You are on page 1of 15

Journal of Drug Delivery Science and Technology 60 (2020) 102082

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


journal homepage: www.elsevier.com/locate/jddst

Review article

Recent developments in formulation design for improving oral


bioavailability of curcumin: A review
Zhenqi Liu, John D. Smart, Ananth S. Pannala *
Biomaterials and Drug Delivery Research Group, School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK

A R T I C L E I N F O A B S T R A C T

Keywords: Curcumin, a yellow-orange substance that is extracted from the spice turmeric (Curcuma longa, Zinziberaceae),
Curcumin has been attributed with a wide range of pharmacological activities for the prevention and treatment of several
Oral bioavailability disease conditions such as arthritis, hypertension, diabetes, Alzheimer’s, antibacterial and cancer to name a few.
Delivery system
However, its potential for use as an orally delivered medicinal product is hindered by its poor solubility and
Liposomes
bioavailability. The low oral bioavailability of curcumin is caused by several factors including low aqueous
Clinical studies
Soluplus® solubility, poor intestinal permeability, unstable at alkaline pH and rapid metabolism. To improve curcumin’s
Piperine poor oral bioavailability, different formulation strategies such as incorporation into nanoparticles, liposomes,
micelles, micro/nano-emulsions and solid dispersions as well as co-administration with piperine have been
investigated in both animal models as well as human supplementation studies. In this review, novel formulations
of curcumin for oral delivery that were developed in recent years are reviewed and discussed.

1. Introduction wounds [10], while in China turmeric is regularly used as herbal


medication for treating diseases that are associated with abdominal pain
Curcumin is a yellow orange coloured crystalline compound that is [11]. With the advancement of science and technology, it had been
extracted from the spice turmeric (Curcuma longa, Zingiberaceae). discovered that curcumin is the key ingredient that contributes to most
Chemically, curcumin is known as diferuloylmethane (C21H20O6) with a of the therapeutic effects of turmeric [12,13]. Curcumin was found to
molecular mass of 368.37 g/mol. It is a polyphenol compound and has a have a wide spectrum of pharmacological activities. Several studies on
melting point of 183 ◦ C. The IUPAC name of curcumin is 1,7-bis (4- curcumin showed that it has anti-inflammatory [14], antimicrobial [15,
hydroxy-3-methoxy phenyl)-1,6-heptadiene-3,5-dione (1E-6E). The two 16,17,18], antirheumatic [19], immunomodulatory [20] and
aryl rings in curcumin contain ortho-methoxy phenolic groups are anti-tumour effects [21]. The anti-tumour effect of curcumin is associ­
symmetrically linked to a β-diketone moiety [1,2,3,4]. Curcumin ex­ ated with the suppression of early growth of response-1-gene product
hibits a pH-dependent keto–enol tautomerism, where in an acidic or (EGR-1), protein tyrosine kinase cascade and mitogen-activated protein
neutral solution, the keto form of curcumin is predominant while in an kinases (MAPK) pathway [22]. Curcumin also demonstrates antioxidant
alkaline medium the enol form of curcumin becomes predominant property based on its ability to scavenge free radicals due to the electron
(Fig. 1) [1,5,6]. The colour of curcumin also changes at different pH donating property of the phenolic group [23,24,25,26,27]. The
levels forming a bright yellow coloured solution at pH range 2.5–7.0 anti-inflammatory effect of curcumin is associated with its ability to
with the colour changing to dark red when the pH is increased above 7 inhibit NFκB (Nuclear factor κB) to bind with DNA. The inhibition of
[1,4]. NFκB-DNA binding lead to suppression of pro-inflammatory molecules
The use of turmeric for medical treatment in Asia can be traced back MMP-3 (matrix metalloprotease 3) and MMP-9 (matrix metalloprotease
thousands of years [2]. Turmeric has been widely used since ancient 9). It also results in reduction in pro-inflammatory cytokines, such as
times as a herbal medicine for treating many conditions such as TNFα (tumour necrosis factor 1α), IL1β (interleukin 1β) and IL8 (inter­
cough/inflammation, respiratory diseases, flu, sinusitis, liver disorders, leukin 8) [4]. In addition, curcumin also exert anti-inflammatory effects
rheumatism and abdominal pain [7,8,9]. In India, turmeric is tradi­ by binding to proteins COX-2 (prostaglandin-endoperoxide synthase 2),
tionally used for treating infections, sprains/swelling and healing burn which leads to reduction in COX-2 expression, prostaglandin and

* Corresponding author. School of Pharmacy and Biomolecular Sciences University of Brighton Lewes Road Brighton, BN2 4GJ, UK.
E-mail address: a.s.v.pannala@brighton.ac.uk (A.S. Pannala).

https://doi.org/10.1016/j.jddst.2020.102082
Received 17 July 2020; Received in revised form 2 September 2020; Accepted 9 September 2020
Available online 13 September 2020
1773-2247/© 2020 Elsevier B.V. All rights reserved.
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

suppression of amyloid β protein aggregates leads to the reduction of


reactive oxygen species (ROS) and cytochemokines, which decreases
oxidative stress induced damage and neuroinflammation to the brain
[38,40,39].
The safety of curcumin was proven in several animal tests and human
studies [41,42,43,44]. An oral dose as high as 12 g per day was shown to
be well tolerated in humans [42]. However, despite its wide pharma­
cological potentials and high dose tolerance, curcumin is still not
approved as a therapeutic agent for oral delivery. The main reason is
because curcumin has very poor oral bioavailability so it can be barely
absorbed in the human body [1]. Curcumin plasma concentration was
under the detection limit at an oral dose of 12 g in healthy male subjects
[45]. To overcome curcumin’s problem of bioavailability, numerous
research studies have been reported every year. In this review, factors
that account for the poor oral bioavailability of curcumin are first
explained in detail followed by a review of recent efforts in improving
curcumin’s oral bioavailability.

2. Routes of drug administration reported for curcumin

The delivery routes of curcumin include oral administration, intra­


venous (IV) injection, nasal administration, topical administration,
subcutaneous delivery and intraperitoneal (IP) administration [46].
Among the delivery routes, oral delivery is regarded as the most
accepted route for drug administration. It is also the most preferred
administration routes over other delivery routes due to its convenience,
Fig. 1. Keto-Enol tautomerism of curcumin. high patient compliance, cost effectiveness and ease of production [47].
Besides, oral administration is the most studied delivery route for cur­
thromboxane synthesis. Subsequently, curcumin inhibits the activity of cumin. In most of studies, curcumin was delivered to the subjects (either
5-LOX (Arachidonate 5-lipoxygenase) and leukotriene synthesis. The humans or animals) through oral administration [46]. Orally delivered
decreased expression of these compound attributes to the curcumin has exhibited various pharmacological effects such as
anti-inflammatory effects of curcumin [28]. The anti-bacterial effect of anti-inflammatory, anti-tumour, antioxidant, antimicrobial, antidia­
curcumin is believed to be associated with its ability to inhibit the for­ betic, immunomodulatory, etc.
mation of FtsZ ring (Z ring) at the site of division of bacterial cells, which In IV administration, curcumin is injected directly into blood vessels,
disrupts the division of the bacterial cells [29]. Curcumin also showed which helps curcumin bypass the physiological barriers against drug
anti-viral effect on several viruses such as HIV, influenza, HSV-1, cox­ absorption, leading to highest bioavailability and fastest effect among all
sackievirus and HBV. The anti-viral activities of curcumin include delivery routes. However, compared with the oral route, IV injection has
inhibiting the HIV-1 LTR-directed gene expression, HIV-1/HIV-2 pro­ some major drawbacks such as poor patience adherence, inconvenience,
teases, haemagglutination, reducing HSV-1, coxsackievirus and HBV needle-associated pain, risk of unsafe needle use and the need for trained
replication [30]. The pharmacological effect of curcumin on rheumatic healthcare personnel [48].
diseases like osteoarthritis is based on its ability to in inhibit the Topical delivery is administration of drugs directly on to the body
expression of metalloproteinase-3 enzymes (MMP3). MMP3 is highly surfaces such as skin or mucous membranes. Compared with oral
expressed in synovial cells in patients with osteoarthritis, which results administration, curcumin delivered though topical route showed phar­
in the degradation of connective tissue components. By inhibiting the macological effects related mostly to the skin conditions such as skin
expression of MMP3, curcumin inhibits the proliferation of osteoar­ inflammation, wound healing and skin cancer among others [49,50,51,
thritis synovial cells, increases the cell apoptosis, and eventually ease 52].
the inflammation and cartilage degradation of osteoarthritis [31]. Nasal delivery is a drug administration route where the drug is
Although the exact mechanisms are still unknown, the immunomodu­ insufflated through the nose and absorbed via the nasal mucosa. Cur­
latory effects of curcumin are most likely due to its ability to inhibit cumin was used in nasal administration for better brain targeting and it
NF-κB target genes that are involved in inducing immune responses showed pharmacological effects on neurodegenerative diseases such as
[32]. Additionally, activity in protecting the heart and kidney against Alzheimer’s and Parkinson’s diseases [46]. Nasal delivery is an easier
oxidative injury was reported in several studies [33,34–36]. Curcumin way for delivering curcumin when compared with IV injection. How­
shows heart protecting effect through several mechanisms that include ever, it is still relatively inconvenient when compared to oral delivery
inhibiting cardiomyocyte fibrosis, increasing ventricular hypertrophy route since there is a possibility of nasal irritation for some patients [53].
and related-gene expression [33]. Curcumin protects against renal Intraperitoneal injection is an administration route where the drug is
injury by inhibiting lipid peroxidation in kidneys, which reduces injected directly into the peritoneum (body cavity) and in subcutaneous
oxidative injury to the kidneys. It also increases levels of natural anti­ injection the drug is injected into the tissue layer between the skin and
oxidants, kidney glutathione content and glutathione peroxidase activ­ the muscle. These two delivery routes were reported more often and
ity in kidney tissues, which restores kidney functions [35]. Curcumin used in the administration of curcumin to animals than to humans [46].
also showed therapeutic activity in diabetes due to its effect in The routes of administration played important roles in the thera­
increasing sensitivity to insulin and reducing blood glucose levels in peutic effects of a drug substance. Among all the delivery routes of
humans [37]. Other studies showed that curcumin could be a promising curcumin, oral administration is an effective route to deliver curcumin
drug candidate for treating or preventing neurodegenerative diseases with numerous advantages over other administration routes. Therefore,
such as Alzheimer’s disease (AD) [38,39], Parkinson’s disease (PD) and it is suggested to focus more on the studies of oral administration of
brain tumours [21,40]. It has been reported that curcumin suppresses curcumin and the development of curcumin formulation for oral
the formation of amyloid β protein aggregates in the brain. The delivery.

