|
beyond reason
진정한 암의 완치는 '기쁨, 감사, 축복, 내면의 평화' 540이상 치유의 에너지장에서 일어난다
Review
Possibilities of Fucoidan Utilization in theDevelopment of Pharmaceutical Dosage Forms
Aleksandra Citkowska, Marta Szekalska and KatarzynaWinnicka *
Department of Pharmaceutical Technology, Medical University of Białystok, Mickiewicza 2c,
15-222 Białystok, Poland
* Correspondence: kwin@umb.edu.pl; Tel.: +48-85-748-5616
Received: 11 July 2019; Accepted: 2 August 2019; Published: 5 August 2019
Abstract:
Fucoidan is a polysaccharide built from L-fucose molecules. The main source of this
polysaccharide is the extracellular matrix of brownseaweed ( Phaeophyta), but it can be also isolated from invertebrates such as sea urchins ( Echinoidea) and sea cucumbers ( Holothuroidea). Interest in fucoidan is related to its broad biological activity, including possible antioxidant, anti-inflammatory, anti fungal, antiviral or antithrombotic ects. The potential application of fucoidan in the pharmaceutical technology is also due to its ionic nature. The negative charge of the molecule results from the presence of sulfate residues in the C-2 and C-4 positions, occasionally in C-3, allowing the formation of complexes with other oppositely charged molecules.
Fucoidan is non-toxic, biodegradable and biocompatible compound approved by Food and Drug Administration (FDA) as Generally Recognized As Safe (GRAS) category as food ingredient. Fucoidan plays an important role in the pharmaceutical technology, so in this work aspects concerning its pharmaceutical characteristics and designing of various dosage forms (nanoparticles, liposomes, microparticles, and semisolid formulations) with fucoidan itself and with its combinations with other polymers or components that give a positive charge were reviewed. Advantages and limitations of fucoidan utilization in the pharmaceutical technology were also discussed.
Keywords: polysaccharide; marine-derived; fucoidan; pharmaceutical formulations; multifunctional
polymer; fucospheres
1. Introduction
Fucoidan belongs to the large group of sulfated, rich in l-fucose polysaccharides, which was
first found by Kylin in 1913 [1]. The main source of fucoidan are marine brown algae ( Phaeophyta:
Laminariaceae, Fucaceae, Chordariaceae, Alariaceae), but it has been also isolated from marine invertebrates such as sea cucumber ( Holothuroidea: Stichopodidae, Holothuriidae), from sea urchins eggs ( Echinoidea: Strongylocentrotidae, Arbaciidae) [2], and from seagrasses ( Cymodoceaceae) [3]. The chemical structure and molecular weight of fucoidan are various in dependence of the species from which it is extracted, growth environment, season of harvesting and the method of extraction used [2,3].
Fucoidans isolated from the majority of brown algae are branched and have a backbone of alternating (1!3)- and (1!4)-linked -l-fucose residues. Sometimes fucose molecules are connected by 1!2 linkage
(Figure 1) [3,4]. Studies demonstrated that the most of echinoderms and some families of brown algae
( Laminariaceae and Chordariaceae) are the source of fucoidan with linear chain built of (1!3)-linked
-l-fucose residues [3–5]. The negative charge of this biopolymer results from the presence of sulfate
groups, which are mainly substituted on C-2 and C-4 and occasionally on C-3 positions [2,4]. Besides
fucose, fucoidan may contain other sugars, for example: xylose, arabinose, rhamnose, glucose, galactose
and uronic acid or even protein and acetyl groups. Both molecular weight (in the range between 10
and 2000 kDa), as well as the varied percentage shared in sugar and non-sugar components makes the
Mar. Drugs 2019, 17, 458; doi:10.3390/md17080458 www.mdpi.com/journal/marinedrugs
Mar. Drugs 2019, 17, 458 2 of 20
analysis of fucoidan structure dicult [3,4]. Furthermore, the structure of fucoidan depends on the
species of algae, but it can be dierent even for the same species thus the term “fucoidan” does not refer
to one specific structure, but includes a diverse group of sulfated polysaccharides containing fucose.
Mar. Drugs 2019, 17, x 2 of 21
structure of fucoidan depends on the species of algae, but it can be different even for the same species
thus the term “fucoidan” does not refer to one specific structure, but includes a diverse group of
sulfated polysaccharides containing fucose.
Figure 1. Scheme of the fucoidan structure; R‐ carbohydrate substituents: xylose, arabinose,
rhamnose, glucose, galactose, or uronic acid and non‐carbohydrate substituents: sulfate or acetate
residues.
