Feng Guan
*a,
Qi Wang
*b,
Yongping Bao
b and
Yimin Chao
c
aSchool of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, P. R. China. E-mail: guanfeng@hljucm.net
bNorwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK. E-mail: q.wang1@uea.ac.uk; y.bao@uea.ac.uk
cSchool of Chemistry, University of East Anglia, Norwich NR4 7TJ, UK. E-mail: y.chao@uea.ac.uk
First published on 11th February 2021
Rheumatoid arthritis (RA) is a common worldwide chronic autoimmune disease, characterised by synovial hyperplasia, inflammatory cell infiltration, pannus formation and destruction of articular cartilage and bone matrix. It is one of the most common forms of osteoarthritis bestowing high rates of both disability and death. Increasing attention has been paid to the use of natural medicines and natural products in the treatment of RA and patients' acceptance has increased year by year because of their high efficacy and safety. Flavonoids are a group of important secondary metabolites occurring in many plants which have rich biological activities such as anti-rheumatic, vasodilator, and anti-tumor effects. Many successful medical treatments of RA appear to be attributable to the application of flavonoids. Quercetin, a representative active member of the flavonoid family, is found abundantly in many plants, e.g. apples, berries, cabbages, onions, and ginkgo. In recent years, progress has been made in the research of its anti-rheumatoid effects which indicate that it is potentially a noteworthy prodrug for the treatment of RA. However, the poor solubility of quercetin affects its bioavailability and clinical efficacy. This review aims to provide an up to date summary of the biological effects and mechanism of action of quercetin for the treatment of RA, and the research progress made towards nano formulations of quercetin to improve its solubility and efficacy.
However, long-term administration of these drugs may cause gastrointestinal discomfort, nausea, vomiting, and bleeding, or other adverse reactions such as to the central nervous system or cardiovascular system. Moreover, improper application of hormones may even aggravate the disease. When the disease is difficult to control, biological agents such as tumour necrosis factor inhibitors, abatacept, rituximab, and tocilizumab are often used in clinic. However their use is limited by high cost and adverse events i.e. reactions and infections at infusion and injection sites.4 Increasing attention has been paid to the use of natural medicines or natural products in the treatment of RA, and the patients' acceptance has increased year by year because of their high efficacy and safety.
The flavonoids are vital secondary metabolites of many plants with the basic structural skeleton of 2-phenyl chromogenic ketone and consist of C6–C3–C6. It is a polyphenolic compound comprising two aromatic rings (A and B) and a heterocyclic ring (C). There are some flavonoid compounds that have a three-carbon chain but without a ring C. Some flavonoids exist in the form of dimers, trimers and even thioflavones. The main classes of natural flavonoids are flavones, isoflavones, flavanols, dihydroflavones, dihydroflavanols, bioflavonoids, triflavonoids, thioflavones, etc. (Fig. 1). Flavonoids have a number of biological activities (Fig. 2) including anti-inflammatory, analgesic, anti-rheumatic, vasodilator, anti-aging, and anti-tumour effects.10,11 The success of many medical treatments can be attributed to the application of flavonoids and new scientific studies of these compounds and their derivatives have focused on the activities above.12,13
Quercetin (5,7,3′,4′-tetrahydroxyflavonol, C15H10O7) is a representative member of the flavonoid's family. It is found abundantly in a variety of foods including apples, berries, Brassica vegetables, grapes, onions, shallots, as well as many medicinal plants including Ginkgo biloba, Hypericum perforatum, and Sambucus canadensis.14–16 Quercetin frequently occurs as quercetin glycosides where polyhydroxyl substitution appears in its structure.17 The quercetin glycosides derivatives that have been identified include quercetin-3-O-rhamnoside (quercitrin), quercetin-3-O-glycoside (isoquercitrin), quercetin-3-O-rutinoside (rutin), and quercetin-7-O-glycoside (quercimeritrin) (Fig. 3).18–20 Quercetin possesses the typical pharmacological effects of flavonoids, such as anti-inflammatory, analgesic, anti-rheumatic, antioxidant, anti-tumour, etc.12,21–24. In recent years, new progress has been made in the research of its anti-rheumatoid effects which indicate that it is safe to use, with few side effects and thus a noteworthy prodrug for the treatment of RA.