2
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

3. Factors that account for curcumin’s poor oral bioavailability jejunum and colon. The absorption of curcumin in the ileum was the
poorest [79].
According to the Biopharmaceutics Classification System (BCS), Caco-2 cell lines (Fig. 2) possess the same cell polarity and compact
curcumin could be classified as a class IV substance since it has poor single-cell layer just like human intestinal epithelial cells and as a result,
aqueous solubility and limited permeability across the intestinal mem­ they are commonly used as the cell model for testing the intestinal
branes [54]. Apart from the poor solubility and intestinal permeability, permeability/absorption of drugs [80,81,82,83,77,84]. The results from
curcumin also has low serum concentration, poor pH dependent permeability test for curcumin reported by Zeng et al. [85] in Caco-2 cell
aqueous-stability and rapid metabolism in animals and humans, which lines exhibited that there was a decrease of permeability from the apical
are believed to be the main reasons that account for the poor oral side to the basolateral side (A to B direction) with an increase in cur­
bioavailability of curcumin [55,56,57,58,59,60,61,62,63,64,65,66,67, cumin concentration. Conversely, permeability from the basolateral side
68,69]. to the apical side (B to A direction) increased along with an increase in
the concentration of curcumin added. Furthermore, curcumin at con­
3.1. Poor aqueous and gastro-intestinal fluid solubility centration of 5 μg/ml, the value of PappA-B was greater than PappB-A
which indicated an increased absorption of curcumin. However, when
Curcumin is hydrophobic so it has extremely poor water solubility the curcumin concentration was increased to 10 μg/ml and 20 μg/ml,
[42]. Its aqueous solubility is also pH dependent. At acidic or neutral pH, the value of PappA-B became lower than PappB-A, indicating that the
it is practically insoluble. The maximum solubility of curcumin in amount of curcumin that is being pumped out from the cells is higher
aqueous solution at pH 5 was only 11 ng/ml [55,66]. It was reported than the amount being absorbed at this concentration range. It also
that the solubility increased to 0.1 μg/ml at pH 6.8. However, it dropped showed that the intestinal permeability of curcumin does not increase in
to 0.06 μg/ml when the pH was increased to 7.4 [60]. For any orally a concentration dependent manner for curcumin [85].
administrated drug to be absorbed, it needs to dissolve in the gastroin­ In transcellular studies, apparent permeability coefficient (Papp) was
testinal (GI) fluids. GI solubility of curcumin tested in simulated gas­ used to represent the amount of compound transported per unit time
troenteric fluids reported that curcumin showed equilibrium solubility [79]. It was reported that the value of Papp is correlated to the extent of
of 2.5 μg/ml in the fasted simulated gastric fluid ((pH1.6), 3 μg/ml in drug molecules that penetrate across the intestinal tract of the drugs [80,
simulated gastric fluid (pH1.2) and 2 μg/ml simulated intestinal fluid 86]). However, in another curcumin permeability study conducted using
(pH6.8) [70]. Compared with the solubilities tested in water or buffer Caco-2 cell line it was reported that efflux pathways do not play a role in
solutions, curcumin solubilities measured in simulated gastroenteric curcumin intestinal permeability. This observation was based on Papp
fluids seems to be much higher but still quite low. This reason for the (A to B) value (2.93 ± 0.94 × 10− 6 cm/s) was found higher than the
higher solubilities tested in the simulated gastroenteric fluids is un­ value of Papp (B to A) (2.55 ± 0.02 × 10− 6 cm/s) at curcumin con­
known but it might be due to the presence of pepsin in the simulated centration of 62.6 μg/ml (170 μM), [78].
gastroenteric fluids. There were studies which showed that pepsin can An efflux pump like P-glycoprotein could also affect the permeability
improve the solubility of lipophilic drugs although the mechanisms of curcumin. P-glycoprotein is an ATP dependent efflux pump that can
behind have not been founded yet [71,72]. pump drugs out of the cells thus reducing the bioavailability. In the
Curcumin has a low plasma concentration even after administering a small intestine, the expression of P-glycoprotein is present at ileum and
high oral dose. A clinical trial conducted in healthy human males even jejunum [87]. An in vitro study using Caco-2 cells showed that
after an oral dose of curcumin as high as 12 g the plasma curcumin co-administration of P-glycoprotein inhibitor such as verapamil, along
concentration was too low to be detected [45]. with curcumin could increase the rate absorption of curcumin [84]. This
In contrast, when 1 g/kg of curcumin was delivered orally to indicated that curcumin could potentially be a P-glycoprotein substrate
Sprague– Dawley rats and measuring plasma curcumin concentration and blocking P-glycoprotein can help to improve the permeability as
showed that the amount of curcumin present in the plasma was less than well as the drug absorption in the intestinal tract. Wand et al. [79] have
1% of the administrated dose [9]. A similar result was also reported by investigated a concentration dependent effect of verapamil on the ab­
Chang et al. [73], after an oral dose of 1 g/kg of curcumin in rodents sorption of curcumin in rat intestines. No obvious change to the ab­
resulted in curcumin plasma concentration of 15 ng/ml after 50 min sorption of curcumin was observed when verapamil concentration was
[73]. increased from 0.05 mmol/L to 0.1 mmol/L. When verapamil concen­
When at the same dose, human showed much lower curcumin tration was increased to 0.5 mmol/L, the rate of curcumin absorption
plasma concentration than rats. The same dose of curcumin (2 g/kg) was was increased by approximately 3%. This showed that P-glycoprotein
delivered orally to rats and humans respectively. The maximum plasma inhibitor like verapamil is capable to improve both the rate and the
concentration (Cmax) in rats was found to be 1.35 ± 0.23 μg/ml amount of curcumin absorbed in small intestines [79].
measured 50 min after taking the dose, whereas the Cmax of curcumin in Overall, from these studies, it can be concluded that curcumin has
humans was only 0.006 ± 0.005 μg/ml measured 1 h after administra­ poor permeability across intestinal barriers even after dissolution, thus
tion [8]. confirming that the curcumin can be classified as a class IV drug ac­
The results from these studies further suggests that curcumin has a cording to biochemical classification scheme (poorly soluble and poorly
very poor plasma concentration even after a high oral dose in both ro­ permeable).
dents and humans. When the same oral dose was administered, rats were
found to have higher plasma concentration of curcumin than humans.

3.2. Low permeability (absorption)

Apart from solubility, permeability is another main factor that affect


oral drug administration since the absorption of drugs depends on its
permeability across the biological membrane of the GI tract after
dissolution [74]. Experimental results in in vivo animal models and
simulated human intestinal epithelium barrier have shown that curcu­
min is poorly permeable across intestinal membranes even after disso­
lution [75,76,77,78]. It was found that in the rat small intestine, Fig. 2. The structure of Caco-2 cell monolayer grown at a permeable fil­
curcumin was best absorbed at the duodenal segment, followed by the ter support.

3
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

3.3. High metabolism rate of curcumin 3.4. Chemical stability of curcumin

Curcumin delivered orally undergoes extensive metabolic reduction Like its aqueous solubility, the chemical stability of curcumin in
and conjugation, which result in poor bioavailability [88]. The mecha­ aqueous solution is affected by change in pH. Curcumin is most stable at
nisms of curcumin metabolism are shown in Figs. 3 and 4. pH 1–6, when in the environment of stomach or small intestine. How­
The Phase I metabolism includes the reduction of the four double ever, the aqueous solubility becomes extremely poor at this pH range
bonds of the heptadiene-3, 5-dione structure of curcumin. The reduced while the stability of curcumin decreases significantly when the pH is
curcumin metabolites dihydrocurcumin, tetrahydrocurcumin, and hex­ above 7 [4,69]. In a stability test, using 5 mM curcumin solution (dis­
ahydrocurcumin were detected in plasma (rats and humans) following solved in methanol and added to 0.1 M phosphate buffer solution at pH
curcumin oral supplementation. The reduction processes were catalysed 7.2) incubated at 37 ◦ C for different lengths of time and analysed by
by NADPH-dependent curcumin reductases that exist in the liver [89, HPLC, the results showed that 90% of the curcumin was degraded within
57]. E. coli, the microorganism that commonly exists in small intestine 30 min to various degradation products including vanillin, ferulic acid
and colon, was found to have the ability to catalyse the metabolic and feruloyl methane (Fig. 5). Vanillin was identified to be the major
reduction processes of curcumin [90]. degradation product in this assay following first-order kinetics [69].
During the Phase II metabolism, glucuronidation and sulfation of Overall, the stability problem of curcumin really affects the oral
curcumin and the reduced metabolites take place. The conjugation ac­ bioavailability since curcumin can be rapidly degraded at alkaline so­
tivities are catalysed by UDP-glucuronosyltransferases and sulfo­ lutions and is poorly soluble in acidic aqueous solutions (even though
transferase respectively [91]. The conjugated metabolites include more stable).
curcumin sulfate, curcumin glucuronide sulfate, curcumin glucuronide,
dihydro curcumin glucuronide, tetrahydro curcumin glucuronide and 4. Novel formulations developed for improving curcumin’s oral
hexahydro curcumin glucuronide. In vivo studies in rats and humans bioavailability
showed that the curcumin glucuronide and curcumin glucur­
onide/sulfate conjugates were found in the plasma, jejunum and livers Numerous efforts have been made to find a way to overcome the low
[92,89,57,91]. An in vivo study conducted in rats showed that the oral bioavailability of curcumin and a great number of novel formula­
UDP-glucuronosyltransferases and sulfotransferase enzymes activities tions have been developed by researchers to achieve this goal. Details of
were found in livers, kidneys, small intestine mucosa and colon intes­ recent developments in curcumin formulations are discussed in the
tinal mucosa. No conjugated enzyme activities were found in the plasma following sections.
[92]. These observations might indicate that the conjugated curcumin
metabolites are probably produced in these tissues before present in the
plasma circulation. The glucuronidation activity was found to occur 4.1. Co-administration of adjuvants
more extensively in human intestines than in rats, while it became less
extensive in human liver than in rats. The difference of metabolism Some adjuvants can decrease the metabolism of curcumin which can
extension may be due to the differences in conjugation-hydrolysing help to improve the bioavailability of curcumin.
enzymes content in intestines & livers of humans and rats [89,57,91]. Piperine (Fig. 6) is an alkaloid naturally present in black pepper and
is the substance that gives black pepper (Piper nigrum) its pungency. Co-

Fig. 3. Sulphation and glucuronidation of curcumin (adapted from Ref. [4]).

4
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

Fig. 4. Reduction and glucuronidation of curcumin (adapted from Ref. [4]).

administration of piperine with curcumin was found to improve the


bioavailability of curcumin [93,8]. Piperine inhibits the enzyme
UDP-glucuronyltransferase in the liver thereby limiting the extent of
curcumin glucuronidation. Because of this process, more curcumin is
available for absorption [94]. An in vivo study was conducted in both
rats and humans at an oral dose of 2 g/kg of curcumin with 20 mg/kg
piperine, administered simultaneously [8]. The relative bioavailability
of curcumin in rats was increased by 1.54-fold while in the human
volunteers it increased by 20-fold. However, even though extent of in­
crease of bioavailability of curcumin was higher in human than in rats,

Fig. 5. Degradation products of curcumin (adapted from Ref. [69]).

Fig. 6. Chemical structure of Piperine.

5
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

the amount of curcumin actually being absorbed was in fact higher in


rats than in human (Fig. 7). In another clinical trial, 2 g of curcumin and
5 mg of piperine were co-administered orally to healthy human volun­
teers [1]. The test results revealed that there was an increase of ab­
sorption of curcumin by 200%, compared with that of taking curcumin
alone (Fig. 8). Another study by Zeng et al. [94] investigated the effect of
pre-administration of piperine on the oral bioavailability of curcumin. In
this study, rats received 20 mg/kg piperine initially followed by cur­
cumin (200 mg/kg) at various time intervals between 0.5 and 8 h after
piperine administration. The rats that received pre-administration of
piperine before the curcumin all showed remarkable enhancement in
curcumin oral bioavailability, especially at 6 h after taking piperine with
AUC0-t increased 97-fold when compared to pure curcumin. In contrast,
rats that received the same oral dose of piperine and curcumin at the Fig. 8. Comparison of curcumin oral absorption results of different piperine
same time exhibited much lower enhancement in AUC0-t, by 1.67-fold strategies tested in healthy human volunteers. (A) 2 g/kg curcumin with 20 mg/
relative to pure curcumin (Fig. 9). It was also found that piperine cau­ kg piperine and (B) 2 g/kg curcumin control administered in rats [8]; (C) 5 mg
ses time-dependent and selective suppression of piperine along with 2 g of curcumin and (D) 2 g curcumin control [1].
UDP-glucuronyltransferase and sulfotransferases, which reducing cur­
cumin metabolism [95,94].
Apart from piperine, several other natural compounds such as sili­
binin, and quercetin have been shown to inhibit the effects oF UDP-
glucuronyltransferase [96,97]. Quercetin is a plant flavonol that is
present in many fruits, vegetables and grains [98]. Silibinin is a flavo­
noid that extracted from the medicinal plant Silybum marianum (milk
thistle) [99]. The effect of co-delivering natural curcumin metabolism
enzyme inhibitors on the oral bioavailability of curcumin was studied
using a self-emulsion formulation of curcumin. The self-emulsion
formulation (containing 100 mg/kg curcumin) was delivered orally to
mice with or without either piperine (125 mg/kg), quercetin (100
mg/kg), or silibinin (100 mg/kg). The pharmacokinetic results showed
that piperine co-administration showed the highest Cmax of curcumin.
However, curcumin concentration in the plasma was highly variable
(0–2.4 μM curcumin plasma levels), which was deemed unacceptable. Fig. 9. Comparison of curcumin oral absorption results of two different
Co-administration of quercetin or silibinin showed much more stable piperine strategies tested in rats. (A) 2 g/kg curcumin with 20 mg/kg piperine
plasma levels of curcumin and both increased the average Cmax of cur­ and (B) 2 g/kg curcumin control administered in rats [8]; (C) initial dose of 20
cumin. Silibinin co-delivery resulted in higher Cmax of curcumin than mg/kg piperine followed by 200 mg/kg of curcumin after six hours and (D) 200
quercetin so the bioavailability study was only conducted for silibinin. mg/kg curcumin control [94].
The results reported that co-administration of silibinin improved the
overall bioavailability of curcumin by approximately 3.5-fold (AUC0–6 trihydroxystilbene), is a natural plant compound that is present in
0.2613 ± 0.0368 Vs. 0.0808 ± 0.0469 h μmol/L for SMEDDS without grapes, berries and other plants. All the three compounds showed an
adjuvant co-delivery) [100]. increase in curcumin permeability across the Caco-2 cell monolayers.
An in vitro Caco-2 cell monolayers permeability study examined how Quercetin and resveratrol increased the curcumin permeability by 1.46-
oral co-administration of piperine, quercetin or resveratrol can affect the fold and 1.25-fold, respectively. The highest effect on curcumin
intestinal absorption of curcumin. Resveratrol (trans-3,5,4′ - permeability was from piperine, which increased the permeability by
2.33-fold. Interestingly, when quercetin and resveratrol were combined
together with curcumin, the permeability increased by 1.85-fold. This
indicated that there is an additive effect of resveratrol and quercetin on
curcumin absorption [98].
Co-delivery of natural compounds like piperine, quercetin, resvera­
trol and silibinin decreased the metabolism of curcumin which eventu­
ally increased the absorption of curcumin. It is a promising and easy way
to improve the oral bioavailability of curcumin and could be investi­
gated further in the development of novel drug delivery systems.

4.2. Nanoparticles

Nanoparticle-based delivery system is another effective strategy to


improve the oral bioavailability of poorly water-soluble drugs. The
nano-size of the particle (10–1000 nm) leads to a greater surface area for
the drug particles thereby increasing the area of physical interaction
with the solvent. According to Noyes-Whitney equation, the dissolution
Fig. 7. Comparison of curcumin oral absorption results of the curcumin +
piperine co-administration strategy in rats and humans. (A) 2 g/kg curcumin rate is directly proportional to the surface area. Therefore, nano-sized
with 20 mg/kg piperine and (B) 2 g/kg curcumin control administered in rats; drug particles with higher surface area are likely to be dissolved more
(C) 2 g/kg curcumin with 20 mg/kg piperine administered to human volun­ rapidly. Furthermore, reducing the drug particle size to nanometer size,
teers; *: Pure curcumin absorption in humans was reported to be below the according to the Ostwald-Freundlich equation, can increase the satura­
detection limit [8]. tion solubility of the drug [101,102,103].