The process of fucoidan extraction consists of several stages. The first step involves cleaning the
algae, drying and grinding it or cutting fresh macroalgae into pieces and leaving in the dark, damp
place to get exudate. Then different solvents (for example acetone, ethanol, mixture of acetone and
ethanol or mixture of methanol, chloroform with water) are used to remove lipids, phenols and
proteins. Fucoidan could be extracted by traditional solvent extraction (with water, ethanol or diluted
acid) or using modern methods such as ultrasound‐assisted extraction (UAE), microwave‐assisted
extraction (MAE) or enzyme‐assisted extraction (EAE) [3,6]. New technique used to obtain fucoidan
is also an extraction with 0.5% ethylenediaminetetraacetic acid (EDTA) at 70 °C. The unquestionable
advantage of this procedure is that EDTA enables simultaneous removal of pigments and fucoidan
extraction with high yield [7]. Regardless of the method used for extraction to obtain a specific
polysaccharide, the next process is purification, most often by applying chromatographic methods or
using molecular cut off membranes [6]. The wide interest of fucoidan in medicine and pharmacy is
increasing due to its broad spectrum of biological activities. The bioactivity of fucoidan depends on
several factors, predominantly on the content of sulfate groups and the molecular weight of the
polysaccharide [3,4,8]. It was found that some fucoidans might exhibit antioxidant [9], anticoagulant
[10], antibacterial [11,12], antifungal, antileishmanial [12], anti‐inflammatory, and
immunomodulatory [13] activity. Positive fucoidan impact on the treatment of patients with cancer
diseases by improving the effectiveness of chemotherapy and reducing its side effects was also
reported [2,3,14,15].
Despite many publications about fucoidan multidirectional biological activity, and many
scientific reports on its use in drug delivery, there are still no registered drug products with fucoidan.
It is currently available only in cosmetics, functional foods and dietary supplements as a regenerative,
anti‐inflammatory, and anti‐cancer factor for patients with immune‐compromised, musculoskeletal,
cardiovascular, or gut diseases. However, fucoidan as synergistic anticancer agent has been subjected
to clinical examination. It was observed that co‐administration of fucoidan resulted in no significant
Figure 1. Scheme of the fucoidan structure; R- carbohydrate substituents: xylose, arabinose, rhamnose,
glucose, galactose, or uronic acid and non-carbohydrate substituents: sulfate or acetate residues.
The process of fucoidan extraction consists of several stages. The first step involves cleaning the
algae, drying and grinding it or cutting fresh macroalgae into pieces and leaving in the dark, damp
place to get exudate. Then dierent solvents (for example acetone, ethanol, mixture of acetone and
ethanol or mixture of methanol, chloroform with water) are used to remove lipids, phenols and proteins.
Fucoidan could be extracted by traditional solvent extraction (with water, ethanol or diluted acid) or
using modern methods such as ultrasound-assisted extraction (UAE), microwave-assisted extraction
(MAE) or enzyme-assisted extraction (EAE) [3,6]. New technique used to obtain fucoidan is also an
extraction with 0.5% ethylenediaminetetraacetic acid (EDTA) at 70 C. The unquestionable advantage
of this procedure is that EDTA enables simultaneous removal of pigments and fucoidan extraction
with high yield [7]. Regardless of the method used for extraction to obtain a specific polysaccharide,
the next process is purification, most often by applying chromatographic methods or using molecular
cut o membranes [6]. The wide interest of fucoidan in medicine and pharmacy is increasing due
to its broad spectrum of biological activities. The bioactivity of fucoidan depends on several factors,
predominantly on the content of sulfate groups and the molecular weight of the polysaccharide [3,4,8].
It was found that some fucoidans might exhibit antioxidant [9], anticoagulant [10], antibacterial [11,12],
antifungal, antileishmanial [12], anti-inflammatory, and immunomodulatory [13] activity. Positive
fucoidan impact on the treatment of patients with cancer diseases by improving the eectiveness of
chemotherapy and reducing its side eects was also reported [2,3,14,15].