Despite showing promising potential for medicinal use, the real-life application of quercetin has been largely limited due to its poor solubility and bioavailability.13,18 The solubility of quercetin in water is 7 μg mL−1, and it is absorbed and metabolised rapidly after entering the body. Quercetin has strong first pass effect and its bioavailability is very low, less than 3.6%. Quercetin is inactivated by combining with sugar molecules into gluconic acid and so on. Therefore, there is a need to employ modern nanotechnology to improve its solubility and bioavailability, so as to better deploy its anti-rheumatoid effects. This review is intended to provide an insight into the pharmacological function and mechanisms of action of therapeutic use of quercetin for RA. The recent research progress using nano formulation as a strategy to increase/improve quercetin potential in RA treatment have also been summarised. Our review is mainly based on the published journal papers in recent 15 years, excluding patent literature.
Liao and Lin studied the pharmacological effects of quercetin on systemic inflammation in septic mice.27 The sepsis mouse model was established by intraperitoneally (i.p.) injecting lipopolysaccharide (LPS). LPS is an important trigger of inflammatory response, which can stimulate a variety of cells in vivo, especially macrophages to synthesize and release many endogenous bioactive factors, leading to inflammatory response. They then administered quercetin to the septic mice in a prophylactic or therapeutic manner. Their results suggested that quercetin administration i.p. at a high dose of 0.15 μmol to each mouse could significantly (P < 0.05) increase Interleukin 10 (IL-10) secretion and had strong anti-inflammatory potential. Huang et al. similarly observed the anti-inflammatory effect of quercetin.28 Their results showed that quercetin at 100, 200, and 400 mg kg−1 could significantly inhibit the auricle swelling of rats caused by xylol, and the degree of swelling and inhibition rate were significantly different from those in the control group (P < 0.01). This suggests that quercetin can have a good inhibitory effect on inflammatory response.
RAW264.7 is a monocyte/macrophage-like cell line that has been frequently used to study immune function. Zhou et al. proved that quercetin significantly inhibited the increase of nitric oxide (NO), Tumour Necrosis Factor alpha (TNF-α), Interleukin 18 (IL-18) and Interleukin 6 (IL-6) in Raw264.7 cells induced by LPS (P < 0.01) showing that it has a good anti-inflammatory effect in vitro.29 These results agreed with the findings from Paul et al.30 and Cessak et al.31 which suggested that quercetin is an effective inhibitor for TNF-α and IL-6. Ren et al. also studied the protective effect of quercetin on LPS induced inflammation in RAW264.7 cells.32 Their results suggested that the protective effect may be related to the regulation of Toll-like receptor 4/nuclear factor kappa-light-chain-enhancer of activated B (TLR4/NF-κB) signalling pathway.
Chronic inflammation, a process linked to increased oxidative stress, may induce many diseases. Yeh et al. investigated the effects of β-carotene on the inflammatory reaction of macrophage model cells (differentiated HL-60 cells and RAW264.7 cells) and their modulation by quercetin or naringenin.33 Their results demonstrated that quercetin partially suppressed the pro-inflammatory effects, synergistically enhanced the inhibitory effects of β-carotene on the secretion of pro-inflammatory mediators and the DNA damaging ability of PMA-stimulated HL-60 cells. The mechanism of action was associated with its antioxidant activity and inhibition of the production of pro-inflammatory cytokines. Avila et al. summarised that quercetin showed a mixed inhibition mechanism towards Adenosine triphosphate (ATP) and that the binding site of quercetin overlaps with both ATP and inhibitor of nuclear factor kappa B (IκBα) binding sites.34
Extracellular High-Mobility Group Box-1 (HMGB-1) is an important late-stage inflammatory transmitter with strong inflammatory activity. It contributes to the pathogenesis of numerous chronic inflammatory and autoimmune diseases including RA. Musumeci et al. indicated that quercetin was a HMGB1 inhibitor and it could limit the activation of mitogen-activated protein kinase.35
Liu DN has reported that quercetin had no significant effect on paw withdrawal thermal latency in naïve rats. However, it could significantly increase the threshold of mechanical paw contraction response.36 The study also showed that quercetin has a significant inhibitory effect on bee venom induced spontaneous nociceptive response, pain score, thermal and mechanical pain sensitivity, and also on bee venom induced local inflammatory response.36 This suggests that quercetin's analgesic effect may be related to the blocking of pro-inflammatory factors. In addition, quercetin has a considerable inhibitory effect on the ipsilateral mechanical hyperalgesia and contralateral mechanical hyperalgesia caused by sciatic nerve branch injury model.