6
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

A list of approved nanodrugs is shown at Table 1. In comparison with with that of free curcumin suspension.
the conventional formulations, these nanodrugs benefited from Another novel curcumin-loaded nanoparticle was developed by
increased dissolution rate and saturation solubility, which leads to using an ionotropic gelation method. Alginate and polysorbate combi­
higher bioavailability [104]. nations were used to form the nanoparticles to entrap curcumin [111].
Nanoparticle technology is widely used for delivering poorly-water This formulation showed low release of curcumin in gastric and intes­
soluble drugs or substances [105,106,107,108]). A curcumin loaded tinal environments but a sustained release within the colon. This phe­
lipopolysaccharide nanoparticles (C-LPNCs) was developed to improve nomenon was due to colonic microflora, which helped to digest the
the oral bioavailability of curcumin [109]. Soluthin® MD, a alginate thus allowing curcumin to be released from the nanoparticles.
phosphatidylcholine-maltodextrin based hydrophilic lipopolysaccha­ In the human body, at a curcumin dose of 100 mg, the nanoparticle
ride, was used as the nanocarrier to load curcumin. Poloxamer 188 was formulation increased the oral bioavailability of curcumin by 5-fold than
added as a stabilizer to prevent aggregation and size growth of the that of free curcumin suspension. Since most of the curcumin was
C-LPNCs. A graphical scheme of this formulation is shown in Fig. 10. released and absorbed in the colon, this formulation might be a prom­
Burst dissolution of the curcumin was observed in the first 30 min at pH ising approach for oral delivery of curcumin for targeting colonic
7.4 buffer solution, which was due to the amorphous distribution of the diseases.
curcumin particles in the C-LPNCs. Subsequently the dissolution rate Applying coating to the surface of nanoparticles may help to prevent
was reported to gradually reduce and became relatively stable after 12 agglomeration to occur. Commonly used polymers for such coating are
h. After 24 h, 80% of the curcumin in the C-LPNCs dissolved in the so­ polyethylene glycol (PEG), poly (vinylpyrrolidone) (PVP), dextran,
lution. The C-LPNCs were evaluated in vivo to investigate its effect on the chitosan, pullulan etc. Surfactants like sodium oleate and dodecylamine
oral bioavailability. A curcumin equivalent dose of 50 mg/kg was have also been used for nanoparticle coating [112]. In another study, a
administrated orally to Albino Wister rats. The Cmax and AUC0–t of curcumin-loaded solid lipid nanoparticles (SLNs) with N-carboxymethyl
curcumin was found to have increased by 104.8-fold and 55.2-fold chitosan (NCC) coating was prepared through a modified hot homoge­
respectively when administrated as the C-LPNCS, compared with the nization and sonication method [113]. The NCC coated SLN samples
unformulated curcumin. Lymphatic transport was found to be involved exhibited 9.5-fold higher oral bioavailability than that of curcumin so­
in the oral absorption of C-LPNCs, which can reduce curcumin hepatic lution. Inhibition of burst release (over-rapid release) of curcumin was
metabolism in liver thus contributing to the increase in curcumin observed in NCC coated SLNs in simulated gastric fluid (pH1.2) thus
bioavailability. In addition, maltodextrin in the formulation can protect indicating that NCC coating was able to address the burst release
curcumin from degradation in the harsh environment of the gastroin­ problem that SLNs usually have in an acidic environment.
testinal tract, which could be another reason for the improvement of the Among the nanoparticle formulations mentioned above, the nano­
oral bioavailability. Anticancer activity of C-LPNCs against colon cancer suspension prepared by CO2 assisted in situ nano-amorphization method
was investigated in vivo in colon-26 tumour bearing rats. A curcumin [110] exhibited the highest curcumin oral bioavailability with the
equivalent dose of 50 mg/kg was delivered orally daily to the rats for 30 AUC0-t value of 365.73 μg h/ml in rats. A comparison of the curcumin
days. At the end of the study, 100% survival rate was observed in the oral absorption data between each nanoparticle formulation is shown in
group being treated with the C-LPNCs while 33.33% survival rate was Fig. 11.
observed in the group receiving pure curcumin [109].
In another study, a simple method called CO2-assisted in situ nano- 4.3. Liposomes
amorphization was used to prepare curcumin-lipopolysaccharide
based oral nanosuspension to improve the bioavailability and anti­ Liposomes are a type of drug delivery system which contain a
cancer efficacy of curcumin [110]. In this method, curcumin, citric acid phospholipid bilayer structure giving them an ability to encapsulate
and a stabilizer, Brij78, were first dissolved in ethanol. The solvent was both hydrophilic and hydrophobic substances [1]. As a drug delivery
then removed via evaporation, resulting in the formation of a solid approach, liposomes have the advantage of being biodegradable,
mixture. Subsequently carbonated solution was added to the solid nontoxic, and remarkably biocompatible [114].
mixture to form the curcumin loaded nanosuspension. In an in vivo test A hybrid liposomal formulation that consisted of chitosan and soy­
in rats, it was reported that the oral bioavailability of curcumin had bean lecithin was developed as a drug vehicle for curcumin [115].
improved by 3.70-fold from the CUR/Brij78 nanosuspensions compared Bioavailability of curcumin from the liposomal formulation was testing
using caco-2 cells. The hybrid liposomes exhibited higher cellular up­
take rate than that of phospholipid liposome, 1.66-fold higher at 0.5 h
Table 1
after incubation and 1.256-fold higher after 4 h incubation. The re­
A List of FDA approved nanodrugs [104].
searchers reported a sustained release of curcumin, increased positive
Trade name Generic name Route of Indication
charge and decreased hydrophobicity from the hybrid liposomal
administration
formulation were the main reasons for the increased curcumin
Emend® Aprepitant as Oral Preventing nausea and bioavailability.
nanocrystal vomiting
In another study, a liposomal formulation coated with chitosan was
Megace ES® Megestrol Oral Treating loss of appetite and
acetate as wasting syndrome in people tested in simulated gastric fluid (pH between 2 and 3) and in simulated
nanocrystal with acquired intestinal fluid (pH 7). It was reported that around 80% of the curcumin
immunodeficiency loaded in the liposome dissolved in the simulated gastric and the
syndrome (AIDS).
simulated intestinal fluids [116].
Rapamune® Rapamycin Oral Immunosuppressant
(sirolimus) as In the research area of curcumin liposomal formulation, the current
nanocrystals trend is to use hybrid liposomal formulations to help to improve the
formulated in bioavailability of curcumin. The studies mentioned above showed that
tablets the hybrid liposomal formulations have better performance in
Tricor® Fenofibrate as Oral Treating
improving bioavailability of curcumin than the conventional curcumin-
nanocrystals hypercholesterolemia and
hypertriglyceridemia loaded liposomes and the unformulated curcumin.
Triglide® Fenofibrate as Oral Treating
nanocrystals hypercholesterolemia and 4.4. Micelles and polymer micelles
hypertriglyceridemia

*Compared with the conventional formulations. Micelles are spherical aggregates of surfactants that form

7
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

Fig. 10. Graphic scheme of the Curcumin loaded phosphatidylcholine-maltodextrin based lipopolysaccharide nanoparticles (C-LPNCs) formulation (adapted
from Ref. [109].

comparison with free curcumin. The improved oral bioavailability and


anti-cancer effects of curcumin was attributed to the controlled release
of curcumin from the CUR-MM, increase of curcumin solubility from
micellization and inhibition of P-glycoprotein by the surfactants.
In another study, a sophorolipid nano-micelle formulation was pre­
pared by using a ‘pH-driven method’ [120]. In vivo test results showed
that it increased curcumin’s oral bioavailability by 3.6-fold compared
with the unformulated curcumin. This proves that biosurfactants like
sophorolipid, obtained from natural sources, could be used for formu­
lating drug delivery systems to enhance the oral bioavailability of cur­
cumin. The preparation method used in this study, the pH-driven
method, does not require heat and organic solvents, thus making it a
simple and cost-effective way for producing nano-sized curcumin
products.
Fig. 11. Comparison of curcumin oral absorption results of the novel curcumin-
A clinical trial was conducted among 23 healthy human candidates
loaded nanoparticle formulations at the curcumin equivalent oral doses of (A) (13 male and 10 female) to assess a micellar curcumin formulation that
50 mg/kg for the curcumin loaded lipopolysaccharide nanoparticles, curcumin composed of micronised curcumin powder and Tween-80 [121]. The
control concentrations below limit of detection in this study [109]; (B) 50 result of pharmacokinetics studies showed that the ingestion of a single
mg/kg for the nanosuspension prepared by CO2-assisted in situ oral dose of 500 mg micelle encapsulated curcumin (equivalent to 410
nano-amorphization method and (C) curcumin control [110]; (D) 100 mg for mg curcumin) resulted in a Cmax of 3.228 μmol/L compared to 0.007
the Alginate-polysorbate 80 nanoparticles and (E) [111]; and (F) 50 mg/kg for μmol/L of free curcumin powder. Both male and female subjects have
the solid lipid nanoparticles (SLNs) with N-carboxymethyl chitosan (NCC) almost the same Tmax, but the female subjects exhibited higher Cmax of
coating, tested in rats, curcumin control concentrations was reported to be 3.701 μmol/L in comparison with 2.612 μmol/L for male subjects,
below the limit of detection [113].
indicating that women might be absorbing more curcumin than men in
this study.
spontaneously when the surfactants reach a critical micelle concentra­ A novel self-assembled, curcumin-loaded polymeric micelle was
tion (CMC) at a certain temperature in aqueous solution [117]. The size developed for improving curcumin oral absorption [122]. It consisted of
of micelles normally range between 20 and 80 nm. Typical micelles are di-tocopherol polyethylene glycol 2000 succinate (TPGS2 K), octadeca­
comprised of an outer shell formed by hydrophilic head groups and a noic acid, 12-hydroxy-, polymer with alpha-hydro-omega-hydroxypoly
core formed by hydrophobic tail groups. Micelles can be used as vesicles (oxy-1,2-ethanediyl) and Pluronic F127. Both in vitro and in vivo test re­
to load and deliver poorly water-soluble drugs. The hydrophobic core sults revealed that the micelle system provided a better intestinal ab­
can encapsulate the lipophilic drugs and the hydrophilic outer shell to sorption of curcumin than free curcumin. Oral administration of the
protect the drugs from being inactivated in the gastrointestinal envi­ micelle formulation in rats resulted in higher AUC0-t of curcumin (870.2
ronment. As a result, solubilization and stability of the drugs are ± 466.78 mg h/L) compared with the standard curcumin (AUC0-t of
improved [118]. 303.58 ± 294.31 mg h/L). Furthermore, lymphatic transport pathways
A novel curcumin-loaded mixed micelle (CUR-MM) that consist of were found to be involved in the absorption of the micelle system, which
two surfactants (Pluronic F-127 and Gelucire® 44/14) was developed helps curcumin to avoid first-pass metabolism.
[119]. The formulation showed that it can enhance the oral bioavail­ Overall, a major increase in oral bioavailability of curcumin was
ability of curcumin and its anti-cancer effect. The optimised CUR-MM demonstrated in the recently developed novel micelles formulations.
formulation with size of 188 ± 3 nm and an entrapment efficiency of These results indicate that micelles are a promising way for delivering
76.45 ± 1.18% w/w exhibited significant enhancement in the oral curcumin orally and worthy of further studies in the future. A compar­
bioavailability of curcumin (by approximately 55-fold) and cytotoxicity ison plot to describe the results of curcumin oral bioavailability from
to human lung cancer cell line A549 (by approximately 300%), in