Despite many publications about fucoidan multidirectional biological activity, and many scientific
reports on its use in drug delivery, there are still no registered drug products with fucoidan. It is
currently available only in cosmetics, functional foods and dietary supplements as a regenerative,
anti-inflammatory, and anti-cancer factor for patients with immune-compromised, musculoskeletal,
cardiovascular, or gut diseases. However, fucoidan as synergistic anticancer agent has been subjected
to clinical examination. It was observed that co-administration of fucoidan resulted in no significant
impact on plasma concentrations of anti-cancer drugs, but might play a role in reducing side eects and
Mar. Drugs 2019, 17, 458 3 of 20
in enhancing the therapeutic eects of conventional anti-cancer therapies [16,17]. The eect of oligo
fucoidan in patients with non-small cell lung cancer treated with platinum-based chemotherapy has
been tested in III-IV stage of clinical trials [18]. Fucoidan was also under clinical evaluation in patients
with metastatic colorectal cancer treated with folinic acid, 5-fluorouracil, irinotecan, and bevacizumab.
It was reported that fucoidan led to alleviate side eects of anti-cancer chemotherapy [19]. Fucoidan
impact on the metabolism of fatty liver and liver fibrosis was also examined, but results have not been
published yet [20].
2. Pharmaceutical Features of Fucoidan
Fucoidan is a pale brown or yellow powder with hygroscopic properties. It easily dissolves in
water but is not soluble in organic solvents. Fucoidan solutions are not highly viscous, so it is not
applied as thickening agent [21–23]. The viscosity of aqueous solutions of fucoidan is low and depends
on many factors, including molecular weight, concentration, number of sulfate groups, degree of
branching of the molecule, as well as temperature and pH [24].
In order to determine the basic properties of fucoidan obtained from Laminaria japonica (Weihai
Century Biocom Seaweed Co., Ltd, Weihai, China), we examined its aqueous solutions (Table 1). The
molecular weight of the tested fucoidan was 10.5 kDa. Both the content of fucose and sulphate groups
were classified at a level higher than 27%. It was observed that with the increasing concentration
of polysaccharide, the pH values of the solutions decreased (5.23 and 4.45 for 1% and 30% solution,
respectively), while their viscosity was increasing and reached a value of 507 mPas for 30% solution,
and no gelation was noted.
Similarly, Jae-Geun et al. have shown that water solutions of fucoidan from Laminaria religiosa,
Undaria pinnatifida, Hizikia fusiforme, and Sargassum fulvellum possessed low apparent viscosity and
were characterized by pseudoplastic behavior [21].
Viscosity of fucoidan is also influenced by algae species, presence of ions, or additional molecules.
Comparing fucoidan solutions from Saccharina longicruris, Ascophyllum nodosum, and Fucus vesiculosus, the highest viscosity was observed when fucoidan isolated from Fucus vesiculosus was applied [25].
Viscosity of fucoidan solutions obtained from Cladosiphom okamuranus increased linearly with an
increase of polymer concentration (up to 2%), and with the addition of sodium chloride, calcium
chloride and sugar. It was stable at pH range from 5.8 to 9.5, indicating that fucoidan molecules are
stable under acidic and alkaline conditions [22]. Contrary to other polysacharides, fucoidan does not
show gelation ability [4,24,26], but upon fucoidan mixing with polymers with opposite net charge,
based on electrostatic interactions, gels might be created.
It should be also emphasized that due to the ability to interact with growth factors, macrophages,
cytokines and P-selectin, inhibition of the P-glycoprotein pump and -glucosidase, as well as to open
tight junctions in intestinal Caco-2 cell monolayer, fucoidan seems to be a valuable adjuvant in the
pharmaceutical technology [2–5]. These properties allow not only to protect the drug substance, but
Mar. Drugs 2019, 17, 458 4 of 20 also to strengthen drug activity, and they are described in more detail in further subsections devoted to
specific formulations.
3. Toxicity of Fucoidan
Fucoidan utilizing in the pharmacy and biomedicine is possible because it is a non-toxic,
biodegradable, and biocompatible compound [4,24]. Fucoidans from Undaria pinnatifida and Fucus
vesiculosus were approved in the USA by Food and Drug Administration (FDA) as Generally
Recognized As Safe (GRAS) category as food ingredients at levels up to 250 mg/day [27]. In Europe
preparations containing Fucus vesiculosus are registered in Austria, Belgium, France, Poland, Spain,
and the United Kingdom [28].
No changes occurred in rats receiving fucoidan from Laminaria japonica for 7 days, even at a dose
of 4000 mg/kg body weight (b.w.). Subchronic toxicity studies conducted over 6 months have reported
that no significant adverse eects were observed when fucoidan was orally administered to animals at
the dose of 300 mg/kg b.w. per day, but when the dose was increased to 900 and 2500 mg/kg b.w. per day, clotting time significantly increased [29]. Nevertheless, low molecular weight fucoidan (LMWF)
from Laminaria japonica did not lead to changes in b.w., water and food consumption, or hematological
parameters after 28 days oral administration, even when the dose was 2000 mg/kg b.w. per day.