In addition to the above studies on the anti-inflammatory and analgesic effects of quercetin monomer, there are also a number related studies of medicinal plant extracts with quercetin as the main component, which further confirm the anti-inflammatory and analgesic effects of quercetin.37–47
Mamani-Matsuda et al. observed that the therapeutic and preventive properties of quercetin in experimental arthritis correlated with decreased macrophage inflammatory mediators.53 Their results indicated that in chronic rat (AA), oral administration of quercetin (30 mg per rat every 2 days, for 10 days) to arthritic rats resulted in a clear decrease of clinical signs compared to untreated controls. The effects of oral administration of quercetin (150 mg kg−1 daily, 28 days) were also investigated in a rat model of adjuvant arthritis by Gardi et al.54. Their results indicated that quercetin lowered levels of Interleukin 1β (IL-1β) (p < 0.003), monocyte chemotactic protein-1 (MCP-1) (p < 0.014) and restored plasma antioxidant capacity.
Effects | Model inducer | Animal | AM | Dose | SW | PT | WT | PWTL | PWMT | TNF-α | IL-1β | IL-1α | IL-6 | IL-8 | IL-10 | IL-17 | NO | CRP | MCP-1 | PGE2 | MMP-1 | MMP-3 | MMP-13 | Ref. |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
a Note: ① AM(administration mode), SW (swelling), PT (pain threshold), WT (writhing times), PWTL (paw withdraw thermal latency), PWMT (paw withdrawal mechanical threshold), IFA (incomplete Freund's adjuvant), CRP(C-reactive protein), MCP-1 (monocyte chemotactic protein-1), PMA (phorbol-12-myristate-13-acetate), Ref. (References). ② — not mentioned, ↑ increase, ↓ reduce. | ||||||||||||||||||||||||
Anti-inflammatory effects | LPS | BALB/c mice | i.p. | 0.15 μmol (each mouse) | — | — | — | — | — | ↑ | ↓ | — | ↑ | — | ↑ | — | — | — | — | — | — | — | — | 27 |
Anti-inflammatory effects | Sodium urate | SD rats | i.g. | 100, 200, 400 mg kg−1 | ↓ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 28 |
Anti-inflammatory effects | Xylene | BALB/c mice | i.g. | 100, 200, 400 mg kg−1 | ↓ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 28 |
Anti-inflammatory effects | LPS | In vitro | — | 2.5, 5, 10 μg mL−1 | — | — | — | — | — | ↓ | ↓ | — | ↓ | — | ↓ | — | ↓ | — | — | — | — | — | — | 29 |
Anti-inflammatory effects | LPS | In vitro | — | 5, 15, 25 μM | — | — | — | — | — | ↓ | ↓ | — | ↓ | — | — | — | ↓ | — | — | — | — | — | — | 32 |
Anti-inflammatory effects | PMA | In vitro | — | 20 μM | — | — | — | — | — | ↓ | — | — | — | ↓ | — | — | — | — | — | — | — | — | — | 33 |
Analgesic effects | Hot plate | BALB/c mice | i.g. | 100, 200, 400 mg kg−1 | — | ↑ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 28 |
Analgesic effects | Acetic acid | BALB/c mice | i.g. | 100, 200, 400 mg kg−1 | — | — | ↓ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 28 |
Analgesic effects | Bee venom | SD rats | i.g. | 40, 80, 120 mg kg−1 | ↓ | — | — | ↑ | ↑ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 37 |
Anti-rheumatic effect | Adjuvant-carrageenan | Wistar rats | i.g. | 80 mg kg−1 | ↓ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 52 |
Anti-rheumatic effect | LPS | — | — | — | — | — | — | — | — | ↓ | — | — | ↓ | — | — | — | — | — | — | — | — | — | — | 30 |
Anti-rheumatic effect | AA | Lewis rats | i.g. | 150 mg kg−1 | ↓ | — | — | — | — | — | ↓ | — | — | — | — | — | — | ↓ | ↓ | — | — | — | — | 54 |
Anti-rheumatic effect | CIA | C57BL/6 mice | i.g. | 30 mg kg−1 | ↓ | — | — | — | — | ↓ | ↓ | ↓ | ↓ | — | — | ↓ | — | — | ↓ | — | — | — | — | 55 |