8
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

different micellar formulations is shown in Fig. 12. [124] showed the highest curcumin oral absorption with the AUC0-t value of
343.8 μg h/ml. In contrast, the other two nanoemulsion formulations [126,
4.5. Curcumin-loaded nano and micro-emulsions 125] have much poorer curcumin bioavailability with AUC0-t value of 0.7
μg h/ml and 2.97 μg h/ml (Fig. 13).
Nanoemulsions, also known as submicron emulsions, is another type of Microemulsion is an isotropic colloidal system of micron-sized droplets
nano-sized drug delivery system. They are single phase, isotropic thermo­ that consists of water, oil, surfactant and cosurfactants. When in a micro­
dynamically stable systems that consist of emulsified oil, water and emulsion system, drug compounds can be entrapped within the oil droplets,
amphiphilic molecules (surfactants). The size of the emulsion droplets which protect them from both enzymatic and chemical degradations thus
typically range between approximately 10–1000 nm [123]. A Thiol modi­ increasing the residence time and bioavailability of the drugs [127]. Inter­
fied chitosan coated, curcumin-piperine loaded oil-in-water nanoemulsion estingly, the droplet size of microemulsion is not in micrometre but between
was developed [124]. The optimised formulation has nano-droplets size of 10 and 100 nm. Microemulsion is fairly similar to nanoemulsion in terms of
110 nm, weight ratio of curcumin: piperine in 100 to 1 and chitosan thio­ the structure. What differentiating it from nanoemulsion is that micro­
lation level of 14–15%. At an oral dose equivalent to 8 mg/kg of curcumin, emulsion is thermodynamically stable whereas nanoemulsion is not. Also,
the AUC0-t value of curcumin increased by 64-fold (343.3 μg h/ml) than that microemulsion normally have a narrow particle size distribution compared
of standard curcumin (5.4 μg h/ml). Comparing with the two piperine to nanoemulsions [128]. A self-emulsifying microemulsion drug delivery
strategies that were reviewed in section 3.1 [8,94], the optimised system (SMEDDS) was developed to improve the oral bioavailability of
curcumin-piperine loaded nanoemulsion formulation is 94-fold and curcumin [129]. A semi-synthetic bicephalous heterolipid known as E1E
1505-fold higher in terms of the AUC0-t values. This indicates that the was used in this formulation, along with Solutol HS15 (a surfactant) and
combination of piperine with the nanoemulsion technology is much more Transcutol HP (a cosurfactant) to form the microemulsion encapsulating
effective than only using piperine alone in improving the oral bioavailability curcumin. An optimised SMEDDS formulation with drug loading efficiency
of curcumin. In another study, a novel lipid nanoemulsion containing cur­ of 70.52 ± 2.46 mg/g was orally administrated to rats at a curcumin dose of
cumin (CNELNs) was prepared and tested for changes in bioavailability in in 50 mg/kg. Plasma analysis showed that the Cmax and AUC 0-t of curcumin
vitro and in vivo models [125]. The mean absorption constants (Ka) and from the SMEDDS were 4.921 ± 0.42 (μg/mL) and 17.30 ± 1.50 (μg h/ml)
effective permeabilities (Peff) over the whole intestine system were respectively, in comparison with free curcumin suspension with 0.186 ±
increased by 2.98-fold and 6.65-fold, respectively. The relative bioavail­ 0.006 (μg/ml) and 0.234 ± 0.023 (μg.h/ml) respectively. Tmax remained
ability of CNELNs to standard curcumin was 733.59%, which was possible unchanged in both SMEDDS and the free curcumin suspension 60 min after
due to the increase of intestinal absorption. CNELNs also displayed stronger oral administration. In another study, a self-microemulsifying curcumin
effect in suppressing the growth of human lung cancer cell A549. A (SME-Cur) entrapped in a hydroxypropylmethyl cellulose (HPMC)-based
curcumin-phospholipid complex was incorporated into a self-nanoemulsion sponge was developed to study oral absorption of curcumin [130]. The in
system consisting of castor oil, Tween 80 and PEG 400 [126]. The combined vivo study was performed in rabbits at a curcumin equivalent oral dose of
formulation showed significant higher serum concentration of curcumin 12.5 mg/kg. The HPMC sponge entrapped SME-Cur showed significantly
than free curcumin and curcumin-phospholipid complex. Using an oral dose higher Cmax and AUC0-t (1.77 ± 0.13 mg/ml and 2615.68 ± 97.79 ng/h/ml,
of 100 mg/kg in rats, the Cmax of curcumin from the combined formulation respectively) than unformulated curcumin at dose of 50 mg/kg (0.30 ± 0.03
was 487.7 ± 53.4 ng/ml, compared to 21.6 ± 3.6 ng/ml for free curcumin mg/ml and 509.49 ± 47.77 ng h/ml, respectively). The in vitro drug release
and 54.6 ± 3.7 ng/ml for curcumin-phospholipid complex. The relative test found that the HPMC-based sponges entrapped SME-Cur provided a
bioavailability of curcumin from the combined formulation was 7.67-fold sustained release of curcumin and led to a higher percentage release
and 52.55-fold higher than that of curcumin-phospholipid complex and compared with the unwrapped SME-Cur and free curcumin. The improved
unformulated curcumin. Several research studies have been carried out to drug release could be attributed to the drug-polymer interaction leads to the
study the potential for using nanoemulsion strategy to improve the oral inhibition of precipitation and crystallization of curcumin in solution.
bioavailability of curcumin. The results of these studies demonstrated that it Compared to nanoemulsions, microemulsions have received less
is a promising strategy to fulfil the goal of improving curcumin’s oral attention since there were only a few studies on their use in improving
bioavailability. Among the nanoemulsion formulations reviewed, piperine the oral bioavailability of curcumin (Fig. 14). However, the studies of
loaded in an oil-in water nanoemulsion with modified chitosan coating microemulsions reviewed in this article both showed considerable

Fig. 12. Comparison of curcumin oral absorption of curcumin loaded micelles


formulation (A) curcumin oral doses of 10 mg/kg for curcumin-loaded mixed Fig. 13. Comparison of curcumin oral absorption results of the novel nano­
micelle and (B) curcumin control [119]; (C) 100 mg/kg for the curcumin loaded emulsion formulations (A) at an oral dose of 8 mg/kg for Chitosan coated,
sophoroliplid-coated nanomicelles and (D) curcumin control [121]; (E) 50 piperine loaded oil in water nanoemulsion (B) curcumin control [124] and (C)
mg/kg for the self-assembled curcumin-loaded polymeric micelle, concentra­ oral dose of 100 mg/kg of curcumin-phospholipid complex incorporated
tion of curcumin from the control sample was reported to be below the limit of self-nanoemulsion, curcumin control values was reported to be below the limit
detection [122]. of detection [126].

9
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

improvement in curcumin’s oral bioavailability [129,130]. All of these Table 2


indicate that microemulsions are still a potent and promising technology A list of FDA approved solid dispersion drugs [141].
to overcome curcumin’s oral delivery problems and it is worthy of Trading Active Carrier Preparation Dosage
further studies in the future. name ingredient method form

Kalydeco® Ivacaftor HPMCAS Spray drying Tablet


Zelboraf® Vemurafenib HPMCAS Coprecipitation Tablet
4.6. Solid dispersion Incivek® Telaprevir HPMCAS Spray drying Tablet
Intelence® Etravirine HPMC Spray drying Tablet
By definition, a solid dispersion is a solid product where at least one Novir® Ritonavir PVP/PA Melt extrusion Tablet
drug is entrapped in a hydrophilic polymeric carrier and the drug(s) are Kaletra® Lopinavir PVP/PA Melt extrusion Tablet
molecularly dispersed in the polymeric carrier [131]. The principle of
solid dispersion to improve the drug bioavailability was to first rapidly
prepared with disodium glycyrrhizin (Na2GA) via ball milling [142].
release the drug particles from the polymeric carrier to create a super­
Na2GA was able to form micelles when dissolved in water and encap­
saturated solution after which the concentration is maintained long
sulate the amorphous curcumin particles. In vivo evaluation showed that
enough for the drug to be absorbed [132,133,134]. Drugs in supersat­
the self-micelle forming solid dispersion provided a 19-fold increase in
uration state tend to precipitate rapidly before the absorption to take
curcumin oral bioavailability over free curcumin. In addition, the solid
place which results in reduced bioavailability. Fortunately, the polymer
dispersion formulation improved curcumin permeability across the
excipients that are commonly used for preparing solid dispersion also
gastrointestinal membranes.
have the ability to inhibit drug precipitation and prolong the supersat­
A curcumin-loaded ternary solid dispersion system was prepared by
uration, such as hydroxypropylmethyl cellulose (HPMC), hydrox­
using mannitol as the hydrophilic carrier and D-α-Tocopheryl poly­
ypropylmethyl cellulose acetate succinate (HPMC-AS) and vinyl
ethylene glycol 1000 succinate (vitamin E TPGs) as the surfactant [66].
polymers such as poly (vinylpyrrolidone) (PVP). The mechanism of how
TPGs was selected due to its safety and potential to inhibit P-glycopro­
polymers prolong drug supersaturation is still not fully understood.
tein efflux. The aqueous solubility of curcumin from the solid dispersion
Nevertheless, it is generally believed to be as a result of hydrogen bond
formulation remained stable across a pH range from 1.2 to 7.4, which
formation between the polymers and the drugs, based on the observa­
diminished the pH-dependent solubility pattern in pure curcumin
tion that polymers are effective in inhibiting precipitation of drugs rich
powder. Pharmacokinetics studies revealed that the Cmax and AUC0-t of
in hydrogen-bond acceptors [135,136,137]. Solid dispersion continues
curcumin were increased by 86- and 65-fold respectively, in comparison
to be a widely studied strategy to improve oral bioavailability of poorly
with pure curcumin powder. The solid dispersion formulation also
water-soluble drug candidates [138,139,140]. A list of FDA approved
exhibited an inhibition effect on P-glycoprotein and this was believed to
solid dispersion drugs is shown at Table 2.
be the main reason for the increased curcumin intestinal absorption
A recent study combined solid dispersion strategy with nanotech­
observed from the solid dispersion system.
nology and developed a novel self-nanomicellizing solid dispersion
A curcumin-loaded solid dispersion formulation was developed by
(NCF) for delivering curcumin orally [81]. Soluplus® (polyvinyl
using Gelucire 50/13 (a non-ionic water-dispersible surfactant) as the
caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer)
carrier material [143]. The solid dispersion formulation was prepared
was used as a carrier for curcumin in the NCF formulation. NCF
by spray drying. The aqueous solubility of curcumin was significantly
exhibited significant increase in aqueous solubility of curcumin (over
increased by the solid dispersion (2700 μg/ml), which was 3600-fold
20,000-fold higher than the curcumin control). Considerable improve­
higher than the unformulated curcumin (0.75 μg/ml). The solid
ment in dissolution rate and small-intestinal permeability of curcumin
dispersion demonstrated rapid release of curcumin at 10 min, with 90%
were also found with the NCF formulation. Furthermore, improved
and 73% of curcumin being released at pH 1.2 and 7.4, respectively. The
curcumin stability in alkaline environment was found. As a result of the
percentage of curcumin released was also considerably higher than pure
increased dissolution, solubility, permeability and alkaline solution
curcumin. Results from rat plasma sample analysis indicated a 5.5-fold
stability, the oral bioavailability of curcumin from the NCF was signif­
increase of curcumin plasma concentration from the solid dispersion
icantly enhanced by 117-fold compared with pure curcumin.
compared with the unformulated curcumin under the same oral dose
In another study, an amorphous curcumin solid dispersion was
(500 mg/kg). Since spray drying is a technique that is suitable for
scaled-up production, solid dispersion formulation developed in this
study could be a promising way to improve curcumin bioavailability
from an industrial perspective.
In another study, an excipient-free co-amorphous curcumin-piperine
solid dispersion (co-amorphous CUR-PIP) was prepared by a melting and
quench cooling method [144]. This formulation has a combined solid
dispersion effect of increasing drug dissolution along with piperine’s
effect of inhibiting the glucuronidation of curcumin in vivo. In this study,
higher dissolution rate and longer-lasting supersaturated curcumin
concentration were observed from the co-amorphous CUR-PIP. This
could be due to piperine’s effects to suppress UDP-glucuronyltransferase
and sulfotransferases in the intestines and liver [94]. In addition, the
permeability of curcumin across the gastrointestinal membrane was
elevated by 2.67-fold compared with pure curcumin, which was linked
to piperine’s ability to inhibit curcumin glucuronosyltransferases. As a
result of faster, longer-lasting supersaturation dissolution and higher GI
permeability, the oral bioavailability of curcumin was enhanced by
2.16- and 1.92-fold with the co-amorphous CUR-PIP relative to the
Fig. 14. Comparison of curcumin oral absorption results of the novel micro­
emulsion formulations at the curcumin equivalent doses of (A) 50 mg/kg for the
crystalline and amorphous curcumin. Comparing with the previously
self-emulsifying microemulsion and (B) curcumin control [129] and (C) 12.5 mentioned strategies that used piperine for curcumin, the co-amorphous
mg/kg for the HPMC sponge entrapped self-microemulsifying curcumin and (D) CUR-PIP showed around 8-fold and 134-fold higher AUC0-t values than
curcumin control [130]. the curcumin-piperine co-administration strategy [8] and the