Dierences were observed only in two biochemical parameters—creatinine (CRE) and triglyceride
(TG) levels were significantly lower than in rats receiving 0.9% saline solution. In histopathological
examination of organs, including brain, heart, kidneys and liver, no discrepancies were observed
between the control group and fucoidan treated rats [30]. However, when the source of fucoidan
was Cladosiphon okamuranus, the dose that did not cause significant changes was 600 mg/kg b.w. per
day and the anticoagulant activity of fucoidan was observed at higher doses (1200 mg/kg b.w. per
day) [31]. No dierences were observed in nutrition, b.w., hematological and biochemical parameters,
as well as in organs subjected to necroscopy and microscopic examination in rats receiving up to
1000 mg/kg b.w. per day fucoidan from Undaria pinnatifida. Increasing the dose to 2000 mg/kg b.w.
per day caused significant changes in mean corpuscular hemoglobin concentration (MCHC), alanine
transaminase (ALT), TG, and high-density lipoprotein (HLD) levels [32]. Observed dierences between
the above-described studies indicate that the potential toxicity of fucoidans depends on the species
from which it is obtained, as well as fucoidan molecular weight.
The safety of oral ingestion of fucoidan has also been studied in humans. Twenty healthy, adult
volunteers received 4.05 g per day of mozuku fucoidan (isolated from Cladosiphon okamuranus) for
two weeks. The blood samples showed a significant decrease in total cholesterol and low-density
lipoprotein cholesterol. In turn, the concentration of chloride ions increased significantly. There were
no changes in the parameters describing the liver and kidneys [33]. Similarly, it was shown in patients
with osteoarthritis that application of fucoidan from Fucus vesiculosus at the dose 300 mg or from Fucus vesiculosus, Macrocystis pyrifera, and Laminaria japonica at 100 mg and 1000 mg did not aect the blood tests results or these changes were clinically negligible [34,35].
Safety of fucoidan was also evaluated at subcutaneous administration. Fucoidan from Fucus
vesiculosus and Laminaria japonica (molecular weight 10–300 kDa) administered subcutaneously and
orally did not cause negative changes in dogs with hemophilia A. Moreover, it also led to improvement
of their hemostasis [36]. Similar conclusions about safety of fucoidan have been drawn from a study
conducted on horses. The solution containing 2500 mg of fucoidan administered intraperitoneally
during abdominal surgery did not have a negative impact on the results of laboratory tests. Most of
the parameters did not dier between the test and the control groups, and the observed dierences
in the amount of leukocytes, neutrophils, antithrombin III, value of hematocrit, or prothrombin time
were within the normal range [37].
Furthermore, the genotoxicity of high and low molecular weight fucoidan was also investigated
with using both in vitro and in vivo model. In bacterial reverse mutation and chromosome aberration
tests fucoidan from Undaria pinnatifida (regardless of the molecular weight) did not show toxic eect
Mar. Drugs 2019, 17, 458 5 of 20
in concentrations 5000 g/plate and 5000 g/mL, respectively. Moreover, in micronucleus assay
conducted in ICR mice, fucoidan at dosages up to 2000 mg/kg b.w. per day did not display evidence
for genotoxicity [38,39].
4. Fucoidan Application in the Pharmaceutical Technology
Fucoidan plays an important role in the design of various dosage forms (Figure 2), especially
nanoparticles and microparticles, films, or hydrogels [4,5,24]. The wide application of fucoidan in
the pharmaceutical technology is due to its ionic nature. Negatively charged polysaccharide allows
the formation of complexes with other, oppositely charged molecules. Polyelectrolyte complexation
is the most commonly used technique for obtaining particles utilizing fucoidan. Other methods are
coacervation, ionic cross-linking, self-assembly, and spray-drying [2]. Fucoidan-based particles served
as carriers of various substances, including drugs (ciprofloxacin [40], gentamicin [41], doxorubicin [42],
isoniazid and rifabutin [43]), growth factors (basic fibroblast growth factor—bFGF [44], proteins (bovine
serum albumin [45], -lactoglobulin [46]), and genes [47]. Interestingly, fucoidan is used not only as an
excipient responsible for drug delivery, but also as a proper substance with therapeutic eects [12,48].