Anti-rheumatic effect | CIA | Wistar rats | i.g. | 150 mg kg−1 | ↓ | — | — | — | — | ↓ | ↓ | — | ↓ | — | — | ↓ | — | — | — | ↓ | — | — | — | 56 |
Anti-rheumatic effect | CIA | Wistar rats | i.p. | 50 mg kg−1 | ↓ | — | — | — | — | — | ↓ | — | — | — | — | — | — | — | — | — | — | — | ↑ | 57 |
Anti-rheumatic effect | CIA | DBA/1 mice | i.g. | 50 mg kg−1 | ↓ | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | — | 58 |
Anti-rheumatic effect | TNF-α | In vitro | — | 50, 100 μM | — | — | — | — | — | — | ↓ | — | ↓ | ↓ | — | — | — | — | — | — | ↓ | ↓ | ↓ | 59 |
Anti-rheumatic effect | Zymosan | Swiss mice | 100 mg kg−1 | ↓ | ↓ | — | — | — | ↓ | ↓ | — | — | — | — | — | — | — | — | — | — | — | — | 108 |
In the studies of Khor et al., quercetin was co-precipitated with dietary fibres into a fast-dissolving nano formulation via antisolvent precipitation. It was found that a high dissolution rate and good storage stability was achieved for quercetin nano formulations with cellulose fibre, resistant starch, or resistant maltodextrin. The nano formulations exhibited higher levels of antioxidant activities in contrast to quercetin alone.92
García-Casas et al. reported that a supercritical antisolvent (SAS) process had been used to precipitate microparticles of quercetin together with nanoparticles of cellulose acetate phthalate (CAP). Release profiles of quercetin were carried out in simulated gastric and intestinal fluids. Higher ratios of quercetin to polymer in the coprecipitates were recommended to achieve faster release and higher solubilities of quercetin.109
Wang et al. reported that amphiphilic chitosan was obtained through grafting of deoxycholic acid modified chitosan and N-acetyl-L-cysteine. Quercetin-loaded nanomicelles (CS-DA-NAC-QNMs) were prepared through a self-assembly method by using amphiphilic chitosan as the wall-material and quercetin as core-material. They demonstrated that there was a bursting release of quercetin from CS-DA-NAC-QNMs for 0 to 8 hours, and then the release rate decreased gradually. After 72 hours, more than 40% of quercetin were released. All the quercetin loaded nano micelles samples showed good hemocompatibility, and their water solubility and biocompatibility was increased significantly.110
Chavoshpour-Natanzi et al. prepared β-Lactoglobulin (BLG) nanoparticles for the encapsulation of quercetin. The nanoparticles had a mean particle size of between 180–300 nm and a loading efficiency (LE) of 13.9%. Protein nanoparticles could be digested at different stages of the gastrointestinal tract, depending on several factors including specificity of proteases e.g. pepsin. This study suggested that nano formulation could overcome BLG resistance to peptic digestion. Thus synthesised BLG-quercetin nanoparticles could achieve controlled release of quercetin under simulated conditions.93
The study performed by Simon et al. used harmless amphiphilic polyoxazolines (POx) to encapsulate quercetin.96 They produced mixed micelles, made of POx and phosphatidylcholine, using a thin film and high-pressure homogeniser process. The obtained nanomicelles that were about 20 nm in diameter with a spherical shape and encapsulation efficiency of 94 ± 4%. They demonstrated improved cell viability and antioxidant activity from these nanomicelles compared to quercetin alone. Subsequently, this group synthesised quercetin encapsulated lipid nanocapsules (LNC) using the same material POx.111 A similar synthesis method has been used as for the production of mixed micelles but implementing an additional short sonication step. The obtained LNC have a well-defined spherical shape and a size of ∼30 nm.
