduction of H. pylori-induced apoptosis 217
In vitro (exposure of Chinese hamster ovary cells to gamma rays and ultraviolet) Protection of cells from radiation and UV light via filtration and blocking of UVC and reduced short UVB wavelengths 218
In vitro (determination of proliferation, migration, and angiogenesis in vascular endothelial cells C166 and embryonic fibroblasts NIH-3T3) and in vivo (wound healing model in mice) Promotion of wound healing of locally transplanted mesenchymal stem cells via boosting of proliferation and angiogenesis and collagen deposition of cells, leading to a high degree of vascularization in vivo 219
In vivo (rat bladder tumor cell model) Reduction of the intestinal damage and preservation of antitumor effectiveness of radiotherapy against bladder tumors 220
Raw clay-based formulation emulsion In vitro (release, skin permeation using human cadaver skin, skin retention study and cell viability study in HaCaT and HDF cell lines) Appropriate delivery of AA-2G with no toxicity detected in cell lines 221
Excipient in drug delivery systems NPs In vitro (solubilization study of co-ground mixtures with drug) Solubilization of clarithromycin, nanoparticle formation for drug AA-2G in the molar ratio of 2[thin space (1/6-em)]:[thin space (1/6-em)]1 (mean PS = 280 nm) 222
NPs (cyclodextrin complex systems containing hydrophilic auxiliary substances “TPGS and AA-2G”) In vitro (phase solubility and saturation solubility studies and dissolution) and in vivo (anti-hypercholesterolemic efficacy in rats) Enhancement of the solubility, wetting, and dispersibility of BCS class II drug “ezetimibe” 223
Solid dispersion (containing AA-2G) In vitro (dissolution study and solid-state characterization) Improvement of dissolution of triamterene via complex formation 224
Water in oil emulsion (containing zinc oxide and AA-2G) In vitro (challenge test for investigation of antimicrobial potential) Improvement of the antimicrobial activity of ZnO via complexing properties of AA-2G towards zinc and enhancing the solubility of the antimicrobial soluble zinc species 225
Integral part of drug-delivery systems Gluco-spanlastics In vitro (physicochemical characterization, morphological examination, differential scanning chromatography, Fourier transform infra-red spectroscopy, antioxidant activity, cytotoxicity assay) AA-2G is successfully integrated as a vesicle component in the formation of an innovative, powerful antioxidant vesicle called glucospanlastics, achieving a 95% loading of Itraconazole 207
Ex vivo (confocal scanning microscopy)
In vivo (Ehrlich ascites carcinoma model, histopathological study, biochemical assessment)


7. Outcomes and future directions

Currently, a renewed solicitude is growing to promote the structural modification of AA to produce functionally derived AA-preserving structures that meet pharmaceutical and medical needs. Indeed, the chemical production of AA derivatives using advanced different processing techniques can assure their high availability and give the opportunity to fabricate a series of economical and cost-effective derivatives. Interestingly, newly developed sustainable and eco-friendly AA derivatives with substituted 1,2,3-triazole moieties were fabricated utilizing both microreactor technology and ultrasonic irradiation (continuous flow process).226 Recently, a novel eco-friendly derivative, ascorbyl-6-O-oleate, was synthesized with a heightened yield via a lipase-catalysed esterification, as well as transesterification based on the reaction between the AP substrate and oleic acid, a successful step towards its commercialization.227 In such a way, production costs can be lowered, allowing them to replace traditional excipients and therapeutic agents while widening their applicability.

Concerning pharmaceutical needs, diverse AA derivatives have been shown to be effective and promising additives in various pharmaceutical formulations, acting as solubilizers, stabilizes and emulsifiers.228–232 From drug-delivery perspectives, if synthesized and validated, the structural derivatives of AA show promise for application as carriers, transforming the drug-delivery arena and yielding transformative implications for futuristic pharmaceutical research. In particular, the amphiphilic derivatives can be exploited as tools in the fabrication of liposomes and their newer-generation variants, as well as micelles via self-assembly.227 For example, octanoyl-6-O-ascorbic acid, decanoyl-6-O-ascorbic acid and trisodium L-ascorbyl 2-phosphate 6-palmitate were employed in the production of micellar dispersions that were found to be effective as solubilization vehicles for various hydrophobic drugs.230,233,234 Additionally, vesicular systems based on the integration of amphiphilic alkyl ester AA derivatives, such as AP, ascorbyl myristate and ascorbyl laurate, into the phospholipid bilayer were investigated in a previous study.235

In consideration of their bioactivity, the fabrication and evolution of such functional molecular entities could expand the pool of therapeutic alternatives with diversified bioactive attributes available to patients suffering from inflammation- and oxidative-stress-related chronic diseases, as well as cancer.236,237

Considerably, shifting specific DDS is expected to potentiate and amplify therapeutic interventions that elicit antioxidant, anti-inflammatory and antiproliferative activities. Optimally, the versatility and potential of integrated delivery systems to enhance the therapeutic performance of combination therapies is substantial, spanning applications specifically in cancer treatment and oxidative-damage-related disorders, by enabling the synergistic action of such derivatives and other bioactive compounds.

Indeed, in earlier works, some AA derivative-based delivery systems have been attested to be efficacious medications. In this context, exploiting AP in the structure of the promising vesicles, i.e., ASP, to enclose the repurposed anticancer drug, itraconazole, yielded tunable nano-platforms with potentiated anticancer activity, verified both in vitro and in vivo.187 Strikingly, the aforementioned unloaded vesicles, when embedded in a cream base and tested clinically for the management of candidiasis associated with diaper dermatitis, exhibited reasonable curative consequences, owing to their role as carriers to boost skin deposition alongside their antioxidant potential and capability to neutralize reactive oxygen species at affected inflammatory sites.188 It is worth mentioning that tolerability and safety following topical application were verified, with no reported adverse effects.

Moreover, the application scope of some AA derivatives has been explored in the field of cosmetics, being linked to the preservation of the antioxidant and melanogenesis-inhibiting features of AA. As a consequence, they can offer distinctive effects towards skin whitening, pigmentation-disorder remediation and antiaging.189,238

Indeed, some AA ethers and esters, including tetrahexyldecyl ascorbate, AP, ascorbyl linoleate and ascorbyl stearate, have demonstrated striking clinical outcomes in the field of cosmetics, functioning as antioxidants, skin protectants, fragrance excipients, and skin-conditioning agents.239 Such derivatives receive regulatory approval and show widespread tolerability when applied topically.

Interestingly, several derivatives have already been marketed in cosmetic products, including serums, lotions and creams.

When exploring AA derivatives in the market, the effectiveness of several standout products has been demonstrated. For instance, Mad Hippie Vitamin C Serum240 and TruSkin Vitamin C Facial Serum241 utilize sodium ascorbyl phosphate, making them excellent choices for targeting brightening and acne control. Sunday Riley C.E.O. 15% Vitamin C Brightening Serum242 and Peter Thomas Roth Potent-C Power Serum243 feature THD ascorbate, known for its superior absorption and brightening properties. Meanwhile, products like Ole Henriksen Banana Bright 15% Vitamin C Serum244 and Allies of Skin 20% Vitamin C Brighten + Firm Serum245 incorporate 3-O ethyl ascorbic acid, which effectively addresses hyperpigmentation. Additionally, The Ordinary 12% AA-2G Solution246 and Inkey List 15% Vitamin C and EGF Serum247 leverage AA-2G for its brightening and collagen-supporting benefits. These products are suitable for diverse skin types and concerns, showcasing the versatility of AA derivatives in skincare.

Looking ahead, contemporary eco-sustainable synthetic methods for AA derivation, optimally for mass production, and great discoveries and advancements in delivery systems based on such valuable derivatives could be explored to achieve breakthroughs in various pharmaceutical and medical fields. As a consequence, tunable and scalable medicines based on AA derivatives with modulated drug release and amplified therapeutic response should be anticipated in the pharmaceutical market.

8. Limitations

Despite its striking role in the drug-delivery arena, the claim linking AA derivatives to therapeutics and functional additives and carriers has distinct limitations and challenges that must be fully addressed.

For large-scale implementation and scalability, eco-sustainable production techniques, as well as the extension of the manufacturing to the industrial scale, are still demanded. In this scenario, synthetic techniques should ensure the production of customized derivatives, establishing appropriate yields and stability. Optimizing the costs of the exploited resources and involved equipment can assist the production of cost-effective and available AA derivatives for commercial applications.

Although they are valuable therapeutic agents, their pharmacological concentration doses and tolerable levels when used as therapeutics, as well as the associated possible risks of over-supplementation, limit their application. The long-term safety profile of the majority of AA derivatives remains unclear. Accordingly, precise monitoring of dosing and the course of treatment is imperative. The setting and approval of the intake dose is considered necessary. Pharmacological evaluation and a mechanistic rationale for identifying their therapeutic utilization are necessary as well. Although in vitro findings and animal studies have asserted the bioactivity of individual AA derivatives and their associated delivery systems, there is presently no conclusive proof establishing their impact on human tissues. It is crucial to perform clinical investigations to validate animal models and to probe their translation to human subjects.

Another key area that needs further research is the verification of their conversion into AA following their administration, as the conversion can be affected by differences in the physiology of the intended routes. Following oral ingestion, AA derivatives have been stated to be readily available AA sources.248 This is not the case for topical application. In light of this, the fate of the applied AA derivatives, in terms of their skin absorption or conversion into AA after dermal penetration, should be understood. Measuring the concentrations of both AA and its derivative in the skin via analysis of the skin after their application and comparing the AA concentration to its baseline skin level could help in this respect. Importantly, the suitability of skin-oriented delivery systems composed of AA derivatives should be determined via profiling ex vivo skin deposition and permeation using Franz diffusion cells.249

Regarding their role as drug carriers and functional excipients, research into their usage, concentration levels, physicochemical properties, solid-state characterizations, and imaging, should be emphasized. More research is also needed concerning their adverse reactions, interaction with other ingredients, toxicity and tolerance. In general, empirical validation of delivery systems covering in vitro, ex vivo and in vivo performances should be conducted. Delivery-system-related issues must be scientifically interpreted pending powerful empirical advocacy. For example, the proposed link between nanoparticle attributes (i.e., nanosize, surface charge, drug encapsulation) and in vivo cellular uptake in different cell cultures, cytocompatibility, distribution, pharmacokinetics and pharmacodynamics warrants further considerable exploration. Differences in the fate and behavior of different nanoplatforms inside the body are still questionable and need to be investigated.

It is worth mentioning that the suggested methodologies for administration involving new delivery systems via nasal, otic and pulmonary routes, as well as pharmacodynamics tests for hypertension and CNS-related diseases (such as stroke), present practical constraints due to dependence on specialized equipment and operators, invasiveness, and sometimes complexity and limited accessibility. Of note, little in vitro-in vivo correlation data were found for AA derivative-based delivery systems. From translational perspectives, their toxicological reports on animal tissues, as well as human ones, should be explored. Intense clinical experimentation should be assessed and precisely optimized in order to figure out their empirical significance and broad acceptance in the drug-delivery field.