Mar. Drugs 2019, 17, x 5 of 21
conducted in ICR mice, fucoidan at dosages up to 2000 mg/kg b.w. per day did not display evidence
for genotoxicity [38,39].
4. Fucoidan Application in the Pharmaceutical Technology
Fucoidan plays an important role in the design of various dosage forms (Figure 2), especially
nanoparticles and microparticles, films, or hydrogels [4,5,24]. The wide application of fucoidan in the
pharmaceutical technology is due to its ionic nature. Negatively charged polysaccharide allows the
formation of complexes with other, oppositely charged molecules. Polyelectrolyte complexation is
the most commonly used technique for obtaining particles utilizing fucoidan. Other methods are
coacervation, ionic cross‐linking, self‐assembly, and spray‐drying [2]. Fucoidan‐based particles
served as carriers of various substances, including drugs (ciprofloxacin [40], gentamicin [41],
doxorubicin [42], isoniazid and rifabutin [43]), growth factors (basic fibroblast growth factor—bFGF
[44], proteins (bovine serum albumin [45], β‐lactoglobulin [46]), and genes [47]. Interestingly,
fucoidan is used not only as an excipient responsible for drug delivery, but also as a proper substance
with therapeutic effects [12,48].
Figure 2. Schematic presentation of pharmaceutical dosage forms designed with fucoidan
utilization.
4.1. Nanoparticles
Nanoparticles are usually spherical particles with diameters from 10 to 1000 nm. Depending on
the composition and the method of locating active substance in the nanoparticle, nanocapsules and
nanospheres can be distinguished. In nanospheres the drug is homogenously dissolved or suspended
in a polymer matrix. In turn, nanocapsules are the systems in which the drug is enclosed in a reservoir
surrounded by a polymer membrane. Nanoparticles are valuable multicompartment carriers in
modern pharmaceutical technology as they provide protection of the drug substance against
chemical and enzymatic degradation, and enable controlled or targeted drug release. As a
consequence, lower concentration of active substance is required to achieve a therapeutic effect,
which contributes to the reduction of side effects [4]. Examples of fucoidan‐based nanoparticles are
presented in Table 2. To obtain nanoparticles with fucoidan utilization, self‐assembly, and ionotropic
cross‐linking is commonly used. Self‐assembly method is a process in which participating molecules,
as a result of interactions between them, organize themselves into specific structures without
introducing additional elements [49]. The ionotropic cross‐linking is based on the interaction of the
negatively charged fucoidan with the positively charged polymer molecule (e.g. chitosan) [50].
Figure 2. Schematic presentation of pharmaceutical dosage forms designed with fucoidan utilization.
4.1. Nanoparticles
Nanoparticles are usually spherical particles with diameters from 10 to 1000 nm. Depending
on the composition and the method of locating active substance in the nanoparticle, nanocapsules
and nanospheres can be distinguished. In nanospheres the drug is homogenously dissolved or
suspended in a polymer matrix. In turn, nanocapsules are the systems in which the drug is enclosed
in a reservoir surrounded by a polymer membrane. Nanoparticles are valuable multicompartment
carriers in modern pharmaceutical technology as they provide protection of the drug substance
against chemical and enzymatic degradation, and enable controlled or targeted drug release. As a
consequence, lower concentration of active substance is required to achieve a therapeutic eect, which
contributes to the reduction of side eects [4]. Examples of fucoidan-based nanoparticles are presented
in Table 2. To obtain nanoparticles with fucoidan utilization, self-assembly, and ionotropic cross-linking
is commonly used. Self-assembly method is a process in which participating molecules, as a result of
interactions between them, organize themselves into specific structures without introducing additional
elements [49]. The ionotropic cross-linking is based on the interaction of the negatively charged
fucoidan with the positively charged polymer molecule (e.g. chitosan) [50].
Mar. Drugs 2019, 17, 458 6 of 20
Table 2. Characteristic of selected fucoidan-based nanoparticles.
Fucoidan
(Source/Modification/Molecular
Weight)
Copolymer/Positive
Charge Donor Drug Method of Obtaining Application Route of
Administration Ref.