10
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

pre-treatment of piperine strategy [94]. However, it is around 11-fold


lower compared to the curcumin-piperine loaded nanoemulsion [124].
A comparative graph between the solid dispersion formulation and the
nanoemulsion formulation is shown in Fig. 15.
The results from the studies mentioned above suggest that solubility
and dissolution of curcumin can be greatly improved by solid dispersion
strategies, thus leading to improvement in curcumin’s oral bioavail­
ability. The selection of carriers and surfactants also play a vital role in
the performance of solid dispersion formulations in improving oral
bioavailability of curcumin.
Among all the solid dispersion formulations reviewed in this article,
the self-nanomicellising solid dispersion prepared using Soluplus [81]
exhibited significantly higher curcumin oral absorption compared with
the other solid dispersion formulations. A comparative graph is shown in
Fig. 16.
Fig. 16. Comparative graph of curcumin oral absorption results of the novel
5. Conclusions solid dispersion formulations at the curcumin equivalent oral doses of: (A) 47
mg/kg for Soluplus containing self-nanomicellizing solid dispersion and (B)
Decades of studies have proved that curcumin can be well-tolerated curcumin control [81]; (C) 150 mg/kg for ball milled curcumin mixed with
in human bodies, with numerous therapeutic potentials. However, Na2GA in a solid dispersion and (D) curcumin control [142] and (E) 100 mg/kg
despite curcumin’s pharmacological efficacy and safety, it is still not for co-amorphous curcumin-piperine solid dispersion, curcumin levels from the
licenced as a valid therapeutic agent mainly due to the very poor oral control sample were below the limit of detection; The AUC0-t values of the pure
bioavailability of curcumin. As discussed earlier in this article, the main curcumin (control) of this study was reported to be below the limit of detec­
factors that contribute to the poor oral bioavailability of curcumin are tion [144].
low aqueous solubility, poor intestinal permeability, chemical stability,
rapid metabolism and elimination. vitro model, NCF (equivalent to 10 μg/ml of curcumin) displayed better
A range of novel formulations have been developed in recent years efficacy than CUR control (10 μg/ml) against cytotoxicity which induced
and many of them have demonstrated considerable improvement in the by CuSO4, H2O2 and Aβ42. This indicated that NCF has better neuro­
oral bioavailability of curcumin. A summary of these novel de­ protective effect than the unformulated curcumin. As for the in vivo
velopments is given in Table 3. The curcumin oral absorption results of study, NCF, curcumin control and donepezil (a medication used for
the formulation with obvious increase in curcumin bioavailability are treating Alzheimer’s diseases) were orally administrated to mice in
summarised and compared in Fig. 17. From this data it can be sum­ order to investigate their long-term (3 months) efficacy against Alz­
marised that the self-nanomicellizing solid dispersion (NCF) prepared by heimer’s diseases. The efficacy was evaluated by behavioural tests of the
Soluplus showed the highest curcumin oral bioavailability with the mice and the results showed that the NCF has significantly better effi­
AUC0-t value of 1998.6 ± 361.5 μg h/ml at a curcumin equivalent dose cacy to repair the cognitive functions and behaviour of mice than the
of 47 mg/kg in rats [81]. This shows the great potential of solid curcumin control and even donepezil. Also, it has been reported that it
dispersion strategy for the application in improving curcumin absorp­ prevents any further deterioration in the behaviour of mice. During the
tion via oral administration. Whether improvement of oral bioavail­ in vivo study, the formulation was well-tolerated with no obvious
ability of curcumin can improve the therapeutic efficacy of the toxicity. This makes NCF a safe and effective option for the treatment of
compound needs to be evaluated. A follow-up study was carried out to Alzheimer’s diseases. It also proved that the improvement of the oral
evaluate the efficacy of NCF against Alzheimer’s disease in in vitro bioavailability of curcumin could lead to good therapeutic effect [145].
(SHSY5Y695 APP human neuroblastoma cell line with induced toxicity) However, it is difficult to directly compare each formulation because
and in vivo (transgenic mice with Alzheimer’s diseases) models. In the in of the differences in administered dose, experimental designs, and
methods of analysis. No cross-over studies have been conducted for the
novel curcumin formulations mentioned in this article. It is suggested
that in order to design standardised controlled trials for different cur­
cumin formulations in the future, it is imperative to understand and
verify which formulation is the best in terms of oral bioavailability and
therapeutic effect.
In summary, curcumin has been demonstrated to have a wide range
of therapeutic effects. However, due to its physical properties it is seri­
ously lacking in aqueous solubility, which has been related to its poor
bioavailability. Advances in the development of novel formulations
should assist in improving bioavailability, which can then be related to
categorically demonstrate curcumin’s therapeutic effect in a given
condition.

Funding

This research did not receive any specific grant from funding
agencies in the public, commercial or not-for-profit sectors.

Fig. 15. Comparison of the curcumin oral absorption results at the curcumin CRediT Author Statement
equivalent oral doses of (A) 8 mg/kg for the curcumin-piperine loaded nano­
emulsion formulation [124] and (B) 100 mg/kg for the curcumin-piperine Zhenqi Liu: Investigation, Writing - original draft, Visualization,
loaded solid dispersion formulation [144]. Methodology, Resources. John D. Smart: Supervision, Reviewing.

11
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

Table 3
A summary of the novel formulations for improving curcumin’s oral bioavailability.
Subjects Amount of curcumin Formulation Improved the oral bioavailability of References
administrated curcumin by

Humans 2g 2 g of curcumin + 5 mg of piperine was delivered orally simultaneously 2-fold [1]


Rats 200 mg/kg Pre-administration of 20 mg/kg piperine, then 200 mg/kg curcumin 97-fold [94]
after 6 h
Albino Wister rats 50 mg/kg Curcumin loaded phosphatidylcholine-maltodextrin based 55.2-fold [109]
nanoparticles (C-LPNCs)
Male SD (Sprague 50 mg/kg Curcumin loaded nanosuspension, prepared by citric acid and Brij78 3.7-fold [79,110]
Dawley) rats
Humans 100 mg Curcumin loaded, Alginate and polysorbate nanoparticles 5-fold [111]
Male SD rats 50 mg/kg Curcumin-loaded solid lipid nanoparticles (SLN) with N- 9.5-fold [113]
carboxymethyl chitosan (NCC) coating
Caco-2 cells 4 μg/ml Curcumin-loaded hybrid liposomal formulation that consist of chitosan 1.256-fold higher 4 h after incubation. [115]
and phospholipid
Male wistar rats 10 mg/kg Curcumin-loaded mixed micelle (consist of Pluronic F-127 and 55-fold [119]
Gelucire® 44/14)
Male SD rats 100 mg/kg Curcumin-loaded sophorolipid based nanomicelle 3.6-fold [120]
Rats 50 mg/kg Self-assembled, polymeric curcumin-loaded micelles, consist of TPGS2 2.87-fold [122]
K, HS15 and Pluronic F127
Male Wistar albino 8 mg/kg Thiol modified chitosan coated, curcumin-piperine loaded, oil-in water 64-fold [124]
rats nanoemulsion
Male SD rats 100 mg/kg Curcumin-loaded self-nanoemulsifying drug delivery system 1.94-fold [125]
(SNEDDS), consist of ethyl oleate, tween 80 and PEG 600
Rats 100 mg/kg Curcumin-phospholipid complex, entrapped in a self-nanoemulsion 52.55-fold [126]
that consist of caster oil, Tween 80 and PEG 400
Rats 50 mg/kg Self-emulsifying curcumin loaded microemulsion that consist of E1E, 73-fold [129]
Solutol HS and Transcutol HP
Adult male Wistar 12.5 mg/kg Self-microemulsifying curcumin (SME-Cur) wrapped in a 5-fold [130]
rats hydroxyprophylmethyl cellulose (HPMC)-based sponge
Male SD rats 47 mg/kg Self-nanomicellizing curcumin-loaded Soluplus solid dispersion 117-fold higher than that of 150 mg/ [81]
kg curcumin suspension
Male S D rats 150 mg/kg Ball milled curcumin loaded, disodium glycyrrhizin (Na2GA) solid 19-fold [142]
dispersion
Male SD rats 30 mg/kg Curcumin-loaded ternary solid dispersion system, prepared with 65-fold higher than 200 mg/kg oral [66]
Mannitol and TPGs dose of pure curcumin
Rats 500 mg/kg Curcumin-Gelucire 50/13 solid dispersion 5.5-fold [143]
Rats 100 mg/kg curcumin-piperine solid dispersion 1.92-fold [144]

Ananth S. Pannala: Conceptualization, Supervision, Writing - review &


editing.

Declaration of competing interest

The authors declare that there are no conflict of interests.

References

[1] P. Anand, A. Kunnumakkara, R. Newman, B. Aggarwal, Bioavailability of


Curcumin: Problems and Promises. Mol. Pharm 4 (6) (2007) 807–818, https://
doi.org/10.1021/mp700113r.
[2] A. Araiza-Calahorra, M. Akhtar, A. Sarkar, Recent advances in emulsion-based
delivery approaches for curcumin: from encapsulation to bioaccessibility, Trends
Food Sci. Technol. 71 (2018) 155–169, https://doi.org/10.1016/j.
tifs.2017.11.009.
[3] R. Carolina Alves, R. Perosa Fernandes, B. Fonseca-Santos, F. Damiani Victorelli,
M. Chorilli, A critical review of the properties and analytical methods for the
determination of curcumin in biological and pharmaceutical matrices, Crit. Rev.
Fig. 17. Comparison of curcumin oral absorption results of the different cur­ Anal. Chem. 49 (2) (2018) 138–149, https://doi.org/10.1080/
10408347.2018.1489216.
cumin oral bioavailability enhancement strategies, at the curcumin oral doses
[4] T. Esatbeyoglu, P. Hubbe, I.M.A. Ernst, D. Chin, A.E. Wagner, G. Rimbach,
of: (A) 47 mg/kg for the Soluplus containing self-nanomicellising solid Curcumin – from molecule to biological function, Angew. Chem. Int. Ed. Engl. 51
dispersion [81], (B) 50 mg/kg for the Nanosuspension prepared by CO2-assisted (22) (2012) 5308–5322, https://doi.org/10.1002/anie.201107724.
in situ nano-amorphization method [110], (C) 8 mg/kg for curcumin + piperine [5] M. Bernabé-Pineda, M. Ramı ́rez-Silva, M. Romero-Romo, E. González-Vergara,
loaded nanoemulsion with thiol modified chitosan coating [124], (D) 100 A. Rojas-Hernández, Determination of acidity constants of curcumin in aqueous
solution and apparent rate constant of its decomposition, Spectrochim. Acta Mol.
mg/kg for the co-amorphous curcumin-piperine solid dispersion [144] and (E)
Biomol. Spectrosc. 60 (5) (2004) 1091–1097, https://doi.org/10.1016/S1386-
50 mg/kg for the self-emulsifying microemulsion [129]. 1425(03)00342-1.
[6] S. Jovanovic, S. Steenken, C. Boone, M. Simic, H-atom transfer is A preferred
antioxidant mechanism of curcumin, J. Am. Chem. Soc. 121 (41) (1999)
9677–9681, https://doi.org/10.1021/ja991446m.
[7] V. Ravindranath, N. Chandrasekhara, Absorption and tissue distribution of
curcumin in rats, Toxicology 16 (3) (1980) 259–265, https://doi.org/10.1016/
0300-483x(80)90122-5.