9. Conclusions and perspectives

AA is a highly valued multifunctional antioxidant biomolecule, but its instability and delivery-related issues have resulted in the evolution of various derivatives. These derivatives have shown effectiveness in mitigating oxidative stress, potentiating antiproliferative efficacy, promoting collagen synthesis and reducing hyperpigmentation in the skin. Of these, AP is a hydrophobic derivative that exhibits good skin penetration and antioxidant activity, and a hydrophilic derivative AA-2G has been studied for its power to inhibit melanin production and improve skin brightening. However, to further enhance the delivery and performance of these AA derivatives, researchers have explored their incorporation into delivery systems either as an integral part of the system or as a drug. Modern evolutions in pharmaceutical delivery involving nanoplatforms, emulsions and gels can benefit the pharmaceutical community in this respect. The design of such innovative therapeutics can optimally offer better and potentiated curative efficacy with improved life quality.

However, adequate design and rigorous monitoring are required for modulation of their physicochemical, bioactive and toxicological features. The recognition of different biological barriers, transport pathways and complexity of various encountered permeation and targeting challenges will assist in the formulation of tailored platforms with respect to their localization in the affected site of action, distribution, targeting and prolonged intended action. In this context, a focus on the technological features is needed, encompassing a proper choice of other components as well as the formulation of delivery systems and industrial techniques for the design and subsequent technology transfer. Importantly, continued research in this area is warranted to further elucidate the structure–activity relationships and degradability while developing even more efficacious and stable AA derivative-based delivery systems for pharmaceutical and medical fields. In light of this, their stability for prolonged periods, assuring the maintenance of their physicochemical, bioactive and AA-preserving benefits over time, degradability and in-depth awareness of their link with the composition of the delivery vehicle should be taken into consideration. Besides, experimental studies using in vivo animal models and imaging tools to probe the in vivo behavior and fate are considered mandatory. Increased research into the profiles of the molecular expression of different genes of signaling pathways can provide an opportunity to demonstrate their linked mechanistic leverage and, hence, their fit-for-purpose biological performance. Moreover, the utilization of computer simulations and pharmacokinetic profiling studies and their advancements can assist in this respect. Yet, the assessments of their related toxicity and cogent in vivo animal and clinical studies are still limited and require further studies.

Overall, AA derivatives, particularly when incorporated in nanoparticle delivery systems and scrutinized against all the aforementioned aspects, could represent a promising approach to harness the power of AA for improved biological impacts in various therapeutic modalities. The attested achievement in the realm of cosmetics could set the substantial impetus to design various delivery platforms, based on such functional additives, with prevalent clinical utility in the management of systemic disorders. As a consequence, such functional entities in marketed delivery platforms would soon provide an efficient cure for a wide range of oxidative stress and inflammation-related ailments.

Author contributions

Caroline Lamie: formal analysis, and writing – original draft. Enas Elmowafy, formal analysis, supervision, and review & editing. Dalia A. Attia, formal analysis, supervision and review & editing. Nahed D. Mortada, formal analysis, supervision and review & editing.

Conflicts of interest

The authors have no conflicts of interest to disclose.

Data availability

In this review article, no primary research findings, software, or code was included and no new data were generated or analyzed.

Acknowledgements

This study did not obtain any specific funding from grant agencies.