Aditya et al. reported a comparative study of solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), and lipid nano emulsions (LNE) of quercetin.112 Encapsulation efficiency (EE) of quercetin in these nanocarriers was above 90%. Maximum bio-accessibility was observed with NLC and LNE (W60%) compared to SLN (W35%) and free quercetin solution (W7%).
Tran et al. developed a quercetin-containing self-nanoemulsifying drug delivery system (Q-SNEDDS). Oil-in-water nanoemulsions were formed to improve quercetin oral bioavailability.98 Following oral administration of Q-SNEDDS in rats (15 mg kg−1), the maximum concentration (Cmax) of plasma quercetin after 24 h was 3.75 ± 0.96 mg L−1, increased by approximately three-fold compared with the native quercetin group (1.20 ± 0.17 mg L−1). The results suggested that Q-SNEDDS can enhance the solubility and oral bioavailability of quercetin. Collectively, Q-SNEDDS increased quercetin Cmax and area under the concentration curve (AUC), from 6.7 ± 1.4 L−1 h−1 to 14.0 ± 2.8 L−1 h−1, without affecting its elimination kinetics, suggesting that Q-SNEDDS improved quercetin bioavailability by enhancing its absorption.
Dinesh Kumar et al. have also studied biodegradable polymeric nanoparticles for the effective delivery of quercetin. The results suggest that optimised formulation of nanoparticles could promote the controlled release and improve the physical stability of quercetin.113
Lee et al. investigated the antioxidative and anti-inflammatory activities of quercetin-loaded silica nanoparticles (QLSNs).94 QLSNs were synthesised using an oil-in-water microemulsion method. The nanoparticles showed comparable cell viability to that of the free quercetin, while the amounts of proinflammatory cytokines produced by macrophages, such as TNF-κB, IL-6, and IL-1β, were significantly reduced.
Caddeo et al. prepared cross-linked chitosan liposomes of quercetin and confirmed that the system had acid resistance and promoted the release under alkaline conditions.114 In addition, Hao et al. proposed a facile electrostatic deposition method to prepare quercetin nanoliposomes coated with chitosan.115 The obtained Q-NPs have high EE (71.14%) and the storage stability and antioxidant activity was improved compared with native quercetin.
Penalva et al. studied the use of zein nanoparticles as a carrier for the oral delivery of quercetin. Quercetin and 2-hydroxypropyl-β-cyclodextrin were encapsulated together in zein nanoparticles. They showed that nanoparticles provided high and sustained levels of quercetin in plasma after oral administration. The Cmax of plasma quercetin was 176 ± 13.4 μg mL−1. The mean values obtained for AUC and the half-life of the terminal phase (t1/2) were 167 ± 8.21 μg h mL−1 and 0.60 ± 0.35 h, respectively. The mean residence time (MRT) was 1.60 ± 0.12 h, whereas the quercetin clearance and its volume of distribution were calculated to be 30 mL h−1 and 26 mL h−1, respectively. The relative oral bioavailability was calculated to be about 60%.116 They further optimised the preparative process of quercetin loaded casein nanoparticles and evaluated the pharmacokinetics of the nanoparticles after oral administration to Wistar rats117 showing that the relative oral bioavailability of quercetin in nanoparticles (close to 37%) was about 9-times higher than the oral solution of quercetin in a mixture of PEG 400 and water. Another study by Li et al. also used zein and soluble soybean polysaccharide (SSPS) nanoparticles. The EE of quercetin was greatly improved to 82.5% and the photochemical stability and 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS+) scavenging ability of quercetin in such nanoparticles were significantly enhanced.100
Pivetta et al. produced nanostructured lipid carriers to load quercetin. Their results indicated that the nanoparticles exhibited a low recrystallization index (13.03%) which is important to obtain high entrapment efficiency (97.42%) and avoid drug expulsion during the storage time.118 Furthermore, in a reconstructed human skin model, it was observed that the topical formulation of quercetin-NLC presented no phototoxic potential. Therefore, this developed nanostructure is a vehicle with potential for topical administration of quercetin.