12
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

[8] G. Shoba, D. Joy, T. Joseph, M. Majeed, R. Rajendran, P.S. Srinivas, Influence of [33] A. Saeidinia, F. Keihanian, A.E. Butler, R.K. Bagheri, S.L. Atkin, A. Sahebkar,
piperine on the pharmacokinetics of curcumin in animals and human volunteers, Curcumin in heart failure: a choice for complementary therapy? Pharmacol. Res.
Planta Med. 64 (1998) 353–356, https://doi.org/10.1055/s-2006-957450, 04. 131 (2018) 112–119, https://doi.org/10.1016/j.phrs.2018.03.009.
[9] B. Wahlström, G. Blennow, A study on the fate of curcumin in the rat, Acta [34] N. Venkatesan, Curcumin attenuation of acute adriamycin myocardial toxicity in
Pharmacol. Toxicol. 43 (2) (1978) 86–92, https://doi.org/10.1111/j.1600- rats, Br. J. Pharmacol. 124 (3) (1983) 425–427, https://doi.org/10.1038/sj.
0773.1978.tb02240.x. bjp.0701877.
[10] M. Araujo, L. Antunes, C. Takahashi, Protective effect of thiourea, a hydroxyl- [35] N. Venkatesan, Pulmonary protective effects of curcumin against paraquat
radical scavenger, on curcumin-induced chromosomal aberrations in an in vitro toxicity, PL21-28, Life Sci. 66 (2) (2000), https://doi.org/10.1016/s0024-3205
mammalian cell system, Teratog. Carcinog. Mutagen. 21 (2) (2001) 175–180, (99), 00576–00577.
https://doi.org/10.1002/1520-6866(2001)21:2%3C175:aid-tcm6%3E3.0.co;2-v. [36] N. Venkatesan, D. Punithavathi, V. Arumugam, Curcumin prevents adriamycin
[11] B. Aggarwal, Y. Takada, O. Oommen, From chemoprevention to chemotherapy: nephrotoxicity in rats, Br. J. Pharmacol. 129 (2) (2000) 231–234, https://doi.
common targets and common goals, Expert Opin. Investig. Drugs 13 (10) (2004) org/10.1038/sj.bjp.0703067.
1327–1338, https://doi.org/10.1517/13543784.13.10.1327. [37] N. Parsamanesh, M. Moossavi, A. Bahrami, A.E. Butler, A. Sahebkar, Therapeutic
[12] M.A. Mukundan, M.C. Chacko, V.V. Annapurna, K. Krishnaswamy, Effect of potential of curcumin in diabetic complications, Pharmacol. Res. 136 (2018)
turmeric and curcumin on BP-DNA adducts, Carcinogenesis 14 (3) (1993) 181–193, https://doi.org/10.1016/j.phrs.2018.09.012.
493–496, https://doi.org/10.1093/carcin/14.3.493. [38] M. Mutsuga, J.K. Chambersm, K. Uchida, M. Tei, T. Makibuchi, T. Mizorogi,
[13] S. Pal, T. Choudhuri, S. Chattopadhyay, A. Bhattacharya, G.K. Datta, T. Das, A. Takashima, H. Nakayama, Binding of curcumin to senile plaques and cerebral
G. Sa, Mechanisms of curcumin-induced apoptosis of Ehrlich’s ascites carcinoma amyloid angiopathy in the aged brain of various animals and to neurofibrillary
cells, Biochem. Biophys. Res. Commun. 288 (3) (2001) 658–665, https://doi.org/ tangles in Alzheimer’s brain, J. Vet. Med. Sci. 74 (2012) 51–57, https://doi.org/
10.1006/bbrc.2001.5823. 10.1292/jvms.11-0307.
[14] R.C. Srimal, B.N. Dhawan, Pharmacology of diferuloyl methane (curcumin), a [39] B. Ray, S. Bisht, A. Maitra, A. Maitra, D.K. Lahiri, Neuroprotective and
non-steroidal anti-inflammatory agent, J. Pharm. Pharmacol. 25 (6) (1973) neurorescue effects of a novel polymeric nanoparticle formulation of curcumin
447–452, https://doi.org/10.1111/j.2042-7158.1973.tb09131.x. (NanoCurc (TM)) in the neuronal cell culture and animal model: implications for
[15] M.K. Kim, G.J. Choi, H.S. Lee, Fungicidal property of Curcuma longa L. rhizome- Alzheimer’s disease, J. Alzheimers Dis. 23 (2011) 61–77, https://doi.org/
derived curcumin against phytopathogenic fungi in a greenhouse, J. Agric. Food 10.3233/jad-2010-101374.
Chem. 51 (6) (2003) 1578–1581, https://doi.org/10.1021/jf0210369. [40] R.B. Mythri, G. Harish, S.K. Dubey, K. Misra, M.M. Bharath, Glutamoyl diester of
[16] R. Kuttan, P. Bhanumathy, K. Nirmala, M.C. George, Potential anticancer activity the dietary polyphenol curcumin offers improved protection against
of turmeric (Curcuma longa), Canc. Lett. 29 (2) (1985) 197–202, https://doi.org/ peroxynitrite-mediated nitrosative stress and damage of brain mitochondria in
10.1016/0304-3835(85)90159-4. vitro: implications for Parkinson’s disease, Mol. Cell. Biochem. 347 (2011)
[17] S. Prasad, A. Tyagi, Curcumin and its analogues: a potential natural compound 135–143, https://doi.org/10.1007/s11010-010-0621-4.
against HIV infection and AIDS, Food & Funct 6 (11) (2015) 3412–3419, https:// [41] C. Lao, M. Demierre, V. Sondak, Targeting events in melanoma carcinogenesis for
doi.org/10.1039/c5fo00485c. the prevention of melanoma, Expert Rev. Anticancer Ther. 6 (11) (2006)
[18] R.C. Reddy, P.G. Vatsala, V.G. Keshamouni, G. Padmanaban, P.N. Rangarajan, 1559–1568, https://doi.org/10.1586/14737140.6.11.1559.
Curcumin for malaria therapy, Biochem. Biophys. Res. Commun. 326 (2) (2005) [42] C.D. Lao, M.T. Ruffin, D. Normolle, D.D. Heath, S.I. Murray, J.M. Bailey, M.
472–474, https://doi.org/10.1016/j.bbrc.2004.11.051. E. Boggs, J. Crowell, C.L. Rock, D.E. Brenner, Dose escalation of a curcuminoid
[19] C. Senft, M. Polacin, M. Priester, V. Seifert, D. Kögel, J. Weissenberger, The formulation. BMC Complement, Alternative Med. 6 (10) (2006), https://doi.org/
nontoxic natural compound Curcumin exerts anti-proliferative, anti-migratory, 10.1186/1472-6882-6-10.
and anti-invasive properties against malignant gliomas, BMC Canc. 10 (491) [43] R.A. Sharma, S.A. Euden, S.L. Platton, D.N. Cooke, A. Shafayat, H.R. Hewitt, T.
(2010), https://doi.org/10.1186/1471-2407-10-491. H. Marczylo, B. Morgan, D. Hemingway, S.M. Plummer, M. Pirmohamed, A.
[20] I. Jantan, S. Bukhari, N. Lajis, F. Abas, L. Wai, M. Jasamai, Effects of J. Gescher, W.P. Steward, Phase I clinical trial of oral curcumin: biomarkers of
-diarylpentanoid analogues of curcumin on chemiluminescence and chemotactic systemic activity and compliance, Clin. Canc. Res. 10 (20) (2004) 6847–6854,
activities of phagocytes, J. Pharm. Pharmacol. 64 (3) (2011) 404–412, https:// https://doi.org/10.1158/1078-0432.ccr-04-0744.
doi.org/10.1111/j.2042-7158.2011.01423.x. [44] A. Siviero, E. Gallo, V. Maggini, L. Gori, A. Mugelli, F. Firenzuoli, A. Vannacci,
[21] H. Hatcher, R. Planalp, J. Cho, F. Torti, S. Torti, Curcumin: from ancient medicine Curcumin, a golden spice with a low bioavailability, J. Herb. Med. 5 (2) (2015)
to current clinical trials, Cell. Mol. Life Sci. 65 (11) (2008) 1631–1652, https:// 57–70, https://doi.org/10.1016/j.hermed.2015.03.001.
doi.org/10.1007/s00018-008-7452-4. [45] U. Klickovic, D. Doberer, G. Gouya, S. Aschauer, S. Weisshaar, A. Storka,
[22] S. Khan, M. Imran, T.T. Butt, S.W.A. Shah, M. Sohail, A. Malik, S. Das, H.E. Thu, M. Bilban, M. Wolzt, Human pharmacokinetics of high dose oral curcumin and its
A. Adam, Z. Hussain, Curcumin based nanomedicines as efficient nanoplatform effect on heme oxygenase-1 expression in healthy male subjects, BioMed Res. Int.
for treatment of cancer: new developments in reversing cancer drug resistance, 2014 (2014) 1–7, https://doi.org/10.1155/2014/458592.
rapid internalization, and improved anticancer efficacy, Trends Food Sci. [46] S. Prasad, A. Tyagi, B. Aggarwal, Recent developments in delivery,
Technol. 80 (2018) 8–22, https://doi.org/10.1016/j.tifs.2018.07.026. bioavailability, absorption and metabolism of curcumin: the golden pigment from
[23] S.M. Khopde, K.I. Priyadarsini, P. Venkatesan, M.N. Rao, Free radical scavenging golden spice, Canc. Res. Treat. 46 (1) (2014) 2–18, https://doi.org/10.4143/
ability and antioxidant efficiency of curcumin and its substituted analogue, crt.2014.46.1.2.
Bhiophys. Chem. 80 (2) (1999) 85–91, https://doi.org/10.1016/s0301-4622(99) [47] H. Gupta, D. Bhandari, A. Sharma, Recent trends in oral drug delivery: a review,
00070-8. Recent Pat. Drug Deliv. Formul. 3 (2) (2009) 162–173, https://doi.org/10.2174/
[24] T. Masuda, T. Maekawa, K. Hidaka, H. Bando, Y. Takeda, H. Yamaguchi, 187221109788452267.
Chemical studies on antioxidant mechanism of curcumin: analysis of oxidative [48] B. Homayun, X. Lin, H. Choi, Challenges and recent progress in oral drug delivery
coupling products from curcumin and linoleate, J. Agric. Food Chem. 49 (5) systems for biopharmaceuticals, Pharmaceutics 11 (3) (2019) 129, https://doi.
(2001) 2539–2547, https://doi.org/10.1021/jf001442x. org/10.3390/pharmaceutics11030129.
[25] A.J. Ruby, G. Kuttan, K.D. Babu, K.N. Rajasekharan, R. Kuttan, Anti-tumour and [49] L.N. Dovigo, J.C. Carmello, C.A. de Souza Costa, C.E. Vergani, I.L. Brunetti, V.
antioxidant activity of natural curcuminoids, Canc. Lett. 94 (1) (1995) 79–83, S. Bagnato, A.C. Pavarina, Curcumin-mediated photodynamic inactivation of
https://doi.org/10.1016/0304-3835(95)03827-j. Candida albicans in a murine model of oral candidiasis, Med. Mycol. 51 (3)
[26] O. Sharma, Antioxidant activity of curcumin and related compounds, Biochem. (2013) 243–251, https://doi.org/10.3109/13693786.2012.714081.
Pharmacol. 5 (15) (1976) 1811–1812, https://doi.org/10.1016/0006-2952(76) [50] P. Lopez-Jornet, F. Camacho-Alonso, M.J. Jiménez-Torres, A. Orduña-Domingo,
90421-4. F. Gómez-Garcia, Topical curcumin for the healing of carbon dioxide laser skin
[27] Y. Sugiyama, S. Kawakishi, T. Osawa, Involvement of the β-diketone moiety in the wounds in mice, Photomed Laser Surg 29 (12) (2011) 809–814, https://doi.org/
antioxidative mechanism of tetrahydrocurcumin, Biochem. Pharmacol. 52 (4) 10.1089/pho.2011.3004.
(1996) 519–525, https://doi.org/10.1016/0006-2952(96)00302-4. [51] M.M. LoTempio, M.S. Veena, H.L. Steele, B. Ramamurthy, T.S. Ramalingam, A.
[28] J. Hong, M. Bose, J. Ju, J.-H. Ryu, X. Chen, S. Sang, M.-J. Lee, C.S. Yang, N. Cohen, R. Chakrabarti, E.S. Srivatsan, M.B. Wang, Curcumin suppresses
Modulation of arachidonic acid metabolism by curcumin and related β-diketone growth of head and neck squamous cell carcinoma, Clin. Canc. Res. 11 (19 Pt 1)
derivatives: effects on cytosolic phospholipase A 2, cyclooxygenases and 5- (2005) 6994–7002, https://doi.org/10.1158/1078-0432.CCR-05-0301.
lipoxygenase, Carcinogenesis 25 (9) (2004) 1671–1679, https://doi.org/ [52] J. Sun, Y. Zhao, J. Hu, Curcumin inhibits imiquimod-induced psoriasis-like
10.1093/carcin/bgh165. inflammation by inhibiting IL-1 beta and IL-6 production in mice, PloS One 8 (6)
[29] D. Rai, J.K. Singh, N. Roy, D. Panda, Curcumin inhibits FtsZ assembly: an (2013), e67078, https://doi.org/10.1371/journal.pone.0067078.
attractive mechanism for its antibacterial activity, Biochem. J. 410 (1) (2008) [53] S. Türker, E. Onur, Y. Ozer, Nasal route and drug delivery systems, Pharm. World
147–155, https://doi.org/10.1042/BJ20070891. Sci. 26 (3) (2004) 137–142, https://doi.org/10.1023/b:phar.0000026823.82950.
[30] S.Z. Moghadamtousi, H.A. Kadir, P. Hassandarvish, H. Tajik, S. Abubakar, ff.
K. Zandi, A review on antibacterial, antiviral, and antifungal activity of curcumin, [54] D. Paolino, A. Vero, D. Cosco, T.M.G. Pecora, S. Cianciolo, M. Fresta,
BioMed Res. Int. 2014 (186864) (2014), https://doi.org/10.1155/2014/186864. R. Pignatello, Improvement of oral bioavailability of curcumin upon
[31] M. Yang, U. Akbar, C. Mohan, Curcumin in autoimmune and rheumatic diseases, microencapsulation with methacrylic copolymers, Front. Pharmacol. 7 (485)
Nutrients 11 (5) (2019) 1004, https://doi.org/10.3390/nu11051004. (2016), https://doi.org/10.3389/fphar.2016.00485.
[32] V.S. Yadav, K.P. Mishra, D.P. Singh, S. Mehrotra, V.K. Singh, Immunomodulatory [55] A. Gantait, T. Barman, P.K. Mukherjee, Validated method for the estimation of
effects of curcumin, Immunopharmacol. Immunotoxicol. 27 (3) (2005) 485–497, curcumin in turmeric powder, Indian J. Tradit. Know 10 (2) (2001) 247–250. http
https://doi.org/10.1080/08923970500242244. ://nopr.niscair.res.in/handle/123456789/11499.
[56] G. Holder, J. Plummer, A. Ryan, The metabolism and excretion of curcumin (1,7-
bis-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) in the rat,