References

  1. İ. Gulcin, Antioxidants and antioxidant methods: An updated overview, Arch. Toxicol., 2020, 94(3), 651–715 CrossRef PubMed.
  2. P. D. Prenzler, D. Ryan and K. Robards, Introduction to basic principles of antioxidant activity, 2021 Search PubMed.
  3. E. A. Decker, Antioxidant mechanisms, Food Sci. Technol., 1998, 24, 397–422 Search PubMed.
  4. A. M. Rizzo, P. Berselli, S. Zava, G. Montorfano, M. Negroni, P. Corsetto and B. Berra, Endogenous antioxidants and radical scavengers, in Bio-farms for Nutraceuticals: Functional Food and Safety Control by Biosensors, Springer, 2010, pp. 52–67 Search PubMed.
  5. N. Noguchi and E. Niki, Chemistry of active oxygen species and antioxidants, in Antioxidant Status, Diet, Nutrition, and Health, CRC Press, 2019, pp. 1–20 Search PubMed.
  6. J. Vaya and M. Aviram, Nutritional antioxidants mechanisms of action, analyses of activities and medical applications, Curr. Med. Chem. Immunol. Endocr. Metab. Agents, 2001, 1(1), 99–117 CrossRef CAS.
  7. O. I. Aruoma, Free radicals, oxidative stress, and antioxidants in human health and disease, J. Am. Oil Chem. Soc., 1998, 75(2), 199–212 CrossRef CAS PubMed.
  8. A. A. Adwas, A. Elsayed, A. E. Azab and F. A. Quwaydir, Oxidative stress and antioxidant mechanisms in human body, J. Appl. Biotechnol. Bioeng., 2019, 6(1), 43–47 Search PubMed.
  9. A. Phaniendra, D. B. Jestadi and L. Periyasamy, Free radicals: properties, sources, targets, and their implication in various diseases, Indian J. Clin. Biochem., 2015, 30, 11–26 CrossRef CAS PubMed.
  10. O. Uchechi, J. D. N. Ogbonna and A. A. Attama, Nanoparticles for dermal and transdermal drug delivery, Application of Nanotechnology in Drug Delivery, 2014, 4, pp. 193–227 Search PubMed.
  11. B. Hu, Y. Ouyang, T. Zhao, Z. Wang, Q. Yan, Q. Qian, W. Wang and S. Wang, Antioxidant Hydrogels: Antioxidant Mechanisms, Design Strategies, and Applications in the Treatment of Oxidative Stress-Related Diseases, Adv. Healthcare Mater., 2024, 13(11), 2303817 CrossRef CAS PubMed.
  12. N. F. Santos-Sánchez, R. Salas-Coronado, C. Villanueva-Cañongo and B. Hernández-Carlos, Antioxidant compounds and their antioxidant mechanism, Antioxidants, 2019, 10, 1–29 Search PubMed.
  13. M. Mitra, S. Mitra and D. K. Nandi, Vitamins, micronutrients, antioxidants, and nutraceuticals in neuroprotection: An overview, A Review on Diverse Neurological Disorders, 2024, pp. 585–601 Search PubMed.
  14. H. K. Krishnamurthy, M. Pereira, V. Jayaraman, K. Krishna, T. Wang, K. Bei and J. J. Rajasekaran, Oxidative Stress: Mechanisms, Quantification and its Role in Human Aging, ScienceOpen, 2024 Search PubMed.
  15. M. Sharifi-Rad, N. V. Anil Kumar, P. Zucca, E. M. Varoni, L. Dini, E. Panzarini, J. Rajkovic, P. V. Tsouh Fokou, E. Azzini and I. Peluso, Lifestyle, oxidative stress, and antioxidants: back and forth in the pathophysiology of chronic diseases, Front. Physiol., 2020, 11, 694 CrossRef PubMed.
  16. M. I. Anik, N. Mahmud, A. A. Masud, M. I. Khan, M. N. Islam, S. Uddin and M. K. Hossain, Role of reactive oxygen species in aging and age-related diseases: A review, ACS Appl. Bio Mater., 2022, 5(9), 4028–4054 CrossRef CAS PubMed.
  17. L. Hecker, Mechanisms and consequences of oxidative stress in lung disease: therapeutic implications for an aging populace, Am. J. Physiol. Lung Cell. Mol. Physiol., 2018, 314(4), L642–L653 CrossRef CAS.
  18. F. Hecht, M. Zocchi, F. Alimohammadi and I. S. Harris, Regulation of antioxidants in cancer, Mol. Cell, 2024, 84(1), 23–33 CrossRef CAS PubMed.
  19. M. J. Iqbal, A. Kabeer, Z. Abbas, H. A. Siddiqui, D. Calina, J. Sharifi-Rad and W. C. Cho, Interplay of oxidative stress, cellular communication and signaling pathways in cancer, Cell Commun. Signal., 2024, 22(1), 7 CrossRef CAS PubMed.
  20. M. W. Davey, M. V. Montagu, D. Inze, M. Sanmartin, A. Kanellis, N. Smirnoff, I. J. J. Benzie, J. J. Strain, D. Favell and J. Fletcher, Plant L-ascorbic acid: chemistry, function, metabolism, bioavailability and effects of processing, J. Sci. Food Agric., 2000, 80(7), 825–860 CrossRef CAS.
  21. C. Liu, J. Li, C. Wang and Q. Ren, Fabrication of crosslinked starch microspheres via water-in-water Pickering emulsion and their application in controlled release of Vitamin C, J. Dispersion Sci. Technol., 2024, 1–13 Search PubMed.
  22. Y. C. Boo, Ascorbic acid (vitamin C) as a cosmeceutical to increase dermal collagen for skin antiaging purposes: emerging combination therapies, Antioxidants, 2022, 11(9), 1663 CrossRef CAS PubMed.
  23. T. Comunian, A. Babazadeh, A. Rehman, R. Shaddel, S. Akbari-Alavijeh, S. Boostani and S. M. Jafari, Protection and controlled release of vitamin C by different micro/nanocarriers, Crit. Rev. Food Sci. Nutr., 2022, 62(12), 3301–3322 CrossRef CAS PubMed.
  24. X. Yin, K. Chen, H. Cheng, X. Chen, S. Feng, Y. Song and L. Liang, Chemical stability of ascorbic acid integrated into commercial products: A review on bioactivity and delivery technology, Antioxidants, 2022, 11(1), 153 CrossRef CAS PubMed.
  25. A. Murugadoss, R. Pasricha and A. Chattopadhyay, Ascorbic acid as a mediator and template for assembling metallic nanoparticles, J. Colloid Interface Sci., 2007, 311(1), 303–310 CrossRef CAS PubMed.
  26. G. Suriati, M. Mariatti and A. Azizan, Synthesis of silver nanoparticles by chemical reduction method: Effect of reducing agent and surfactant concentration, Int. J. Automot. Mech. Eng., 2014, 10, 1920–1927 CrossRef CAS.
  27. C. Gutiérrez-Wing, J. J. Velázquez-Salazar and M. José-Yacamán, Procedures for the synthesis and capping of metal nanoparticles, Nanoparticles in Biology and Medicine: Methods and Protocols, 2020, pp. 3–20 Search PubMed.
  28. M. A. Sheraz, M. F. Khan, S. Ahmed, S. H. Kazi and I. Ahmad, Stability and stabilization of ascorbic acid, Household Pers. Care Today, 2015, 10, 22–25 Search PubMed.
  29. M. A. Sheraz, M. F. Khan, S. Ahmed, S. H. Kazi, S. R. Khattak and I. Ahmad, Factors affecting formulation characteristics and stability of ascorbic acid in water-in-oil creams, Int. J. Cosmet. Sci., 2014, 36(5), 494–504 CrossRef CAS PubMed.
  30. N. Khalid, I. Kobayashi, M. A. Neves, K. Uemura, M. Nakajima and H. Nabetani, Monodisperse W/O/W emulsions encapsulating l-ascorbic acid: Insights on their formulation using microchannel emulsification and stability studies, Colloids Surf., A, 2014, 458, 69–77 CrossRef CAS.
  31. S. Jacob, A. Shirwaikar, I. Elsayed, T. J. Thomas and S. Anoop, Formulation and Evaluation of Stable Ascorbic Acid Multiple Emulsion, President's Message, 2015 Search PubMed.
  32. J. D. Hoyos-Leyva, A. Chavez-Salazar, F. Castellanos-Galeano, L. A. Bello-Perez and J. Alvarez-Ramirez, Physical and chemical stability of L-ascorbic acid microencapsulated into taro starch spherical aggregates by spray drying, Food Hydrocolloids, 2018, 83, 143–152 CrossRef CAS.
  33. B. Bordignon, J. Chiron and M. Fontés, Ascorbic acid derivatives as a new class of antiproliferative molecules, Cancer Lett., 2013, 338(2), 317–327 CrossRef CAS PubMed.
  34. P. Jakubek, K. Suliborska, M. Kuczyńska, M. Asaduzzaman, K. Parchem, I. Koss-Mikołajczyk, B. Kusznierewicz, W. Chrzanowski, J. Namieśnik and A. Bartoszek, The comparison of antioxidant properties and nutrigenomic redox-related activities of vitamin C, C-vitamers, and other common ascorbic acid derivatives, Free Radic. Biol. Med., 2023, 209, 239–251 CrossRef CAS PubMed.
  35. A. Stolić Jovanović, M. Martinović, A. Žugić, I. Nešić, T. Tosti, S. Blagojević and V. M. Tadić, Derivatives of L-Ascorbic Acid in Emulgel: Development and Comprehensive Evaluation of the Topical Delivery System, Pharmaceutics, 2023, 15(3), 813 CrossRef PubMed.
  36. S. Chambial, S. Dwivedi, K. K. Shukla, P. J. John and P. Sharma, Vitamin C in disease prevention and cure: an overview, Indian J. Clin. Biochem., 2013, 28, 314–328 CrossRef CAS PubMed.
  37. M. Percival, Nutritional support for connective tissue repair and wound healing, Clin. Nutr. Insight, 1997, 6(98), 1–4 Search PubMed.
  38. P. Aghajanian, S. Hall, M. D. Wongworawat and S. Mohan, The roles and mechanisms of actions of vitamin C in bone: new developments, J. Bone Miner. Res., 2015, 30(11), 1945–1955 CrossRef CAS PubMed.
  39. S. J. Ballaz and G. V. Rebec, Neurobiology of vitamin C: Expanding the focus from antioxidant to endogenous neuromodulator, Pharmacol. Res., 2019, 146, 104321 CrossRef CAS PubMed.
  40. R. Figueroa-Méndez and S. Rivas-Arancibia, Vitamin C in health and disease: its role in the metabolism of cells and redox state in the brain, Front. Physiol., 2015, 6, 397 Search PubMed.
  41. J. Kocot, D. Luchowska-Kocot, M. Kiełczykowska, I. Musik and J. Kurzepa, Does vitamin C influence neurodegenerative diseases and psychiatric disorders?, Nutrients, 2017, 9(7), 659 CrossRef PubMed.
  42. V. Sasidharan Nair and J. Huehn, Impact of vitamin C on the development, differentiation and functional properties of T cells, Eur. J. Microbiol. Immunol., 2024, 14(2), 67–74 Search PubMed.
  43. L. Mititelu-Tartau, M. Bogdan and M. Ciocoiu, Vitamin C from bench to bedside, Front. Nutr., 2024, 11, 1406342 CrossRef PubMed.
  44. A. Sorice, E. Guerriero, F. Capone, G. Colonna, G. Castello and S. Costantini, Ascorbic acid: its role in immune system and chronic inflammation diseases, Mini Rev. Med. Chem., 2014, 14(5), 444–452 CrossRef CAS PubMed.
  45. A. S. Jamshidovich, Ascorbic acid: its role in immune system, chronic inflammation diseases and on the antioxidant effects, Eur. J. Mod. Med. Practice, 2023, 3(11), 57–60 Search PubMed.
  46. A. Gęgotek and E. Skrzydlewska, Antioxidative and anti-inflammatory activity of ascorbic acid, Antioxidants, 2022, 11(10), 1993 CrossRef PubMed.
  47. R. J. Jariwalla and S. Harakeh, Antiviral and immunomodulatory activities of ascorbic acid, Subcellular Biochemistry: Ascorbic Acid: Biochemistry and Biomedical Cell Biology, 1996, pp. 215–231 Search PubMed.