Research by Gokhale et al. reported a quercetin loaded nano emulsion (NE)-based gel for the effective of management RA.119 This study showed that quercetin-NE has no toxic effect on synoviocytes and a strong inhibitory effect on LPS-induced TNF-α production. It has also exhibited adequate rheological behaviour with a good texture profile and improved drug permeation compared to a free quercetin gel. In addition, the gel was found to be non-irritating and inhibited the formation of paw edema in rats induced by Freund's complete adjuvant (CFA) over 24 hours. Another study performed by Ghatak and Iyyaswami used casein particles to encapsulate quercetin to improve its water solubility and bioavailability.101 A maximum encapsulation yield of 97% could be achieved with the addition of 0.5% (w/v) sodium caseinate, 0.1 M of calcium chloride, 0.5 M of di potassium hydrogen phosphate, 0.1 mM CTAB and 1 M of sodium citrate at a pH of 7.
Mohan et al. reported on TiO2 nanotubes that were loaded initially with quercetin (TNTQ) and then additionally with chitosan coated on the top of the quercetin (TNTQC) to various thicknesses. The drug release of TNTQ and TNTQC were studied in Hanks' solution for 192 hours. The results showed that the release of drug into the local environment during that duration was constant and the local concentration of the drug could be controlled and tuned by controlling the thickness of the chitosan (0.6, 1 and 3 μm).120
Zong et al. studied in vitro release of quercetin-loaded mixed micelles composed of Pluronic P123/Poloxamer 188, and their pharmacokinetics in rat.121 The results indicated that quercetin-loaded mixed micelles have high entrapment efficiency and loading efficiency which could improve the release behaviour in vitro. The nano formulation of quercetin also prolonged the circulation time of quercetin and significantly enhanced the bioavailability of quercetin. Hui et al. prepared and characterised amphiphilic chitosan/quercetin nano micelles (ACS-QNMs) using a novel amphiphilic chitosan (ACS).97 ACS has deoxycholic acid (DA) as the hydrophobic group and n-acetyl-L-cysteine (NAC) as the hydrophilic group. The results showed that quercetin could be released in vivo and was stable when stored at room temperature after being embedded in nano micelles.
Zhao et al. prepared a new nanodrug delivery system (quercetin@mesoporous hydroxyapatite, QUE@MHAs) and investigated its release in vitro.122 The results showed that QUE@MHAs have good stability and a slow drug release rate.
Tan Qi studied the preparation and evaluation of quercetin-loaded lecithin–chitosan nanoparticles for topical delivery. Quercetin nanoparticles were prepared using organic solvent injection. The results demonstrated that the nanoparticles could clearly increase the amount of drug retention in especially in the epidermis and also in the dermis, and further enhance antioxidation and anti-inflammatory effects.123
Guo et al. evaluated the potential of quercetin-loaded nanostructured lipid carriers (QT-NLCs) as a topical delivery system.124 The nanoparticles were prepared by the method of emulsion evaporation–solidification at low temperature. The results showed that QT-NLCs could promote the permeation of quercetin, increase the amount of quercetin retention in epidermis and dermis, and enhance the effect of anti-oxidation and anti-inflammation exerted by quercetin.