13
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

Xenobiotica 8 (12) (1978) 761–768, https://doi.org/10.3109/ [79] Y.-H. Wang, X.-M. Ke, C.-H. Zhang, R.-P. Yang, Absorption mechanism of three
00498257809069589. curcumin constituents through in situ intestinal perfusion method, Braz. J. Med.
[57] C. Ireson, S. Orr, D. Jones, R. Verschoyle, C. Lim, J. Luo, L. Howells, S. Plummer, Biol. Res. 50 (11) (2017) E6353, https://doi.org/10.1590/1414-431x20176353.
R. Jukes, M. Williams, W. Steward, Characterization of metabolites of the [80] P. Artursson, J. Karlsson, Correlation between oral drug absorption in humans
chemopreventive agent curcumin in human and rat hepatocytes and in the rat in and apparent drug permeability coefficients in human intestinal epithelial (Caco-
vivo, and evaluation of their ability to inhibit phorbol ester-induced 2) cells, Biochem. Biophys. Res. Commun. 175 (3) (1991) 880–885, https://doi.
prostaglandin E2 production, Canc. Res. 61 (3) (2001) 1058–1064. https:// org/10.1016/0006-291x(91)91647-u.
cancerres.aacrjournals.org/content/61/3/1058. [81] A. Parikh, K. Kathawala, Y. Song, X.-F. Zhou, S. Garg, Curcumin-loaded self-
[58] A.B. Kunnumakkara, P. Anand, B.B. Aggarwal, Curcumin inhibits proliferation, nanomicellizing solid dispersion system: part I: development, optimization,
invasion, angiogenesis and metastasis of different cancers through interaction characterization, and oral bioavailability. Drug Deliv, Transl. Res. 8 (5) (2018)
with multiple cell signaling proteins, Canc. Lett. 269 (2008) 199–225, https:// 1389–1405, https://doi.org/10.1007/s13346-018-0543-3.
doi.org/10.1016/j.canlet.2008.03.009. [82] Y. Sambuy, I. De Angelis, G. Ranaldi, M.L. Scarino, A. Stammati, F. Zucco, The
[59] B.T. Kurien, A. Singh, H. Matsumoto, R.H. Scofield, Improving the solubility and Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-
pharmacological efficacy of curcumin by heat treatment, Assay drug Dev. related factors on Caco-2 cell functional characteristics, Cell Biol. Toxicol. 21 (1)
Technol. 5 (4) (2007) 567–576, https://doi.org/10.1089/adt.2007.064. (2005) 1–26, https://doi.org/10.1007/s10565-005-0085-6.
[60] M.K. Modasiya, V.M. Patel, Studies on solubility of curcumin, Int. J. Pharm. Life [83] R.B. van Breemen, Y. Li, Caco-2 cell permeability assays to measure drug
Sci. 3 (3) (2012) 1490–1497. http://www.ijplsjournal.com/issues%20PDF%20fi absorption, Expert Opin. Drug Metab. Toxicol. 1 (2) (2005) 175–185, https://doi.
les/march%202012/1.pdf. org/10.1517/17425255.1.2.175.
[61] P. Murugan, L. Pari, Effect of tetrahydrocurcumin on plasma antioxidants in [84] M. Xue, Y. Cheng, L. Xu, L. Zhang, Study of the intestinal absorption
streptozotocin–nicotinamide experimental diabetes, J. Basic Clin. Physiol. characteristics of curcumin in vivo and in vitro, J. App. Pharm. 9 (3) (2017),
Pharmacol. 17 (4) (2006) 231–244, https://doi.org/10.1515/ https://doi.org/10.21065/1920-4159.1000246, 1000246.
jbcpp.2006.17.4.231. [85] Z. Zeng, Z. Shen, S. Zhai, J. Xu, H. Liang, Q. Shen, Q. Li, Transport of curcumin
[62] S. Perkins, R. Verschoyle, K. Hill, I. Parveen, M.D. Threadgill, R.A. Sharma, M. derivatives in caco-2 cell monolayers, Eur. J. Pharm. Biopharm. 117 (2017)
L. Williams, W.P. Steward, A.J. Gescher, Chemopreventive efficacy and 123–131, https://doi.org/10.1016/j.ejpb.2017.04.004.
pharmacokinetics of curcumin in the min/+ mouse, a model of familial [86] K. Ozeki, M. Kato, Y. Sakurai, M. Ishigai, T. Kudo, K. Ito, Evaluation of the
adenomatous polyposis, Canc. Epidemiol. Biomarkers Prev. 11 (6) (2002) appropriate time range for estimating the apparent permeability coefficient
535–540. https://cebp.aacrjournals.org/content/11/6/535.long. (Papp) in a transcellular transport study, Int. J. Pharm. 495 (2) (2015) 963–971,
[63] E. Pfeiffer, S.I. Hoehle, S.G. Walch, A. Riess, A.M. Sólyom, M. Metzler, https://doi.org/10.1016/j.ijpharm.2015.09.035.
Curcuminoids form reactive glucuronides in vitro, J. Agric. Food Chem. 55 (2) [87] R. Canaparo, N. Finnström, L. Serpe, A. Nordmark, E. Muntoni, M. Eandi, A. Rane,
(2007) 538–544, https://doi.org/10.1021/jf0623283. G. Zara, Expression of CYP3A isoforms and P-glycoprotein in human stomach,
[64] V. Ravindranath, N. Chandrasekhara, Metabolism of curcumin–studies with [3H] jejunum and ileum, Clin. Exp. Pharmacol. Physiol. 34 (11) (2007) 1138–1144,
curcumin, Toxicology 22 (4) (1981) 337–344, https://doi.org/10.1016/0300- https://doi.org/10.1111/j.1440-1681.2007.04691.x.
483x(81)90027-5. [88] G. Garcea, D. Jones, R. Singh, A. Dennison, P. Farmer, R.A. Sharma, W.
[65] S.K. Sandur, M.K. Pandey, B. Sung, K.S. Ahn, A. Murakami, G. Sethi, P. Limtrakul, P. Steward, A.J. Gescher, D.P. Berry, Detection of curcumin and its metabolites in
V. Badmaev, B.B. Aggarwal, Curcumin, demethoxycurcumin, hepatic tissue and portal blood of patients following oral administration, Br. J.
bisdemethoxycurcumin, tetrahydrocurcumin and turmerones differentially Canc. 90 (5) (2004) 1011–1015, https://doi.org/10.1038/sj.bjc.6601623.
regulate anti-inflammatory and anti-proliferative responses through a ROS- [89] S. Hoehle, E. Pfeiffer, M. Metzler, Glucuronidation of curcuminoids by human
independent mechanism, Carcinogenesis 28 (8) (2007) 1765–1773, https://doi. microsomal and recombinant UDP-glucuronosyltransferases, Mol. Nutr. Food Res.
org/10.1093/carcin/bgm123. 51 (8) (2007) 932–938, https://doi.org/10.1002/mnfr.200600283.
[66] I. Song, J. Cha, M. Choi, Characterization, in vivo and in vitro evaluation of solid [90] A. Hassaninasab, Y. Hashimoto, K. Tomita-Yokotani, M. Kobayashi, Discovery of
dispersion of curcumin containing d-α-Tocopheryl polyethylene glycol 1000 the curcumin metabolic pathway involving a unique enzyme in an intestinal
succinate and mannitol, Molecules 21 (10) (2016) 1386, https://doi.org/ microorganism, Proc. Natl. Acad. Sci. U.S.A. 108 (16) (2011) 6615–6620, https://
10.3390/molecules21101386. doi.org/10.1073/pnas.1016217108.
[67] K. Suresh, A. Nangia, Curcumin: pharmaceutical solids as a platform to improve [91] C. Ireson, D. Jones, S. Orr, M. Coughtrie, D. Boocock, M. Williams, P.B. Farmer,
solubility and bioavailability, CrystEngComm 20 (24) (2018) 3277–3296, W.P. Steward, A.J. Gescher, Metabolism of the cancer chemopreventive agent
https://doi.org/10.1039/C8CE00469B. curcumin in human and rat intestine, Canc. Epidemiol. Biomarkers Prev. 11 (1)
[68] H.H. Tønnesen, J. Karlsen, Studies on curcumin and curcuminoids.VI. Kinetics of (2002) 105–111. https://cebp.aacrjournals.org/content/11/1/105.long.
curcumin degradation in aqueous- solution, Z. Lebensm. Unters. Forsch. 180 [92] A. Asai, T. Miyazawa, Occurrence of orally administered curcuminoid as
(1985) 402–404, https://doi.org/10.1007/bf01027775. glucuronide and glucuronide/sulfate conjugates in rat plasma, Life Sci. 67 (23)
[69] Y.J. Wang, M.H. Pan, A.L. Cheng, L.I. Lin, Y.S. Ho, C.Y. Hsieh, J.K. Lin, Stability of (2000) 2785–2793, https://doi.org/10.1016/s0024-3205(00)00868-7.
curcumin in buffer solutions and characterization of its degradation products, [93] C. Atal, R. Dubey, J. Singh, Biochemical basis of enhanced drug bioavailability by
J. Pharmaceut. Biomed. Anal. 15 (1997) 1867–1876, https://doi.org/10.1016/ piperine: evidence that piperine is a potent inhibitor of drug metabolism,
s0731-7085(96)02024-9. J. Pharmacol. Exp. Therapeut. 232 (1) (1985) 258–262. http://jpet.aspetjournals.
[70] P. Jaisamut, R. Wiwattanapatapee, K. Wiwattanawongsa, Influence of natural org/content/232/1/258.long.
gum on curcumin supersaturation in gastrointestinal fluids, Int. J. Med. Health [94] X. Zeng, D. Cai, Q. Zeng, Z. Chen, G. Zhong, J. Zhuo, H. Gan, X. Huang, Z. Zhao,
Biomed. Bioeng. Pharm. Eng 7 (12) (2013) 906–909, https://doi.org/10.5281/ N. Yao, D. Huang, C. Zhang, D. Sun, Y. Chen, Selective reduction in the expression
zenodo.1089481. of UGTs and SULTs, a novel mechanism by which piperine enhances the
[71] G.N.L. Nascimento, E.V. Montalvão, T.A. Aversi-Ferreira, Study of the pepsin bioavailability of curcumin in rat, Biopharm Drug Dispos. 38 (1) (2017) 3–19,
enzymatic activity in in-vitro dissolution test of bromazepam tablets by UV/VIS https://doi.org/10.1002/bdd.2049.
spectrophotometry, J. Appl. Pharmaceut. Sci. 2 (10) (2012) 11–15, https://doi. [95] L.P. Volak, S. Ghirmai, J.R. Cashman, M.H. Court, Curcuminoids inhibit multiple
org/10.7324/JAPS.2012.21002. human cytochromes P450, UDP-glucuronosyltransferase, and sulfotransferase
[72] S. Pinnamaneni, F.A. Rinaldi, D.A. Jayawickrama, J. Li, M.V. Dali, Effect of enzymes, whereas piperine is a relatively selective CYP3A4 inhibitor, Drug
pepsin on maintaining the supersaturation of the HCl salt of a weakly basic drug: Metab. Dispos 36 (8) (2008) 1594–1605, https://doi.org/10.1124/
a case study, Pharmaceut. Dev. Technol. 21 (3) (2015) 311–320, https://doi.org/ dmd.108.020552.
10.3109/10837450.2014.1003653. [96] K. Grancharov, Z. Naydenova, S. Lozeva, E. Golovinsky, Natural and synthetic
[73] M.T. Chang, T.R. Tsai, C.Y. Lee, Y.S. Wei, Y.J. Chen, C.R. Chen, J.T. Tzen, inhibitors of UDP-glucuronosyltransferase, Pharmacol. Ther. 89 (2) (2001)
Elevating bioavailability of curcumin via encapsulation with a novel formulation 171–186, https://doi.org/10.1016/S0163-7258(00)00109-1.
of artificial oil bodies, J. Agric. Food Chem. 61 (40) (2013) 9666–9671, https:// [97] J.A. Williams, B.J. Ring, V.E. Cantrell, K. Campanale, D.R. Jones, S.D. Hall, S.
doi.org/10.1021/jf4019195. A. Wrighton, Differential modulation of UDP-glucuronosyltransferase 1A1
[74] A. Parikh, K. Kathawala, C.C. Tan, S. Garg, X. Zhou, Development of a novel oral (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1
delivery system of edaravone for enhancing bioavailability, Int. J. Pharm. 515 substrates and other compounds to human liver microsomes, Drug Metab. Dispos.
(1–2) (2016) 490–500, https://doi.org/10.1016/j.ijpharm.2016.10.052. 30 (11) (2002) 1266–1273, https://doi.org/10.1124/dmd.30.11.1266.
[75] D. Gao, H. Liu, J. Lin, Y. Wang, Y. Jiang, Characterization of drug permeability in [98] K.C. Lund, T. Pantuso, Combination effects of quercetin, resveratrol and curcumin
Caco-2 monolayers by mass spectrometry on a membrane-based microfluidic on in vitro intestinal absorption, J. Restor. Med 3 (1) (2014) 112–120, https://
device, Lab Chip 13 (5) (2013) 978–985, https://doi.org/10.1039/c2lc41215b. doi.org/10.14200/jrm.2014.3.0108.
[76] C. Righeschi, M.C. Bergonzi, B. Isacchi, C. Bazzicalupi, P. Gratteri, A. Bilia, [99] C. Cheung, N. Gibbons, D. Wayne Johnson, D. Nicol, Silibinin – a promising new
Enhanced curcumin permeability by SLN formulation: the PAMPA approach, LWT treatment for cancer, Anti Canc. Agents Med. Chem. 10 (3) (2010) 186–195,
- Food Sci. Technol. (Lebensmittel-Wissenschaft -Technol.) 66 (2016) 475–483, https://doi.org/10.2174/1871520611009030186.
https://doi.org/10.1016/j.lwt.2015.11.008. [100] A.E. Grill, B. Koniar, J. Panyam, Co-delivery of natural metabolic inhibitors in a
[77] D.A. Volpe, P.J. Faustino, A.B. Ciavarella, E.B. Asafu-Adjaye, C.D. Ellison, L.X. Yu, self-microemulsifying drug delivery system for improved oral bioavailability of
A.S. Hussain, Classification of drug permeability with a caco-2 cell monolayer curcumin. Drug Deliv, Transl. Res. 4 (4) (2014) 344–352, https://doi.org/
assay, Clin. Res. Regul. Aff. 4 (1) (2007) 39–47, https://doi.org/10.1080/ 10.1007/s13346-014-0199-6.
10601330701273669. [101] P.S. Jahagirdar, P.K. Gupta, S.P. Kulkarni, P.V. Devarajan, Polymeric curcumin
[78] B. Wahlang, Y. Pawar, A. Bansal, Identification of permeability-related hurdles in nanoparticles by a facile in situ method for macrophage targeted delivery, Bioeng.
oral delivery of curcumin using the Caco-2 cell model, Eur. J. Pharm. Biopharm. Transl. Med 4 (1) (2018) 141–151, https://doi.org/10.1002/btm2.10112.
77 (2) (2011) 275–282, https://doi.org/10.1016/j.ejpb.2010.12.006.