  48. I. R. L. Andriolo, L. Venzon and L. M. da Silva, Perspectives About Ascorbic Acid to Treat Inflammatory Bowel Diseases, Drug Res., 2024 Search PubMed.
  49. A. E. Ratajczak, A. Szymczak-Tomczak, M. Skrzypczak-Zielińska, A. M. Rychter, A. Zawada, A. Dobrowolska and I. Krela-Kaźmierczak, Vitamin C deficiency and the risk of osteoporosis in patients with an inflammatory bowel disease, Nutrients, 2020, 12(8), 2263 CrossRef CAS PubMed.
  50. M. Ponec, A. Weerheim, J. Kempenaar, A. Mulder, G. S. Gooris, J. Bouwstra and A. M. Mommaas, The formation of competent barrier lipids in reconstructed human epidermis requires the presence of vitamin C, J. Invest. Dermatol., 1997, 109(3), 348–355 CrossRef CAS PubMed.
  51. S. Ohno, Y. Ohno, N. Suzuki, G.-I. Soma and M. Inoue, High-dose vitamin C (ascorbic acid) therapy in the treatment of patients with advanced cancer, Anticancer Res., 2009, 29(3), 809–815 CAS.
  52. K.-A. Lee, S.-H. Lee, Y.-J. Lee, S. M. Baeg and J.-H. Shim, Hesperidin induces apoptosis by inhibiting Sp1 and its regulatory protein in MSTO-211H cells, Biomol Ther., 2012, 20(3), 273 CrossRef CAS PubMed.
  53. N. Sivaranjani, S. V. Rao and G. Rajeev, Role of reactive oxygen species and antioxidants in atopic dermatitis, J. Clin. Diagn. Res., 2013, 7(12), 2683 CAS.
  54. S. H. Byun and Y. Jeon, Administration of vitamin C in a patient with herpes zoster-a case report, Korean J. Pain, 2011, 24(2), 108–111 CrossRef CAS PubMed.
  55. J. Y. Kwak, S. Park, J. K. Seok, K.-H. Liu and Y. C. Boo, Ascorbyl coumarates as multifunctional cosmeceutical agents that inhibit melanogenesis and enhance collagen synthesis, Arch. Dermatol. Res., 2015, 307(7), 635–643 CrossRef CAS PubMed.
  56. S. Vasanth, A. Dubey, R. Gs, S. A. Lewis, V. M. Ghate, S. A. El-Zahaby and S. Hebbar, Development and investigation of vitamin C-enriched adapalene-loaded transfersome gel: a collegial approach for the treatment of acne vulgaris, AAPS PharmSciTech, 2020, 21(2), 1–17 CrossRef.
  57. D. A. Basketter, I. R. White, P. Kullavanijaya, P. Tresukosol, M. Wichaidit and J. P. McFadden, Influence of vitamin C on the elicitation of allergic contact dermatitis to p-phenylenediamine, Contact Dermat., 2016, 74(6), 368–372 CrossRef CAS.
  58. S. R. Al-Katib, H. A. Al-Wakeel and R. F. Al-Rawaf, Role of Vitamin C as Antioxidant in Psoriasis Patients Treated with NB-UVB Phototherapy, Indian J. Public Health Res. Dev., 2018, 9(10), 375–380 CrossRef.
  59. E. Fernández-García, Skin protection against UV light by dietary antioxidants, Food Funct., 2014, 5(9), 1994–2003 RSC.
  60. S. Dunaway, R. Odin, L. Zhou, L. Ji, Y. Zhang and A. L. Kadekaro, Natural antioxidants: multiple mechanisms to protect skin from solar radiation, Front. Pharmacol., 2018, 9, 392 CrossRef PubMed.
  61. M. Ehrlich, J. Rao, A. Pabby and M. P. Goldman, Improvement in the appearance of wrinkles with topical transforming growth factor β1 and L-ascorbic acid, Dermatol. Surg., 2006, 32(5), 618–625 CAS.
  62. F. Al-Niaimi and N. Y. Z. Chiang, Topical vitamin C and the skin: mechanisms of action and clinical applications, J. Clin. Aesthet. Dermatol., 2017, 10(7), 14 Search PubMed.
  63. A. N. Zaid and R. Al Ramahi, Depigmentation and anti-aging treatment by natural molecules, Curr. Pharm. Des., 2019, 25(20), 2292–2312 CrossRef CAS PubMed.
  64. W. Zhao, A. Yang, J. Wang, D. Huang, Y. Deng, X. Zhang, Q. Qu, W. Ma, R. Xiong and M. Zhu, Potential application of natural bioactive compounds as skin-whitening agents: A review, J. Cosmet. Dermatol., 2022, 21(12), 6669–6687 CrossRef PubMed.
  65. N. Smit, J. Vicanova and S. Pavel, The hunt for natural skin whitening agents, Int. J. Mol. Sci., 2009, 10(12), 5326–5349 CrossRef CAS PubMed.
  66. K. Iqbal, A. Khan and M. Khattak, Biological significance of ascorbic acid (vitamin C) in human health-a review, Pakistan J. Nutr., 2004, 3(1), 5–13 Search PubMed.
  67. S. V. Torti and F. M. Torti, Iron and cancer: more ore to be mined, Nat. Rev. Cancer, 2013, 13(5), 342–355 CrossRef CAS PubMed.
  68. B. Ngo, J. M. Van Riper, L. C. Cantley and J. Yun, Targeting cancer vulnerabilities with high-dose vitamin C, Nat. Rev. Cancer, 2019, 19(5), 271–282 CrossRef CAS PubMed.
  69. M. Uetaki, S. Tabata, F. Nakasuka, T. Soga and M. Tomita, Metabolomic alterations in human cancer cells by vitamin C-induced oxidative stress, Sci. Rep., 2015, 5(1), 13896 CrossRef PubMed.
  70. X. Su, Z. Shen, Q. Yang, F. Sui, J. Pu, J. Ma, S. Ma, D. Yao, M. Ji and P. Hou, Vitamin C kills thyroid cancer cells through ROS-dependent inhibition of MAPK/ERK and PI3K/AKT pathways via distinct mechanisms, Theranostics, 2019, 9(15), 4461 CrossRef CAS.
  71. W. H. Talib, D. A. Ahmed Jum’Ah, Z. S. Attallah, M. S. Jallad, L. T. Al Kury, R. W. Hadi and A. I. Mahmod, Role of vitamins A, C, D, E in cancer prevention and therapy: Therapeutic potentials and mechanisms of action, Front. Nutr., 2024, 10, 1281879 CrossRef PubMed.
  72. H. Dong, G. Zhu, K. Tamada and L. Chen, B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion, Nat. Med., 1999, 5(12), 1365–1369 CrossRef CAS.
  73. S. P. Patel and R. Kurzrock, PD-L1 expression as a predictive biomarker in cancer immunotherapy, Mol. Cancer Therapeut., 2015, 14(4), 847–856 CrossRef CAS PubMed.
  74. N. Mikirova, J. Casciari, A. Rogers and P. Taylor, Effect of high-dose intravenous vitamin C on inflammation in cancer patients, J. Transl. Med., 2012, 10, 1–10 CrossRef PubMed.
  75. N. Mikirova, N. Riordan and J. Casciari, Modulation of cytokines in cancer patients by intravenous ascorbate therapy, Med. Sci. Monit., 2016, 22, 14 CrossRef CAS PubMed.
  76. X. Zhao, M. Liu, C. Li, X. Liu, J. Zhao, H. Ma, S. Zhang and J. Qu, High dose Vitamin C inhibits PD-L1 by ROS-pSTAT3 signal pathway and enhances T cell function in TNBC, Int. Immunopharmacol., 2024, 126, 111321 CrossRef CAS.
  77. C. D. Munialo and V. Kontogiorgos, An investigation into the degradation of ascorbic acid in solutions, J. Food Res., 2014, 2, 106–112 Search PubMed.
  78. H. S. Farah, J. F. Alhmoud, A. Al-Othman, K. M. Alqaisi, A. M. Atoom, K. Shadid, A. Shakya and T. AlQaisi, Effect of pH, temperature and metal salts in different storage conditions on the stability of vitamin C content of yellow bell pepper extracted in aqueous media, Sys. Rev. Pharm., 2020, 11, 661–667 CAS.
  79. V. Gérard, E. Ay, B. Graff, F. Morlet-Savary, C. Galopin, W. Mutilangi and J. Lalevée, Ascorbic acid derivatives as potential substitutes for ascorbic acid to reduce color degradation of drinks containing ascorbic acid and anthocyanins from natural extracts, J. Agric. Food Chem., 2019, 67(43), 12061–12071 CrossRef PubMed.
  80. A. C. Caritá, B. Fonseca-Santos, J. D. Shultz, B. Michniak-Kohn, M. Chorilli and G. R. Leonardi, Vitamin C: One compound, several uses. Advances for delivery, efficiency and stability, Nanomed. Nanotechnol. Biol. Med., 2020, 24, 102117 CrossRef PubMed.
  81. O. Sasidharan, A. Gholap and R. Rastogi, A Review of Clinical Efficacy of Topical Vitamin C and Its Derivatives, Sci. Technol., 2023, 7(2), 20–26 Search PubMed.
  82. A. Dghoughi, F.-E. Nazih, A. Halloub, M. Raji, H. Essabir, M. O. Bensalah and R. Bouhfid, Development of shelf life-extending packaging for vitamin C syrup based on high-density polyethylene and extracted lignin argan shells, Int. J. Biol. Macromol., 2023, 242, 125077 CrossRef CAS PubMed.
  83. M. d. C. Morán, C. Porredon and C. Gibert, Insight into the Antioxidant Activity of Ascorbic Acid-Containing Gelatin Nanoparticles in Simulated Chronic Wound Conditions, Antioxidants, 2024, 13(3), 299 CrossRef PubMed.
  84. Y. Cho, J. T. Kim and H. J. Park, Size-controlled self-aggregated N-acyl chitosan nanoparticles as a vitamin C carrier, Carbohydr. Polym., 2012, 88(3), 1087–1092 CrossRef CAS.
  85. G. Güney, H. M. Kutlu and L. Genç, Preparation and characterization of ascorbic acid loaded solid lipid nanoparticles and investigation of their apoptotic effects, Colloids Surf., B, 2014, 121, 270–280 CrossRef PubMed.
  86. S. Gopi and P. Balakrishnan, Evaluation and clinical comparison studies on liposomal and non-liposomal ascorbic acid (vitamin C) and their enhanced bioavailability, J. Liposome Res., 2021, 31(4), 356–364 CrossRef CAS PubMed.
  87. A. Csorba, G. Katona, M. Budai-Szűcs, D. Balogh-Weiser, A. M. Fadda, C. Caddeo, Á. I. Takács, P. Mátyus, G. T. Balogh and Z. Z. Nagy, Effect of liposomal formulation of ascorbic acid on corneal permeability, Sci. Rep., 2023, 13(1), 3448 CrossRef CAS PubMed.
  88. M. Ren, Z. Han, J. Li, G. Feng and S. Ouyang, Ascorbic acid delivered by mesoporous silica nanoparticles induces the differentiation of human embryonic stem cells into cardiomyocytes, Mater. Sci. Eng. C, 2015, 56, 348–355 CrossRef CAS PubMed.
  89. S. A. Fahmy, A. Ramzy, A. A. Mandour, S. Nasr, A. Abdelnaser, U. Bakowsky and H. M. E.-S. Azzazy, PEGylated chitosan nanoparticles encapsulating ascorbic acid and oxaliplatin exhibit dramatic apoptotic effects against breast cancer cells, Pharmaceutics, 2022, 14(2), 407 CrossRef CAS PubMed.
  90. F. Quiñonero, B. Parra-Torrejón, G. B. Ramírez-Rodríguez, V. Garcés, J. M. Delgado-López, C. Jiménez-Luna, G. Perazzoli, C. Melguizo, J. Prados and R. Ortíz, Combining olaparib and ascorbic acid on nanoparticles to enhance the drug toxic effects in pancreatic cancer, Int. J. Nanomed., 2023, 5075–5093 CrossRef.
  91. M. Sun, Q. Xie, X. Cai, Z. Liu, Y. Wang, X. Dong and Y. Xu, Preparation and characterization of epigallocatechin gallate, ascorbic acid, gelatin, chitosan nanoparticles and their beneficial effect on wound healing of diabetic mice, Int. J. Biol. Macromol., 2020, 148, 777–784 CrossRef CAS PubMed.