Sapino et al. evaluated the potential of aminopropyl functionalised mesoporous silica nanoparticles (NH2-MSN) as topical carrier system for quercetin delivery. The silica nanoparticle vehicle prevented UV-induced degradation of quercetin over time, which showed positive effect on photostability of quercetin. Epidermal accumulation and transdermal permeation were evaluated ex vivo using porcine skin mounted on Franz diffusion cells. The inclusion complexation with the inorganic nanoparticles increased the penetration of quercetin into the skin after 24 hours post-application without transdermal delivery.95
Hatahet et al. tested three approaches to improve quercetin delivery to skin, including liposomes, lipid nanocapsules (LNC) and smartCrystals®.99 They showed that compared to liposome (0.56 mg mL−1), quercetin smartCrystals® and LNC had a better drug loading with 14.4 mg mL−1 and 10.8 mg mL−1 respectively. SmartCrystals® and LNC demonstrated different skin penetration behaviours. Only LNC allow quercetin to be delivered to viable epidermis that holds potential for treatment of skin inflammatory disorders.
In conclusion, quercetin is not only an important drug source for oral administration but can also be used as a transdermal absorbent by employing nanotechnology to enhance its transdermal absorption capacity. It can be seen from all above reports that different materials and forms of nano encapsulation can have many positive effects on quercetin anti-rheumatoid applications. Nano formulation has significantly improved the solubility and bioavailability of native quercetin, and at the same time avoiding its shortcomings. The anti-rheumatoid related nano formulations of quercetin are summarised in Table 2.
Type | Nanocarrier | Preparation method | Antisolvent | Size (nm) | Aim | Ref. |
---|---|---|---|---|---|---|
a Note: — not mentioned; Ref. (reference). | ||||||
Nanoparticles | — | Syringe pump | Deionised water | 170 | Improvement of solubility | 107 |
Nanoparticles | Dietary fiber | Antisolvent precipitation | Water | <100 | Improvement of solubility | 92 |
Nanoparticles | Cellulose acetate phthalate | Supercritical antisolvent | Supercritical CO2 | 145 | Improvement of solubility | 109 |
Nanoparticles | β-Lactoglobulin | Antisolvent precipitation | Acetone | 180–300 | Improvement of solubility | 93 |
Nanoparticles | Thioglycolic acid-capped cadmium telluride quantum dots | Antisolvent precipitation | Acetone | 185 | Enhancement of bioavailability | 102 |
Nanoparticles | Polycaprolactone | Nano-precipitation | Pluronic F127 | 213–257 | Enhancement of bioavailability | 113 |
Nanoparticles | Silica | Oil-in-water microemulsion | Water | 70–140 | Enhancement of bioavailability | 94 |
Nanoparticles | Zein, 2-hydroxypropyl-β-cyclodextrin | Desolvation | Water | 300 | Enhancement of bioavailability | 116 |
Nanoparticles | Casein, 2-hydroxypropyl-β-cyclodextrin | Simple coacervation | Water | 200 | Enhancement of bioavailability | 117 |
Nanoparticles | Natural lipids | Emulsion and sonication | Pluronic F68 | 130 | Enhancement of bioavailability | 118 |
Nanoparticles | Zein, SSPS | Antisolvent precipitation | — | 200 | Enhancement of bioavailability | 100 |
Nanoparticles | Casein | Emulsion | Ethanol | 114.3–482.1 | Enhancement of bioavailability | 101 |
Nanoparticles | Mesoporous hydroxyapatite | Magnetic stirring | Deionised water | 169–179 | Regulation of release rate | 122 |
Nanoparticles | Lecithin, chitosan | Organic solvent injection | Ethanol | 95 | Changes of administration mode | 123 |
Nanoparticles | Soya lecithin, glyceryl monostearate, stearic acid, media chain triglyceride | Emulsion evaporation–solidification | Water | 215 | Changes of administration mode | 124 |
Nanoparticles | Mesoporous silica | Impregnation and magnetic stirring | Methanol | 200–300 | Changes of administration mode | 95 |