14
Z. Liu et al. Journal of Drug Delivery Science and Technology 60 (2020) 102082

[102] E. Merisko-Liversidge, G.G. Liversidge, E.R. Cooper, Nanosizing: a formulation [125] K. Wan, L. Sun, X. Hu, Z. Yan, Y. Zhang, X. Zhang, J. Zhang, Novel nanoemulsion
approach for poorly-water-soluble compounds, Eur. J. Pharmaceut. Sci. 18 (2) based lipid nanosystems for favorable in vitro and in vivo characteristics of
(2003) 113–120, https://doi.org/10.1016/s0928-0987(02)00251-8. curcumin, Int. J. Pharm. 504 (1–2) (2016) 80–88, https://doi.org/10.1016/j.
[103] R.H. Müller, S. Gohla, C.M. Keck, State of the art of nanocrystals – special ijpharm.2016.03.055.
features, production, nanotoxicology aspects and intracellular delivery, Eur. J. [126] M. Shukla, S. Jaiswal, A. Sharma, P.K. Srivastava, A. Arya, A.K. Dwivedi, J. Lal,
Pharm. Biopharm. 78 (1) (2011) 1–9, https://doi.org/10.1016/j. A combination of complexation and self-nanoemulsifying drug delivery system for
ejpb.2011.01.007. enhancing oral bioavailability and anticancer efficacy of curcumin, Drug Dev.
[104] J.U. Junghanns, R.H. Muller, Nanocrystal technology, drug delivery and clinical Ind. Pharm. 43 (5) (2016) 847–861, https://doi.org/10.1080/
applications, Int. J. Nanomed. 3 (3) (2008) 295–309, https://doi.org/10.2147/ 03639045.2016.1239732.
IJN.S595. [127] P. Constantinides, Lipid microemulsion for improving drug dissolution and oral
[105] S. Bisht, G. Feldmann, S. Soni, R. Ravi, C. Karikar, A. Maitra, A. Maitra, Polymeric absorption: physical and biopharmaceutical aspects, Pharm. Res. (N. Y.) 12
nanoparticle-encapsulated curcumin ("nanocurcumin"): a novel strategy for (1995) 1561–1572, https://doi.org/10.1023/a:1016268311867.
human cancer therapy, J. Nanobiotechnol. 5 (1) (2007) 3, https://doi.org/ [128] D. McClements, Nanoemulsions versus microemulsions: terminology, differences,
10.1186/1477-3155-5-3. and similarities, Soft Matter 8 (6) (2012) 1719–1729, https://doi.org/10.1039/
[106] L. Ensign, R. Cone, J. Hanes, Oral drug delivery with polymeric nanoparticles: the c2sm06903b.
gastrointestinal mucus barriers, Adv. Drug Del. Rev. 64 (6) (2012) 557–570, [129] D. Dhumal, P. Kothari, R. Kalhapure, K. Akamanchi, Self-microemulsifying drug
https://doi.org/10.1016/j.addr.2011.12.009. delivery system of curcumin with enhanced solubility and bioavailability using a
[107] R. Singh, J.W. Lillard Jr., Nanoparticle-based targeted drug delivery, Exp. Mol. new semi-synthetic bicephalous heterolipid: in vitro and in vivo evaluation, RSC
Pathol. 86 (3) (2009) 215–223, https://doi.org/10.1016/j.yexmp.2008.12.004. Adv. 5 (110) (2015) 90295–90306, https://doi.org/10.1039/C5RA18112G.
[108] L. Zhang, F.X. Gu, J.M. Chan, A.Z. Wang, R.S. Langer, O.C. Farokhzad, [130] A. Petchsomrit, N. Sermkaew, R. Wiwattanapatapee, Hydroxypropylmethyl
Nanoparticles in medicine: therapeutic applications and developments, Clin. cellulose-based sponges loaded self-microemulsifying curcumin: preparation,
Pharmacol. Ther. 83 (5) (2008) 761–769, https://doi.org/10.1038/sj. characterization, and in vivo oral absorption studies, J. Appl. Polym. Sci. 133 (6)
clpt.6100400. (2015), https://doi.org/10.1002/app.42966, 42966.
[109] S. Chaurasia, R. Patel, P. Chaubey, N. Kumar, G. Khan, B. Mishra, [131] W. Chiou, S. Riegelman, Pharmaceutical applications of solid dispersion systems,
Lipopolysaccharide based oral nanocarriers for the improvement of J. Pharmacol. Sci. 60 (9) (1971) 1281–1302, https://doi.org/10.1002/
bioavailability and anticancer efficacy of curcumin, Carbohydr. Polym. 130 jps.2600600902.
(2015) 9–17, https://doi.org/10.1016/j.carbpol.2015.04.062. [132] D. Craig, The mechanisms of drug release from solid dispersions in water-soluble
[110] Y. Wang, C. Wang, J. Zhao, Y. Ding, L. Li, A cost-effective method to prepare polymers, Int. J. Pharm. 231 (2) (2002) 131–144, https://doi.org/10.1016/
curcumin nanosuspensions with enhanced oral bioavailability, J. Colloid s0378-5173(01)00891-2.
Interface Sci. 485 (2017) 91–98, https://doi.org/10.1016/j.jcis.2016.09.003. [133] S. Kumar, S.K. Gupta, Pharmaceutical solid dispersion technology: a strategy to
[111] R. Govindaraju, R. Karki, J. Chandrasekharappa, M. Santhanam, A.K.K. Shankar, improve dissolution of poorly water-soluble drugs, Recent Pat. Drug Deliv.
H.K. Joshi, G. Divakar, Enhanced water dispersibility of curcumin encapsulated in Formul. 7 (2) (2013) 111–121, https://doi.org/10.2174/
alginate-polysorbate 80 nano particles and bioavailability in healthy human 18722113113079990009.
volunteers, Pharm. Nanotechnol. 7 (1) (2019) 39–56, https://doi.org/10.2174/ [134] T. Vasconcelos, B. Sarmento, P. Costa, Solid dispersions as strategy to improve
2211738507666190122121242. oral bioavailability of poor water soluble drugs, Drug Discov. Today Off. 12
[112] W.H. De Jong, P.J. Borm, Drug delivery and nanoparticles: applications and (23–24) (2007) 1068–1075, https://doi.org/10.1016/j.drudis.2007.09.005.
hazards, Int. J. Nanomed. 3 (2) (2008) 133–149, https://doi.org/10.2147/ijn. [135] J. Bevernage, T. Forier, J. Brouwers, J. Tack, P. Annaert, P. Augustijns, Excipient-
s596. mediated supersaturation stabilization in human intestinal fluids, Mol. Pharm. 8
[113] J. Baek, C. Cho, Surface modification of solid lipid nanoparticles for oral delivery (2011) 564–570, https://doi.org/10.1021/mp100377m.
of curcumin: improvement of bioavailability through enhanced cellular uptake, [136] P. Gao, A. Akrami, F. Alvarez, J. Hu, L. Li, C. Ma, S. Surapaneni, Characterization
and lymphatic uptake, Eur. J. Pharm. Biopharm. 117 (2017) 132–140, https:// and optimization of AMG-517 supersaturatable self-emulsifying drug delivery
doi.org/10.1016/j.ejpb.2017.04.013. system (S-SEDDS) for improved oral absorption, J. Pharmacol. Sci. 98 (2009)
[114] Z.L. Li, S.F. Peng, X. Chen, Y.Q. Zhu, L.Q. Zou, W. Liu, C.M. Liu, Pluronics 516–528, https://doi.org/10.1002/jps.21451.
modified liposomes for curcumin encapsulation: sustained release, stability and [137] D.A. Miller, J.C. DiNunzio, W. Yang, J.W. McGinity, R.O. Williams, Enhanced in
bioaccessibility, Food Res. Int. 108 (2018) 246–253, https://doi.org/10.1016/j. vivo absorption of itraconazole via stabilization of supersaturation following
foodres.2018.03.048. acidic-to-neutral pH transition, Drug Dev. Ind. Pharm. 34 (2008) 890–902,
[115] S. Peng, L. Zou, W. Liu, Z. Li, W. Liu, X. Hu, X. Chen, C. Liu, Hybrid liposomes https://doi.org/10.1080/03639040801929273.
composed of amphiphilic chitosan and phospholipid: preparation, stability and [138] A. Chuah, B. Jacob, Z. Jie, S. Ramesh, S. Mandal, J. Puthan, P. Deshpande,
bioavailability as a carrier for curcumin, Carbohydr. Polym. 156 (2017) 322–332, V. Vaidyanathan, R. Gelling, G. Patel, T. Das, S. Shreeram, Enhanced
https://doi.org/10.1016/j.carbpol.2016.09.060. bioavailability and bioefficacy of an amorphous solid dispersion of curcumin,
[116] F. Cuomo, M. Cofelice, F. Venditti, A. Ceglie, M. Miguel, B. Lindman, F. Lopez, In- Food Chem. 156 (2014) 227–233, https://doi.org/10.1016/j.
vitro digestion of curcumin loaded chitosan-coated liposomes, Colloids Surf. B foodchem.2014.01.108.
Biointerfaces 168 (2018) 29–34, https://doi.org/10.1016/j. [139] S. Onoue, H. Takahashi, Y. Kawabata, Y. Seto, J. Hatanaka, B. Timmermann,
colsurfb.2017.11.047. S. Yamada, Formulation design and photochemical studies on nanocrystal solid
[117] W. Xu, P. Ling, T. Zhang, Polymeric micelles, a promising drug delivery system to dispersion of curcumin with improved oral bioavailability, J. Pharmacol. Sci. 99
enhance bioavailability of poorly water-soluble drugs, J. Drug Deliv. 2013 (2013) (4) (2010) 1871–1881, https://doi.org/10.1002/jps.21964.
1–15, https://doi.org/10.1155/2013/340315. [140] S. Seo, H. Han, M. Chun, H. Choi, Preparation and pharmacokinetic evaluation of
[118] B. Haley, E. Frenkel, Nanoparticles for drug delivery in cancer treatment, Urol. curcumin solid dispersion using Solutol® HS15 as a carrier, Int. J. Pharm. 424
Oncol. 26 (2008) 57–64, https://doi.org/10.1016/j.urolonc.2007.03.015. (1–2) (2012) 18–25, https://doi.org/10.1016/j.ijpharm.2011.12.051.
[119] S. Patil, B. Choudhary, A. Rathore, K. Roy, K. Mahadik, Enhanced oral [141] S. Baghel, H. Cathcart, N. O’Reilly, Polymeric amorphous solid dispersions: a
bioavailability and anticancer activity of novel curcumin loaded mixed micelles review of amorphization, crystallization, stabilization, solid-state
in human lung cancer cells, Phytomedicine 22 (12) (2015) 1103–1111, https:// characterization, and aqueous solubilization of biopharmaceutical classification
doi.org/10.1016/j.phymed.2015.08.006. system class II drugs, J. Pharmacol. Sci. 105 (9) (2016) 2527–2544, https://doi.
[120] S. Peng, Z. Li, L. Zou, W. Liu, C. Liu, D.J. McClements, Enhancement of curcumin org/10.1016/j.xphs.2015.10.008.
bioavailability by encapsulation in sophorolipid-coated nanoparticles: an in vitro [142] Q. Zhang, N.E. Polyakov, Y.S. Chistyachenko, M.V. Khvostov, T.S. Frolova, T.
and in vivo study, J. Agric. Food Chem. 66 (6) (2018) 1488–1497, https://doi. G. Tolstikova, A.V. Dushkin, W. Su, Preparation of curcumin self-micelle solid
org/10.1021/acs.jafc.7b05478. dispersion with enhanced bioavailability and cytotoxic activity by
[121] C. Schiborr, A. Kocher, D. Behnam, J. Jandasek, S. Toelstede, J. Frank, The oral mechanochemistry, Drug Deliv. 25 (1) (2018) 198–209, https://doi.org/10.1080/
bioavailability of curcumin from micronized powder and liquid micelles is 10717544.2017.1422298.
significantly increased in healthy humans and differs between sexes, Mol. Nutr. [143] C.C.C. Teixeira, L.M. Mendonça, M.M. Bergamaschi, R.H.C. Queiroz, G.E.
Food Res. 58 (3) (2014) 516–527, https://doi.org/10.1002/mnfr.201300724. P. Souza, L.M.G. Antunes, L.A.P. Freitas, Microparticles containing curcumin solid
[122] J. Wang, W. Ma, P. Tu, The mechanism of self-assembled mixed micelles in dispersion: stability, bioavailability and anti-inflammatory activity, AAPS
improving curcumin oral absorption: in vitro and in vivo, Colloids Surf. B: PharmSciTech 17 (2) (2015) 252–261, https://doi.org/10.1208/s12249-015-
Biointerfaces 133 (2015) 108–119, https://doi.org/10.1016/j. 0337-6.
colsurfb.2015.05.056. [144] R. Wang, J. Han, A. Jiang, R. Huang, T. Fu, L. Wang, Q. Zheng, W. Li, J. Li,
[123] K. Gurpreet, S. Singh, Review of nanoemulsion formulation and characterization Involvement of metabolism-permeability in enhancing the oral bioavailability of
techniques, Indian J. Pharm. Sci. 80 (5) (2018) 781–789, https://doi.org/ curcumin in excipient-free solid dispersions co-formed with piperine, Int. J.
10.4172/pharmaceutical-sciences.1000422. Pharm. 561 (2019) 9–18, https://doi.org/10.1016/j.ijpharm.2019.02.027.
[124] R. Vecchione, V. Quagliariello, D. Calabria, V. Calcagno, E. De Luca, R.V. Iaffaioli, [145] A. Parikh, K. Kathawala, J. Li, C. Chen, Z. Shan, X. Cao, X.-F. Zhou, S. Garg,
P.A. Netti, Curcumin bioavailability from oil in water nano-emulsions: in vitro Curcumin-loaded self-nanomicellizing solid dispersion system: part II: in vivo
and in vivo study on the dimensional, compositional and interactional safety and efficacy assessment against behavior deficit in Alzheimer disease. Drug
dependence, J. Contr. Release 233 (2016) 88–100, https://doi.org/10.1016/j. Deliv, Transl. Res. 8 (5) (2018) 1406–1420, https://doi.org/10.1007/s13346-
jconrel.2016.05.004. 018-0570-0.

15

You might also like