  92. R. Phul, C. Kaur, U. Farooq and T. Ahmad, Ascorbic acid assisted synthesis, characterization and catalytic application of copper nanoparticles, Int. J. Mater. Sci. Eng., 2018, 2(4), 90–94 Search PubMed.
  93. K. Sun, J. Qiu, J. Liu and Y. Miao, Preparation and characterization of gold nanoparticles using ascorbic acid as reducing agent in reverse micelles, J. Mater. Sci., 2009, 44(3), 754–758 CrossRef CAS.
  94. M. Luty-Błocho, M. Wojnicki and K. Fitzner, Gold Nanoparticles Formation via Au (III) Complex Ions Reduction with l-Ascorbic Acid, Int. J. Chem. Kinet., 2017, 49(11), 789–797 CrossRef.
  95. A. Sood, V. Arora, J. Shah, R. K. Kotnala and T. K. Jain, Ascorbic acid-mediated synthesis and characterisation of iron oxide/gold core–shell nanoparticles, J. Exp. Nanosci., 2016, 11(5), 370–382 CrossRef CAS.
  96. N. Filipović, D. Ušjak, M. T. Milenković, K. Zheng, L. Liverani, A. R. Boccaccini and M. M. Stevanović, Comparative study of the antimicrobial activity of selenium nanoparticles with different surface chemistry and structure, Front. Bioeng. Biotechnol., 2021, 8, 624621 CrossRef PubMed.
  97. A. Zaid Alkilani, R. Hamed, G. Hussein and S. Alnadi, Nanoemulsion-based patch for the dermal delivery of ascorbic acid, J. Dispersion Sci. Technol., 2022, 43(12), 1801–1811 CrossRef CAS.
  98. L. Maione-Silva, E. G. de Castro, T. L. Nascimento, E. R. Cintra, L. C. Moreira, B. A. S. Cintra, M. C. Valadares and E. M. Lima, Ascorbic acid encapsulated into negatively charged liposomes exhibits increased skin permeation, retention and enhances collagen synthesis by fibroblasts, Sci. Rep., 2019, 9(1), 522 CrossRef PubMed.
  99. S. Duarah, R. D. Durai and V. B. Narayanan, Nanoparticle-in-gel system for delivery of vitamin C for topical application, Drug Delivery Transl. Res., 2017, 7, 750–760 CrossRef CAS PubMed.
  100. P. S. Telang, Vitamin C in dermatology, Indian Dermatol. Online J., 2013, 4(2), 143 CrossRef PubMed.
  101. S. Duarah, R. D. Durai and V. B. Narayanan, Nanoparticle-in-gel system for delivery of vitamin C for topical application, Drug Delivery Transl. Res., 2017, 7(5), 750–760 CrossRef CAS PubMed.
  102. T. E. A. Ardjani, S. Daoudi, M. R. Benaissa and J. R. Alvarez-Idaboy, Strategic design, theoretical insights, synthesis, and unveiling antioxidant potential in a novel ascorbic acid analog, J. Mol. Model., 2024, 30(5), 141 CrossRef CAS PubMed.
  103. C. D. Enescu, L. M. Bedford, G. Potts and F. Fahs, A review of topical vitamin C derivatives and their efficacy, J. Cosmet. Dermatol., 2022, 21(6), 2349–2359 CrossRef PubMed.
  104. M. D. E. L. Prieto, Cosmetic Topical Use of Vitamin C, 2023 Search PubMed.
  105. M. Plaza-Oliver, M. J. Santander-Ortega, L. Castro-Vázquez, V. Rodríguez-Robledo, J. González-Fuentes, P. Marcos, M. V. Lozano and M. M. Arroyo-Jiménez, The role of the intestinal-protein corona on the mucodiffusion behaviour of new nanoemulsions stabilised by ascorbyl derivatives, Colloids Surf., B, 2020, 186, 110740 CrossRef CAS PubMed.
  106. M. Imran, B. Titilayo, M. Adil, Q. Mehmood, S. H. Mustafa and Q. Shen, Ascorbyl palmitate: A comprehensive review on its characteristics, synthesis, encapsulation and applications, Process Biochem., 2024, 142, 68–80 CrossRef CAS.
  107. U. F. Saidu, I. Bulama, I. Abubakar, Y. Zayyana, A. Onu, N. Suleiman, A. Abbas, Y. Saidu and L. Bilbis, Chemical modification of ascorbic acid to L-ascorbyl-6-palmitate: A novel approach for improved antioxidant therapy in traumatic brain injury, J. Exp. Clin. Med., 2024, 41(2), 278–285 Search PubMed.
  108. E. Capecchi, D. Piccinino, C. Nascimben, E. Tomaino, N. Ceccotti Vlas, S. Gabellone and R. Saladino, Biosynthesis of novel ascorbic acid esters and their encapsulation in lignin nanoparticles as carriers and Stabilizing Systems, Int. J. Mol. Sci., 2023, 24(10), 9044 CrossRef CAS PubMed.
  109. L. Frungillo, D. Martins, S. Teixeira, M. C. Anazetti, P. da Silva Melo and N. Durán, Targeted antitumoral dehydrocrotonin nanoparticles with L-ascorbic acid 6-stearate, J. Pharmaceut. Sci., 2009, 98(12), 4796–4807 CrossRef CAS PubMed.
  110. D. A. Navarre, M. Zhu and H. Hellmann, Plant antioxidants affect human and gut health, and their biosynthesis is influenced by environment and reactive oxygen species, Oxygen, 2022, 2(3), 348–370 CrossRef CAS.
  111. X. Chen, R. Liu, X. Liu, C. Xu and X. Wang, L-ascorbic Acid-2-Glucoside inhibits Helicobacter pylori-induced apoptosis through mitochondrial pathway in Gastric Epithelial cells, Biomed. Pharmacother., 2018, 97, 75–81 CrossRef CAS PubMed.
  112. W. Dong, Y. Peng, G. Chen, Z. Xie, W. Xu, W. Zhou, J. Mi, L. Lu, Y. Sun and X. Zeng, 2-O-β-D-Glucopyranosyl-L-ascorbic acid, an ascorbic acid derivative isolated from the fruits of Lycium barbarum L., ameliorates high fructose-induced neuroinflammation in mice: Involvement of gut microbiota and leaky gut, Food Sci. Hum. Wellness, 2024, 13(1), 241–253 CrossRef CAS.
  113. K. Huang, Y. Yan, D. Chen, Y. Zhao, W. Dong, X. Zeng and Y. Cao, Ascorbic acid derivative 2-O-β-D-glucopyranosyl-L-ascorbic acid from the fruit of Lycium barbarum modulates microbiota in the small intestine and colon and exerts an immunomodulatory effect on cyclophosphamide-treated BALB/c mice, J. Agric. Food Chem., 2020, 68(40), 11128–11143 CrossRef CAS PubMed.
  114. J. Y. Choi, J. W. Ha and Y. C. Boo, Multifaceted Effects of L-Cysteine, L-Ascorbic Acid, and Their Derivatives on the Viability and Melanin Synthesis of B16/F10 Cells under Different Conditions, Antioxidants, 2024, 13(3), 330 CrossRef CAS PubMed.
  115. E. Wynendaele, J. Urlaub, X. Xu, U. Holzgrabe and B. De Spiegeleer, Quality Control of L-Ascorbic Acid 2-Phosphate Magnesium Using Reversed-Phase Liquid Chromatography, J. Chromatogr. Sci., 2024, bmae031 Search PubMed.
  116. A. Harej, A. M. Macan, V. Stepanić, M. Klobučar, K. Pavelić, S. K. Pavelić and S. Raić-Malić, The antioxidant and antiproliferative activities of 1, 2, 3-triazolyl-L-ascorbic acid derivatives, Int. J. Mol. Sci., 2019, 20(19), 4735 CrossRef CAS PubMed.
  117. S. Shibuya, I. Sakaguchi, S. Ito, E. Kato, K. Watanabe, N. Izuo and T. Shimizu, Topical application of trisodium ascorbyl 6-palmitate 2-phosphate actively supplies ascorbate to skin cells in an ascorbate transporter-independent manner, Nutrients, 2017, 9(7), 645 CrossRef PubMed.
  118. C. Jacques, C. Genies, D. Bacqueville, A. Tourette, N. Borotra, F. Chaves, F. Sanches, A. L. Gaudry, S. Bessou-Touya and H. Duplan, Ascorbic acid 2-glucoside: An ascorbic acid pro-drug with longer-term antioxidant efficacy in skin, Int. J. Cosmet. Sci., 2021, 43(6), 691–702 CrossRef CAS PubMed.
  119. R. Gref, C. Deloménie, A. Maksimenko, E. Gouadon, G. Percoco, E. Lati, D. Desmaële, F. Zouhiri and P. Couvreur, Vitamin C–squalene bioconjugate promotes epidermal thickening and collagen production in human skin, Sci. Rep., 2020, 10(1), 16883 CrossRef CAS PubMed.
  120. L. Xiao and N. Miwa, The lipophilic vitamin C derivative, 6-O-palmitoylascorbate protects human keratinocytes and 3d-human skin equivalents against X-ray-induced oxidative stress and apoptosis more markedly than L-ascorbic acid, J. Cell. Biochem., 2017, 118(2), 318–329 CrossRef CAS PubMed.
  121. R. Mohamed, S. Tarannum, M. Yariswamy, H. K. Vivek, J. M. Siddesha, N. Angaswamy and B. S. Vishwanath, Ascorbic acid 6-palmitate: a potent inhibitor of human and soybean lipoxygenase-dependent lipid peroxidation, J. Pharm. Pharmacol., 2014, 66(6), 769–778 CrossRef CAS PubMed.
  122. I. Yamamoto, A. Tai, Y. Fujinami, K. Sasaki and S. Okazaki, Synthesis and characterization of a series of novel monoacylated ascorbic acid derivatives, 6-O-acyl-2-O-α-D-glucopyranosyl-L-ascorbic acids, as skin antioxidants, J. Med. Chem., 2002, 45(2), 462–468 CrossRef CAS PubMed.
  123. A. Tai, D. Kawasaki, K. Sasaki, E. Gohda and I. Yamamoto, Synthesis and characterization of 6-O-acyl-2-O-α-D-glucopyranosyl-L-ascorbic acids with a branched-acyl chain, Chem. Pharm. Bull., 2003, 51(2), 175–180 CrossRef CAS PubMed.
  124. H. Shibayama, K. Ueda, K. Yoshio, S. Matsuda, M. Hisama and M. Miyazawa, Synthesis and characterization of new ascorbic derivative: sodium isostearyl 2-OL-Ascorbyl Phosphate, J. Oleo Sci., 2005, 54(11), 601–608 CrossRef CAS.
  125. N. Taira, Y. Katsuyama, M. Yoshioka, O. Muraoka and T. Morikawa, Structural requirements of alkylglyceryl-L-ascorbic acid derivatives for melanogenesis inhibitory activity, Int. J. Mol. Sci., 2018, 19(4), 1144 CrossRef PubMed.
  126. K. Wittine, M. S. Babić, D. Makuc, J. Plavec, S. K. Pavelić, M. Sedić, K. Pavelić, P. Leyssen, J. Neyts and J. Balzarini, Novel 1, 2, 4-triazole and imidazole derivatives of L-ascorbic and imino-ascorbic acid: Synthesis, anti-HCV and antitumor activity evaluations, Bioorg. Med. Chem., 2012, 20(11), 3675–3685 CrossRef CAS PubMed.
  127. T. Gazivoda, K. Wittine, I. Lovrić, D. Makuc, J. Plavec, M. Cetina, D. Mrvoš-Sermek, L. Šuman, M. Kralj and K. Pavelić, Synthesis, structural studies, and cytostatic evaluation of 5, 6-di-O-modified L-ascorbic acid derivatives, Carbohydr. Res., 2006, 341(4), 433–442 CrossRef CAS PubMed.
  128. S. R. Deshmukh, A. S. Nalkar, A. P. Sarkate, S. V. Tiwari, D. K. Lokwani and S. R. Thopate, Design, synthesis, and biological evaluation of novel 2, 3-Di-O-Aryl/Alkyl sulfonate derivatives of l-ascorbic acid: Efficient access to novel anticancer agents via in vitro screening, tubulin polymerization inhibition, molecular docking study and ADME predictions, Bioorg. Chem., 2024, 147, 107402 CrossRef CAS PubMed.
  129. B. R. K. Shyamlal, L. Yadav, M. K. Tiwari, M. Mathur, J. I. Prikhodko, I. V. Mashevskaya, D. K. Yadav and S. Chaudhary, Synthesis, bioevaluation, structure-activity relationship and docking studies of natural product inspired (Z)-3-benzylideneisobenzofuran-1 (3H)-ones as highly potent antioxidants and antiplatelet agents, Sci. Rep., 2020, 10(1), 2307 CrossRef CAS PubMed.