Nanomicelles | Amphiphilic chitosan | Grafting deoxycholic acid, N-acetyl-L-cysteine | — | 360–580 | Improvement of solubility | 110 |
Nanomicelles | Polyoxazolines, phosphatidylcholine | Thin film and high pressure homogeniser | Acetonitrile | 20 | Improvement of solubility | 96 |
Nanomicelles | Polyoxazolines, Labrafac®, Lipoid® S75 | Thin film and high pressure homogeniser process | Acetonitrile | 30 | Improvement of solubility | 111 |
Nanomicelles | Pluronic P123/Poloxamer 188 | Film dispersion | Tween80 | — | Regulation of release rate | 121 |
Nanomicelles | Amphiphilic chitosan | Self assembly | Deionised water | 140–600 | Regulation of release rate | 97 |
Nanoliposomes | Cross-linked chitosan | Ultrasonication | TPP aqueous solution | 180 | Enhancement of bioavailability | 114 |
Nanoliposomes | Chitosan | Facile electrostatic deposition | Chloroform, methanol | 350–600 | Enhancement of bioavailability | 115 |
Nanoliposomes | GMS, MCT, soy lecithin | Emulsifying and solidifying | Transcutol | 118–135 | Regulation of release rate | 78 |
Nanoliposomes | DPPC, Cremophor® EL | Magnetic stirring | Alcohol | 179 | Changes of administration mode | 99 |
Nanoemulsions | Lecithin | High pressure homogeniser | — | 73–91 | Enhancement of bioavailability | 112 |
Nanoemulsions | Self-nanoemulsifying drug delivery system | Gentle stirring | Tween 80, PEG 400 | 204–213 | Enhancement of bioavailability | 98 |
Nanoemulsion-based gels | Arachis oil, oleic acid | Gentle stirring | Tween 20, PEG 400 | 137 | Enhancement of bioavailability | 119 |
Nanosuspensions | — | Evaporative precipitation | Hexane | 220 | Improvement of solubility | 106 |
Nanotubes | TiO2 | Top filling | — | 125 (tubes diameters) | Regulation of release rate | 120 |
Nanocapsules | Lipid | Magnetic stirring | Milli Q water | 26 | Changes of administration mode | 99 |
As a plant derived medicine, quercetin is a striking candidate for use in arthritic therapy.126 As summarised above, there are many reports on the effects of quercetin on RA. It has been confirmed that quercetin has significant anti-inflammatory and analgesic effects in vivo and in vitro and furthermore, quercetin and its derivatives also have significant antioxidant effects, which is one of the possible reasons for their significant anti-rheumatic properties.20 At the same time, it is reported that quercetin is mostly well tolerated and safe to use. Doses up to 1000 mg each day for several months have not produced adverse effects on blood parameters, and liver, and kidney function. As a potential bioavailability enhancer for active pharmaceutical ingredients, quercetin can also be used as one of the options in combination therapy for RA.65 Moreover, it has been shown that with the application of nano formulations, quercetin has not only improved oral bioavailability, but also can be used for external transdermal use, which provides a new reference for the treatment of RA.
Move rover, Susanne Andres et al. reviewed the safety aspects of quercetin as a dietary supplement. It showed that based on animal studies, some possible critical safety aspects of quercetin could be identified such as to enhance nephrotoxic effects in the predamaged kidney or to promote tumour development especially in estrogen-dependent cancer.127 Furthermore, when quercetin interacts with some drugs, the bioavailability of may be altered. Therefore, it suggests that, like any potential drug or active ingredient, a very in-depth study on its safety and applicability should be conducted before clinical application. Future clinical studies are needed to verify the safety and efficacy of nano formulated quercetin as a new RA treatment medicine. Future clinical studies are needed to verify the safety and efficacy of Nano formulated quercetin as a new RA treatment medicine.
This journal is © The Royal Society of Chemistry 2021 |