  130. Y. Fujinami, A. Tai and I. Yamamoto, Radical scavenging activity against 1, 1-diphenyl-2-picrylhydrazyl of ascorbic acid 2-glucoside (AA-2G) and 6-acyl-AA-2G, Chem. Pharm. Bull., 2001, 49(5), 642–644 CrossRef CAS PubMed.
  131. L. Saftić Martinović, N. Birkic, V. Miletić, R. Antolović, D. Štanfel and K. Wittine, Antioxidant activity, stability in aqueous medium and molecular docking/dynamics study of 6-Amino-and N-Methyl-6-amino-L-ascorbic acid, Int. J. Mol. Sci., 2023, 24(2), 1410 CrossRef PubMed.
  132. J. Takebayashi, A. Tai, E. Gohda and I. Yamamoto, Characterization of the radical-scavenging reaction of 2-O-substituted ascorbic acid derivatives, AA-2G, AA-2P, and AA-2S: a kinetic and stoichiometric study, Biol. Pharm. Bull., 2006, 29(4), 766–771 CrossRef CAS PubMed.
  133. R. Mohamed, K. K. Dharmappa, S. Tarannum, N. M. Jameel, S. A. Kannum, H. S. Ashrafulla, L. Rai, C. J. Souza, M. A. Shekhar and B. S. Vishwanath, Chemical modification of ascorbic acid and evaluation of its lipophilic derivatives as inhibitors of secretory phospholipase A2 with anti-inflammatory activity, Mol. Cell. Biochem., 2010, 345(1), 69–76 CrossRef CAS PubMed.
  134. S. D. Brinkevich, E. I. Boreko, O. V. Savinova, N. I. Pavlova and O. I. Shadyro, Radical-regulating and antiviral properties of ascorbic acid and its derivatives, Bioorg. Med. Chem. Lett., 2012, 22(7), 2424–2427 CrossRef CAS PubMed.
  135. T. Gazivoda, M. Plevnik, J. Plavec, S. Kraljević, M. Kralj, K. Pavelić, J. Balzarini, E. De Clercq, M. Mintas and S. Raić-Malić, The novel pyrimidine and purine derivatives of L-ascorbic acid: synthesis, one-and two-dimensional 1H and 13C NMR study, cytostatic and antiviral evaluation, Bioorg. Med. Chem., 2005, 13(1), 131–139 CrossRef CAS PubMed.
  136. A. Meves, S. N. Stock, A. Beyerle, M. R. Pittelkow and D. Peus, Vitamin C derivative ascorbyl palmitate promotes ultraviolet-B-induced lipid peroxidation and cytotoxicity in keratinocytes, J. Invest. Dermatol., 2002, 119(5), 1103–1108 CrossRef CAS PubMed.
  137. G. Shinde, P. Desai, S. Shelke, R. Patel, G. Bangale and D. Kulkarni, Mometasone furoate-loaded aspasomal gel for topical treatment of psoriasis: formulation, optimization, in vitro and in vivo performance, J. Dermatol. Treat., 2022, 33(2), 885–896 CrossRef CAS PubMed.
  138. M. H. Aboul-Einien, S. M. Kandil, E. M. Abdou, H. M. Diab and M. S. E. Zaki, Ascorbic acid derivative-loaded modified aspasomes: formulation, in vitro, ex vivo and clinical evaluation for melasma treatment, J. Liposome Res., 2020, 30(1), 54–67 CrossRef CAS PubMed.
  139. A. Leekha, B. S. Gurjar, A. Tyagi, M. A. Rizvi and A. K. Verma, Vitamin C in synergism with cisplatin induces cell death in cervical cancer cells through altered redox cycling and p53 upregulation, J. Cancer Res. Clin. Oncol., 2016, 142(12), 2503–2514 CrossRef CAS PubMed.
  140. D. Gopinath, D. Ravi, B. R. Rao, S. S. Apte, D. Renuka and D. Rambhau, Ascorbyl palmitate vesicles (Aspasomes): formation, characterization and applications, Int. J. Pharm., 2004, 271(1–2), 95–113 CrossRef CAS PubMed.
  141. J. Li, C. Guo, F. Feng, A. Fan, Y. Dai, N. Li, D. Zhao, X. Chen and Y. Lu, Co-delivery of docetaxel and palmitoyl ascorbate by liposome for enhanced synergistic antitumor efficacy, Sci. Rep., 2016, 6(1), 1–8 CrossRef CAS PubMed.
  142. D. Paneva, N. Manolova, M. Argirova and I. Rashkov, Antibacterial electrospun poly (ε-caprolactone)/ascorbyl palmitate nanofibrous materials, Int. J. Pharm., 2011, 416(1), 346–355 CrossRef CAS PubMed.
  143. F. A. Andersen, Final report on the safety assessment of ascorbyl palmitate, ascorbyl dipalmitate, ascorbyl stearate, erythorbic acid, and sodium erythorbate, Int. J. Toxicol., 1999, 18, 1–26 Search PubMed.
  144. J. Kristl, B. Volk, M. Gašperlin, M. Šentjurc and P. Jurkovič, Effect of colloidal carriers on ascorbyl palmitate stability, Eur. J. Pharmaceut. Sci., 2003, 19(4), 181–189 CrossRef CAS PubMed.
  145. G. Shinde, P. Desai, S. Shelke, R. Patel, G. Bangale and D. Kulkarni, Mometasone furoate-loaded aspasomal gel for topical treatment of psoriasis: Formulation, optimization, in vitro and in vivo performance, J. Dermatol. Treat., 2022, 33(2), 885–896 CrossRef CAS PubMed.
  146. K. A. Naidu, Vitamin C in human health and disease is still a mystery? An overview, Nutr. J., 2003, 2(1), 1–10 CrossRef PubMed.
  147. G. G. M. D'Souza, T. Wang, K. Rockwell and V. P. Torchilin, Surface modification of pharmaceutical nanocarriers with ascorbate residues improves their tumor-cell association and killing and the cytotoxic action of encapsulated paclitaxel in vitro, Pharm. Res., 2008, 25(11), 2567–2572 CrossRef PubMed.
  148. G. G. M. D'Souza, T. Wang, K. Rockwell and V. P. Torchilin, Surface modification of pharmaceutical nanocarriers with ascorbate residues improves their tumor-cell association and killing and the cytotoxic action of encapsulated paclitaxel in vitro, Pharm. Res., 2008, 25(11), 2567–2572 CrossRef PubMed.
  149. B. M. Abdel-Hady, B. Ekram, W. E. G. Müller, A. A. M. Gad, X. Wang, H. C. Schröder and E. Tolba, Ascorbyl palmitate–PCL fiber mats loaded with strontium polyphosphate nanoparticles for guided bone regeneration, Polym. Bull., 2024, 81(4), 3355–3374 CrossRef CAS.
  150. M. Tabak, R. Armon, G. Rosenblat, E. Stermer and I. Neeman, Diverse effects of ascorbic acid and palmitoyl ascorbate on Helicobacter pylori survival and growth, FEMS Microbiol. Lett., 2003, 224(2), 247–253 CrossRef CAS PubMed.
  151. S. Han, Structure of Ascorbyl Palmitate Bilayers (Aspasomes) from Molecular Dynamics Simulation, Bull. Korean Chem. Soc., 2018, 39(7), 887–890 CrossRef CAS.
  152. S. Ghosh, B. Mukherjee, S. Chaudhuri, T. Roy, A. Mukherjee and S. Sengupta, Methotrexate aspasomes against rheumatoid arthritis: optimized hydrogel loaded liposomal formulation with in vivo evaluation in Wistar rats, AAPS PharmSciTech, 2018, 19(3), 1320–1336 CrossRef CAS PubMed.
  153. M. Pokorski, M. Marczak, A. Dymecka and P. Suchocki, Ascorbyl palmitate as a carrier of ascorbate into neural tissues, J. Biomed. Sci., 2003, 10(2), 193–198 CrossRef CAS PubMed.
  154. P. Jurkovič, M. Šentjurc, M. Gašperlin, J. Kristl and S. Pečar, Skin protection against ultraviolet induced free radicals with ascorbyl palmitate in microemulsions, Eur. J. Pharm. Biopharm., 2003, 56(1), 59–66 CrossRef PubMed.
  155. Ü. Gönüllü, G. Yener, M. Üner and T. Incegül, Moisturizing potentials of ascorbyl palmitate and calcium ascorbate in various topical formulations, Int. J. Cosmet. Sci., 2004, 26(1), 31–36 CrossRef PubMed.
  156. M. Üner, S. A. Wissing, G. Yener and R. H. Müller, Skin moisturizing effect and skin penetration of ascorbyl palmitate entrapped in solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) incorporated into hydrogel, Int. J. Pharmaceut. Sci., 2005, 60(10), 751–755 Search PubMed.
  157. V. Teeranachaideekul, E. B. Souto, R. H. Müller and V. B. Junyaprasert, Physicochemical characterization and in vitro release studies of ascorbyl palmitate-loaded semi-solid nanostructured lipid carriers (NLC gels), J. Microencapsul., 2008, 25(2), 111–120 CrossRef CAS PubMed.
  158. R. R. Sawant, O. Vaze, G. G. M. D'Souza, K. Rockwell and V. P. Torchilin, Palmitoyl ascorbate-loaded polymeric micelles: cancer cell targeting and cytotoxicity, Pharm. Res., 2011, 28(2), 301–308 CrossRef CAS.
  159. F. R. Favarin, É. M. Forrati, V. A. Bassoto, S. da Silva Gündel, M. C. Velho, C. M. Ledur, C. M. Verdi, J. G. Lemos, M. R. Sagrillo and S. B. Fagan, Ascorbic acid and ascorbyl palmitate-loaded liposomes: Development, characterization, stability evaluation, in vitro security profile, antimicrobial and antioxidant activities, Food Chem., 2024, 460, 140569 CrossRef CAS PubMed.
  160. L. Li, H. Wang, J. Ye, Y. Chen, R. Wang, D. Jin and Y. Liu, Mechanism Study on Nanoparticle Negative Surface Charge Modification by Ascorbyl Palmitate and Its Improvement of Tumor Targeting Ability, Molecules, 2022, 27(14), 4408 CrossRef CAS PubMed.
  161. J. Zhai, J. Mantaj and D. Vllasaliu, Ascorbyl palmitate hydrogel for local, intestinal delivery of macromolecules, Pharmaceutics, 2018, 10(4), 188 CrossRef CAS PubMed.
  162. M. Francois, T. Luhong and W. Honghua, Immobilization of Human Sperms by Gossypol Incorporated into l-Ascorbyl Palmitate Coagel, Biomed. J. Sci. Tech. Res., 2019, 14(5), 1–9 Search PubMed.
  163. E. Elmowafy, S. Hammad and M. E. Soliman, Exploiting Ascorbyl Palmitate Assisted-Chondroitin Sulphate/Chitosan Nanoparticles for Intra-articular Delivery of Lipoic Acid, Carbohydr. Polym. Technol. Appl., 2024, 100459 CAS.
  164. Y. Inoue, M. Hibino, I. Murata and I. Kanamoto, A nanocarrier skin-targeted drug delivery system using an ascorbic acid derivative, Pharm. Res., 2018, 35, 1–14 CrossRef CAS PubMed.
  165. K. Moribe, S. Maruyama, Y. Inoue, T. Suzuki, T. Fukami, K. Tomono, K. Higashi, Y. Tozuka and K. Yamamoto, Ascorbyl dipalmitate/PEG-lipid nanoparticles as a novel carrier for hydrophobic drugs, Int. J. Pharm., 2010, 387(1–2), 236–243 CrossRef CAS.
  166. Z. Chen, K. Higashi, R. Shidara, K. Ueda, T. Morita, W. Limwikrant, K. Yamamoto and K. Moribe, The nanostructure of rod-like ascorbyl dipalmitate nanoparticles stabilized by a small amount of DSPE-PEG, Int. J. Pharm., 2021, 602, 120599 CrossRef CAS PubMed.
  167. M. Plaza-Oliver, A. Beloqui, M. J. Santander-Ortega, L. Castro-Vázquez, V. Rodríguez-Robledo, M. M. Arroyo-Jiménez, V. Préat and M. V. Lozano, Ascorbyl-dipalmitate-stabilised nanoemulsions as a potential localised treatment of inflammatory bowel diseases, Int. J. Pharm., 2020, 586, 119533 CrossRef CAS PubMed.
  168. P. Sonkaew, A. Sane and P. Suppakul, Antioxidant activities of curcumin and ascorbyl dipalmitate nanoparticles and their activities after incorporation into cellulose-based packaging films, J. Agric. Food Chem., 2012, 60(21), 5388–5399 CrossRef CAS PubMed.
  169. S. Jain, N. Patel, M. K. Shah, P. Khatri and N. Vora, Recent advances in lipid-based vesicles and particulate carriers for topical and transdermal application, J. Pharmaceut. Sci., 2017, 106(2), 423–445 CrossRef CAS PubMed.
  170. R. Arora, S. S. Katiyar, V. Kushwah and S. Jain, Solid lipid nanoparticles and nanostructured lipid carrier-based nanotherapeutics in treatment of psoriasis: a comparative study, Expet Opin. Drug Deliv., 2017, 14(2), 165–177 CrossRef CAS PubMed.
  171. S. B. More, T. D. Nandgude and S. S. Poddar, Vesicles as a tool for enhanced topical drug delivery, Asian J. Pharm., 2016, 10(3), S196–S208 CAS.
  172. K. Thakur, G. Sharma, B. Singh and O. P. Katare, Topical drug delivery of anti-infectives employing lipid-based nanocarriers: Dermatokinetics as an important tool, Curr. Pharm. Des., 2018, 24(43), 5108–5128 CrossRef CAS PubMed.
  173. M. Sala, R. Diab, A. Elaissari and H. Fessi, Lipid nanocarriers as skin drug delivery systems: Properties, mechanisms of skin interactions and medical applications, Int. J. Pharm., 2018, 535(1–2), 1–17 CrossRef CAS PubMed.
  174. M. Estanqueiro, J. Conceição, M. H. Amaral and J. M. S. Lobo, The role of liposomes and lipid nanoparticles in the skin hydration, in Nanobiomaterials in Galenic Formulations and Cosmetics, Elsevier, 2016, pp. 297–326 Search PubMed.
  175. M. Elmowafy, Skin penetration/permeation success determinants of nanocarriers: Pursuit of a perfect formulation, Colloids Surf., B, 2021, 203, 111748 CrossRef CAS PubMed.
  176. G. Cevc, Transfersomes, liposomes and other lipid suspensions on the skin: permeation enhancement, vesicle penetration, and transdermal drug delivery, Crit. Rev. Ther. Drug Carrier Syst., 1996, 13(3–4) CAS.
  177. V. K. Rapalli, V. Kaul, T. Waghule, S. Gorantla, S. Sharma, A. Roy, S. K. Dubey and G. Singhvi, Curcumin loaded nanostructured lipid carriers for enhanced skin retained topical delivery: optimization, scale-up, in-vitro characterization and assessment of ex-vivo skin deposition, Eur. J. Pharm. Sci., 2020, 152, 105–438 CrossRef PubMed.
  178. A. C. Sintov and H. V. Levy, A microemulsion-based system for the dermal delivery of therapeutics, Innovat. Pharmaceut. Technol., 2007, 23, 68–72 CAS.
  179. S. S. Amer, M. Nasr, R. T. A. Abdel-Aziz, N. H. Moftah, A. El Shaer, E. Polycarpou, W. Mamdouh and O. Sammour, Cosm-nutraceutical nanovesicles for acne treatment: Physicochemical characterization and exploratory clinical experimentation, Int. J. Pharm., 2020, 577, 119092 CrossRef CAS PubMed.
  180. S. Hatem, M. Nasr, N. H. Moftah, M. H. Ragai, A. S. Geneidi and S. A. Elkheshen, Melatonin vitamin C-based nanovesicles for treatment of androgenic alopecia: design, characterization and clinical appraisal, Eur. J. Pharmaceut. Sci., 2018, 122, 246–253 CrossRef CAS PubMed.
  181. S. S. Amer, M. Nasr, R. T. A. Abdel-Aziz, N. H. Moftah, A. El Shaer, E. Polycarpou, W. Mamdouh and O. Sammour, Cosm-nutraceutical nanovesicles for acne treatment: Physicochemical characterization and exploratory clinical experimentation, Int. J. Pharm., 2020, 577, 119092 CrossRef CAS PubMed.
  182. P. Ozturk, O. Arican, E. B. Kurutas, T. Karakas and M. Gungor, Local oxidative stress in interdigital tinea pedis, J. Dermatol., 2013, 40(2), 114–117 CrossRef CAS PubMed.
  183. R. Jukanti, G. Devraj, A. S. Shashank and R. Devraj, Biodistribution of ascorbyl palmitate loaded doxorubicin pegylated liposomes in solid tumor bearing mice, J. Microencapsul., 2011, 28(2), 142–149 CrossRef CAS PubMed.
  184. S. Hatem, M. Nasr, N. H. Moftah, M. H. Ragai, A. S. Geneidi and S. A. Elkheshen, Melatonin vitamin C-based nanovesicles for treatment of androgenic alopecia: design, characterization and clinical appraisal, Eur. J. Pharm. Sci., 2018, 122, 246–253 CrossRef CAS PubMed.
  185. R. M. Khalil, A. Abdelbary, S. K. E. Arini, M. Basha, H. A. El-Hashemy and F. Farouk, Development of tizanidine loaded aspasomes as transdermal delivery system: ex-vivo and in-vivo evaluation, J. Liposome Res., 2021, 31(1), 19–29 CrossRef CAS PubMed.
  186. N. d'Avanzo, M. C. Cristiano, L. Di Marzio, M. C. Bruno, D. Paolino, C. Celia and M. Fresta, Multidrug Idebenone/Naproxen Co-loaded Aspasomes for Significant in vivo Anti-inflammatory Activity, ChemMedChem, 2022, 17(9), e202200067 CrossRef PubMed.
  187. C. Lamie, E. Elmowafy, D. A. Attia, M. M. Elmazar and N. D. Mortada, Diversifying the skin cancer-fighting worthwhile frontiers: How relevant are the itraconazole/ascorbyl palmitate nanovectors?, Nanomed. Nanotechnol. Biol. Med., 2022, 43, 102561 CrossRef CAS PubMed.
  188. C. Lamie, E. Elmowafy, M. H. Ragaie, D. A. Attia and N. D. Mortada, Assessment of antifungal efficacy of itraconazole loaded aspasomal cream: comparative clinical study, Drug Deliv., 2022, 29(1), 1345–1357 CrossRef CAS PubMed.
  189. M. H. Aboul-Einien, S. M. Kandil, E. M. Abdou, H. M. Diab and M. S. E. Zaki, Ascorbic acid derivative-loaded modified aspasomes: formulation, in vitro, ex vivo and clinical evaluation for melasma treatment, J. Liposome Res., 2020, 30(1), 54–67 CrossRef CAS PubMed.
  190. M. Salah Eleleemy, M. H. Ragaie, B. Hamdy Amin, M. Nasr and O. A. Sammour, Enhanced skin penetration and clinical antifungal activity of eugenol encapsulated in aspasomes, Pharm. Dev. Technol., 2025, 30(4), 372–384 CrossRef CAS PubMed.
  191. M. Shahid, F. Usman and S. R. Abbas, Topical Hydrogel Formulation based on Nano-Aspasomes Encapsulating Caffeic Acid Retinoic Acid for Antiaging Effects, J. Drug Delivery Sci. Technol., 2025, 107360 CrossRef CAS.
  192. S. Hatem, N. H. Moftah, M. H. Ragai and E. El-Maghawry, Development of gallic acid loaded composite nanovesicles for the topical treatment of acne: optimization, characterization, and clinical investigation, Pharm. Dev. Technol., 2024, 29(8), 899–911 CrossRef CAS PubMed.
  193. M. Jaber, B. Jaber, S. Hamed and H. S. AlKhatib, Preparation and evaluation of ascorbyl glucoside and ascorbic acid solid in oil nanodispersions for corneal epithelial wound healing, Int. J. Pharm., 2022, 627, 122227 CrossRef CAS PubMed.
  194. X. Tao, L. Su and J. Wu, Current studies on the enzymatic preparation 2-O-α-d-glucopyranosyl-l-ascorbic acid with cyclodextrin glycosyltransferase, Crit. Rev. Biotechnol., 2019, 39(2), 249–257 CrossRef CAS PubMed.
  195. M. D. Belsito, C. D. Klaassen, D. C. Liebler, J. G. Marks Jr, R. C. Shank, Safety Assessment of Ascorbyl Glucoside and Sodium Ascorbyl Glucoside as Used in Cosmetics, Cosmetic Ingredient Review Expert Panel Members, 2019, vol. 134, p. 14 Search PubMed.
  196. A. C. Caritá, B. Fonseca-Santos, J. D. Shultz, B. Michniak-Kohn, M. Chorilli and G. R. Leonardi, Vitamin C: One compound, several uses. Advances for delivery, efficiency and stability, Nanomed. Nanotechnol. Biol. Med., 2020, 24, 102–117 CrossRef PubMed.
  197. I. Yamamoto, N. Muto, E. Nagata, T. Nakamura and Y. Suzuki, Formation of a stable L-ascorbic acid α-glucoside by mammalian α-glucosidase-catalyzed transglucosylation, Biochim. Biophys. Acta Gen. Subj., 1990, 1035(1), 44–50 CrossRef CAS PubMed.
  198. H. Matsukawa, T. Yagi, H. Matsuda, H. Kawahara, I. Yamamoto, J. Matsuoka and N. Tanaka, Ascorbic acid 2-glucoside prevents sinusoidal endothelial cell apoptosis in supercooled preserved grafts in rat liver transplantation, in Transplantation proceedings, Elsevier, USA, 2000, vol. 32(2), pp. 313–317. Search PubMed.
  199. I. Yamamoto, A. Tai, Y. Fujinami, K. Sasaki and S. Okazaki, Synthesis and characterization of a series of novel monoacylated ascorbic acid derivatives, 6-O-acyl-2-O-α-D-glucopyranosyl-L-ascorbic acids, as skin antioxidants, J. Med. Chem., 2002, 45(2), 462–468 CrossRef CAS PubMed.
  200. W.-Y. Huang, P.-C. Lee, L.-K. Huang, L.-P. Lu and W. C. Liao, Stability studies of ascorbic acid 2-glucoside in cosmetic lotion using surface response methodology, Bioorg. Med. Chem. Lett., 2013, 23(6), 1583–1587 CrossRef CAS PubMed.
  201. Y. Kumano, T. Sakamoto, M. Egawa, I. Iwai, M. Tanaka and I. Yamamoto, In vitro and in vivo prolonged biological activities of novel vitamin C derivative, 2-O-α-D-glucopyranosyl-L-ascorbic acid (AA-2G), in cosmetic fields, J. Nutr. Sci. Vitaminol., 1998, 44(3), 345–359 CrossRef CAS PubMed.
  202. Y. Hanada, A. Iomori, R. Ishii, E. Gohda and A. Tai, Protection of free radical-induced cytotoxicity by 2-O-α-d-glucopyranosyl-l-ascorbic acid in human dermal fibroblasts, Biosc. Biotech. Biochem., 2014, 78(2), 301–306 CrossRef CAS PubMed.
  203. Y. Inoue, S. Yoshimura, Y. Tozuka, K. Moribe, T. Kumamoto, T. Ishikawa and K. Yamamoto, Application of ascorbic acid 2-glucoside as a solubilizing agent for clarithromycin: solubilization and nanoparticle formation, Int. J. Pharm., 2007, 331(1), 38–45 CrossRef CAS PubMed.
  204. K. Moribe, W. Limwikrant, K. Higashi and K. Yamamoto, Drug nanoparticle formulation using ascorbic acid derivatives, J. Drug Deliv., 2011, 2011, 9 Search PubMed.
  205. Y.-C. Lin, In Vitro Evaluation of Permeation Ability and In Vivo Whitening of Ascorbic Acid 2-Glucoside in Microemulsion, Int. J. Pharm. Sci. Rev. Res., 2016, 14178(16), 11 Search PubMed.
  206. C. Jacques, C. Genies, D. Bacqueville, A. Tourette, N. Borotra, F. Chaves, F. Sanches, A. L. Gaudry, S. Bessou-Touya and H. Duplan, Ascorbic acid 2-glucoside: An ascorbic acid pro-drug with longer-term antioxidant efficacy in skin, Int. J. Cosmet. Sci., 2021, 43(6), 691–702 CrossRef CAS PubMed.
  207. C. Lamie, E. Elmowafy, D. Attia and N. D. Mortada, Glucospanlastics: innovative antioxidant and anticancer ascorbyl-2-glucoside vesicles for striking topical performance of repurposed itraconazole, RSC Adv., 2024, 14(36), 26524–26543 RSC.
  208. M. Haramoto, H. Tatemoto and N. Muto, Essential role of ascorbic acid in neural differentiation and development: High levels of ascorbic acid 2-glucoside effectively enhance nerve growth factor-induced neurite formation and elongation in PC12 cells, J. Health Sci., 2008, 54(1), 43–49 Search PubMed.
  209. D. M. Maliakel, T. V. Kagiya and C. K. K. Nair, Prevention of cisplatin-induced nephrotoxicity by glucosides of ascorbic acid and α-tocopherol, Exp. Toxicol. Pathol., 2008, 60(6), 521–527 CrossRef CAS PubMed.
  210. Y. Shimada, H. Tai, A. Tanaka, I. Ikezawa-Suzuki, K. Takagi, Y. Yoshida and H. Yoshie, Effects of ascorbic acid on gingival melanin pigmentation in vitro and in vivo, J. Periodontol., 2009, 80(2), 317–323 CrossRef CAS PubMed.
  211. D. Mathew, T. V. Kagiya and C. K. K. Nair, Protection of gastrointestinal and haematopoietic systems by ascorbic acid-2-glucoside in mice exposed to whole-body gamma radiation, Int. J. Low Radiat., 2010, 7(5), 380–392 CrossRef CAS.
  212. T. G. Jenkins, K. I. Aston and D. T. Carrell, Supplementation of cryomedium with ascorbic acid–2-glucoside (AA2G) improves human sperm post-thaw motility, Fertil. Steril., 2011, 95(6), 2001–2004 CrossRef CAS PubMed.
  213. H. Moteki, Y. Shimamura, M. Kimura and M. Ogihara, Signal transduction pathway for L-ascorbic acid-and L-ascorbic acid 2-glucoside-induced DNA synthesis and cell proliferation in primary cultures of adult rat hepatocytes, Eur. J. Pharmacol., 2012, 683(1–3), 276–284 CrossRef CAS PubMed.
  214. Y. Hanada, A. Iomori, R. Ishii, E. Gohda and A. Tai, Protection of free radical-induced cytotoxicity by 2-O-α-d-glucopyranosyl-l-ascorbic acid in human dermal fibroblasts, Biosc. Biotech. Biochem., 2014, 78(2), 301–306 CrossRef CAS PubMed.
  215. M. Kimura, H. Moteki, M. Uchida, H. Natsume and M. Ogihara, L-ascorbic acid-and L-ascorbic acid 2-glucoside accelerate in vivo liver regeneration and lower serum alanine aminotransaminase activity in 70% partially hepatectomized rats, Biol. Pharm. Bull., 2014, 8(2), 13–39 Search PubMed.
  216. K. Miura and A. Tai, 2-O-α-D-Glucopyranosyl-l-ascorbic acid as an antitumor agent for infusion therapy, Biochem. Biophys. Rep., 2017, 10, 232–236 Search PubMed.
  217. X. Chen, R. Liu, X. Liu, C. Xu and X. Wang, L-ascorbic Acid-2-Glucoside inhibits Helicobacter pylori-induced apoptosis through mitochondrial pathway in Gastric Epithelial cells, Biomed. Pharmacother., 2018, 97, 75–81 CrossRef CAS PubMed.
  218. J. Maeda, A. J. Allum, J. T. Mussallem, C. E. Froning, A. H. Haskins, M. A. Buckner, C. D. Miller and T. A. Kato, Ascorbic acid 2-glucoside pretreatment protects cells from ionizing radiation, UVC, and short wavelength of UVB, Genes, 2020, 11(3), 238 CrossRef CAS PubMed.
  219. Y. Yi, M. Wu, X. Zhou, M. Xiong, Y. Tan, H. Yu, Z. Liu, Y. Wu and Q. Zhang, Ascorbic acid 2-glucoside preconditioning enhances the ability of bone marrow mesenchymal stem cells in promoting wound healing, Stem Cell Res. Ther., 2022, 13(1), 1–17 CrossRef PubMed.
  220. Y. Ito, T. Yamamoto, K. Miyai, J. Take, H. Scherthan, A. Rommel, S. Eder, K. Steinestel, A. Rump and M. Port, Ascorbic acid-2 glucoside mitigates intestinal damage during pelvic radiotherapy in a rat bladder tumor model, Int. J. Radiat. Biol., 2022, 98(5), 942–957 CrossRef CAS PubMed.
  221. J. D. Shultz, G. R. Leonardi, S. R. A. Bertolino, S. L. Cuffini, H. Mohd, A. C. Caritá, W. Luiz-Silva, P. Shah, W. G. T. Chambi and B. Michniak-Kohn, Design and development of raw clay-based formulations emulsions loaded with ascorbyl glucoside, in vitro evaluations on topical delivery and cell viability, J. Dispersion Sci. Technol., 2024, 45(4), 731–742 CrossRef CAS.
  222. Y. Inoue, S. Yoshimura, Y. Tozuka, K. Moribe, T. Kumamoto, T. Ishikawa and K. Yamamoto, Application of ascorbic acid 2-glucoside as a solubilizing agent for clarithromycin: solubilization and nanoparticle formation, Int. J. Pharm., 2007, 331(1), 38–45 CrossRef CAS PubMed.
  223. K. M. R. Srivalli and B. Mishra, Improved aqueous solubility and antihypercholesterolemic activity of ezetimibe on formulating with hydroxypropyl-β-cyclodextrin and hydrophilic auxiliary substances, AAPS PharmSciTech, 2016, 17(2), 272–283 CrossRef CAS PubMed.
  224. H. Onoda, Y. Inoue, T. Ezawa, I. Murata, T. Chantadee, S. Limmatvapirat, T. Oguchi and I. Kanamoto, Preparation and characterization of triamterene complex with ascorbic acid derivatives, Drug Dev. Ind. Pharm., 2020, 46(12), 2032–2040 CrossRef CAS PubMed.
  225. J. Pasquet, Y. Chevalier, E. Couval, D. Bouvier and M.-A. Bolzinger, Zinc oxide as a new antimicrobial preservative of topical products: Interactions with common formulation ingredients, Int. J. Pharm., 2015, 479(1), 88–95 CrossRef CAS PubMed.
  226. A. Meščić, A. Šalić, T. Gregorić, B. Zelić and S. Raić-Malić, Continuous flow-ultrasonic synergy in click reactions for the synthesis of novel 1, 2, 3-triazolyl appended 4, 5-unsaturated L-ascorbic acid derivatives, RSC Adv., 2017, 7(2), 791–800 RSC.
  227. H.-E. Ji, S.-Y. Kim, H. So, V. Prayitno, K.-T. Lee and J.-A. Shin, A Novel Eco-Friendly Process for the Synthesis and Purification of Ascorbyl-6-Oleates, Foods, 2024, 14(1), 70 CrossRef PubMed.
  228. T. S. Kim, E. A. Decker and J. Lee, Antioxidant capacities of α-tocopherol, trolox, ascorbic acid, and ascorbyl palmitate in riboflavin photosensitized oil-in-water emulsions, Food Chem., 2012, 133(1), 68–75 CrossRef CAS.
  229. L. Li, H. Wang, J. Ye, Y. Chen, R. Wang, D. Jin and Y. Liu, Mechanism study on nanoparticle negative surface charge modification by ascorbyl palmitate and its improvement of tumor targeting ability, Molecules, 2022, 27(14), 4408 CrossRef CAS.
  230. S. Palma, R. H. Manzo, D. Allemandi, L. Fratoni and P. L. Nostro, Solubilization of hydrophobic drugs in octanoyl-6-O-ascorbic acid micellar dispersions, J. Pharmaceut. Sci., 2002, 91(8), 1810–1816 CrossRef CAS PubMed.
  231. H. Onoda, Y. Inoue, T. Ezawa, I. Murata, T. Chantadee, S. Limmatvapirat, T. Oguchi and I. Kanamoto, Preparation and characterization of triamterene complex with ascorbic acid derivatives, Drug Dev. Ind. Pharm., 2020, 46(12), 2032–2040 CrossRef CAS PubMed.
  232. S. Palma, P. L. Nostro, R. Manzo and D. Allemandi, Evaluation of the surfactant properties of ascorbyl palmitate sodium salt, Eur. J. Pharmaceut. Sci., 2002, 16(1–2), 37–43 CrossRef CAS PubMed.
  233. A. R. Bilia, M. C. Bergonzi, F. F. Vincieri, P. L. Nostro and G. A. Morris, A diffusion-ordered NMR spectroscopy study of the solubilization of artemisinin by octanoyl-6-O-ascorbic acid micelles, J. Pharmaceut. Sci., 2002, 91(10), 2265–2270 CrossRef CAS PubMed.
  234. Y. Inoue, M. Hibino, I. Murata and I. Kanamoto, A nanocarrier skin-targeted drug delivery system using an ascorbic acid derivative, Pharm. Res., 2018, 35(1), 1 CrossRef CAS PubMed.
  235. F. Giudice, E. E. Ambroggio, M. Mottola and M. L. Fanani, The amphiphilic alkyl ester derivatives of L-ascorbic acid induce reorganization of phospholipid vesicles, Biochim. Biophys. Acta Biomembr., 2016, 1858(9), 2132–2139 CrossRef CAS PubMed.
  236. N. Khansari, Y. Shakiba and M. Mahmoudi, Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer, Recent Pat. Inflamm. Allergy Drug Discov., 2009, 3(1), 73–80 CrossRef CAS PubMed.
  237. R. R. Sawant, O. S. Vaze, T. Wang, G. G. M. D'Souza, K. Rockwell, K. Gada, B.-A. Khaw and V. P. Torchilin, Palmitoyl ascorbate liposomes and free ascorbic acid: comparison of anticancer therapeutic effects upon parenteral administration, Pharm. Res., 2012, 29(2), 375–383 CrossRef CAS.
  238. A. Llamedo, P. Rodríguez, C. de Passos, S. Freitas-Rodriguez, A. M. Coto, R. G. Soengas and R. Alonso-Bartolomé, Liposomal formulation of a vitamin C derivative: a promising strategy to increase skin permeability, J. Liposome Res., 2025, 1–9 Search PubMed.
  239. W. Johnson, I. J. Boyer, W. F. Bergfeld, D. V. Belsito, R. A. Hill, C. D. Klaassen and B Heldreth, Safety assessment of ethers and esters of ascorbic acid as used in cosmetics, Int. J. Toxicol., 2022, 41, 57S–75S CrossRef PubMed.
  240. Mad Hippie, Mad Hippie Vitamin C Serum, 2025, https://madhippie.com/products/vitamin-c-serum Search PubMed.
  241. Signal S, Comparison between TruSkin Vitamin C Facial Serum vs. Mad Hippie Vitamin C Serum, https://skinsignal.com/compare/truskin-vitamin-c-facial-serum-vs-mad-hippie-vitamin-c-serum.
  242. Riley S. C. E. O., 15% Vitamin C Brightening Serum, https://sundayriley.com/products/ceo-vitamin-c-brightening-serum.
  243. Care P. c. s., Potent-C Power Serum, https://www.peterthomasroth.com/products/potent-c-power-serum-1501049.
  244. Byrdie, Ole Henriksen's Banana Bright Serum Gave Me Younger-Looking Skin in a Week, https://www.byrdie.com/ole-henriksen-banana-bright-vitamin-c-serum-review-5409237.
  245. Skin A. O., Allies of Skin 20% Vitamin C Brighten + Firm Serum. https://uk.allies.shop/products/20-vitamin-c-brighten-firm-serum.
  246. Ordinary T., Ascorbyl Glucoside Solution 12%, https://theordinary.com/en-us/ascorbyl-glucoside-solution-12-vitamin-c-100405.html#:%7E:text=AscorbylGlucosideSolution12%25offersalightweight%2Cwater-based,agingbybrighteningandbalancingunevenskintone.
  247. Inkey, Inkey List 15% Vitamin C and EGF Serum, https://uk.theinkeylist.com/products/15-vitamin-c-egf-serum.
  248. A. Tai, Y. Fujinami, K. Matsumoto, D. Kawasaki and I. Yamamoto, Bioavailability of a Series of Novel Acylated Ascorbic Acid Derivatives, 6-O-Acyl-2-O-α-D-glucopyranosyl-L-ascorbic Acids, as an Ascorbic Acid…, Biosc. Biotech. Biochem., 2002, 66(8), 1628–1634 CrossRef CAS PubMed.
  249. M. Gosenca, M. Bešter-Rogač and M. Gašperlin, Lecithin based lamellar liquid crystals as a physiologically acceptable dermal delivery system for ascorbyl palmitate, Eur. J. Pharmaceut. Sci., 2013, 50(1), 114–122 CrossRef CAS.

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.