Acetylated fucoidan
(Fucus vesiculosus) - Doxorubicin Self-assembly and
dialysis
Anticancer therapy
and immunotherapy NA1 51
Fucoidan
(Laminaria japonica, 80 kDa) Protamine Doxorubicin Self-assembly Anticancer therapy Intravenous 52
Fucoidan (Fucus vesiculosus) Polyethyleneimine Doxorubicin Polyelectrolyte
complexation method Anticancer therapy Intravenous 53
Fucoidan (Fucus vesiculosus) Gold nanoparticles Doxorubicin Electrostatic
physisorption Anticancer therapy Ocular 42
Fucoidan (Fucus vesiculosus) Polyallylamine
hydrochloride Copper sulfide Layer-by-layer Anticancer therapy Intratumoral 54
Fucoidan (200–400 kDa) Polyallyamine
hydrochloride Methotrexate Self-assembly Anticancer therapy NA1 55
Fucoidan (Fucus vesiculosus) Chitosan - Coacervation Thrombolytic therapy Oral 56
Fucoidan
(Fucus vesiculosus, 50–190 kDa) Chitosan Methotrexate Self-assembly Skin inflammation Topical (ear skin) 57
Fucoidan Chitosan Curcumin Self-assembly Anticancer therapy Oral 58
Fucoidan (Fucus vesiculosus) O-carboxymethyl chitosan Curcumin Ionotropic crosslinking Penetration enhancer Oral 59
Thiolated fucoidan
(THL-fucoidan) Arginine-modified chitosan Dextran/rhodamine/
curcumin Self-assembly NA1 Oral 60
Fucoidan (Fucus vesiculosus) Chitosan Gentamicin Self-assembly Pulmonary diseases Pulmonary 41
Fucoidan (Fucus vesiculosus) Chitosan Gentamicin Ionotropic crosslinking Pulmonary diseases Pulmonary 50
Fucoidan (Fucus vesiculosus) Chitosan Silver nitrate Self-assembly Antibacterial and
anticancer therapy NA1 61
Fucoidan (20–200 kDa) TPP crosslinked chitosan Ciprofloxacin Self-assembly Infections of
Salmonella NA1 40
Fucoidan
(Fucus vesiculosus, 57.26 kDa) Chitosan Poly-l-lysine Layer-by-layer Antibacterial therapy NA1 62
Fucoidan
(Fucus vesiculosus, 5–50 kDa) Trimethyl chitosan Insulin Self-assembly Diabetes Oral 63
Fucoidan
(Fucus vesiculosus, 80 kDa) Chitosan Basic fibroblast
growth factor Ionotropic crosslinking Neurite extension Nerve tissue 44
Mar. Drugs 2019, 17, 458 7 of 20
Table 2. Cont.
Fucoidan
(Source/Modification/Molecular
Weight)
Copolymer/Positive
Charge Donor Drug Method of Obtaining Application Route of
Administration Ref.
Fucoidan (104 kDa) Isobutylcyanoacrylate Recombinant tissue
plasminogen activator
Redox radical emulsion
polymerization Thrombolytic therapy Retro-orbital
(C57BL/6 mice) 64
Fucoidan (Sargassum cymosum) Isobutylcyanoacrylate -
Anionic emulsion
polymerization and
redox radical emulsion
polymerization
Immunotherapy NA1 65
Fucoidan
Poly(lactide-co-glycolide)
and poly-l-ornitine
(core-shell)
- Layer-by-layer Anticancer therapy NA1 66
Fucoidan (Fucus vesiculosus,
20–200 kDa) - Cisplatin Self-assembly Anticancer therapy
and immunotherapy
Colonic drug
delivery system 67
Fucoidan (Spatoglossum
schr˝oederi, 21 kDa) Hexadecylamine - Self-assembly Anticancer therapy NA1 68
1NA—No data available.
Mar. Drugs 2019, 17, 458 8 of 20
As shown in Table 2, various nanoparticles were tested to determine fucoidan potential in
anticancer drug delivery. As model antitumor drugs, doxorubicin (DOX), copper sulfide (CuS),
methotrexate (MTX), curcumin (CUR), silver nitrate (AgNO3), and cisplatin (CSN) were studied.
In 2013, Lee et al. developed self-organized nanoparticles with acetylated fucoidan from Fucus
vesiculosus. DOX was introduced into the nanoparticles through dialysis and it was released according
to the first order kinetics. Fucoidan was tested as a drug carrier due to its immunomodulatory
properties, ability to produce anticancer cytokines and drug eux pump inhibition [51]. In turn, Lu et
al. created nanoparticles by applying electrostatic interactions between the negatively charged fucoidan
and the cationic peptide–protamine. The self-assembled complex was stable at pH 7.4 (corresponding
to blood pH). When pH decreased to 4.5 (tumor cell pH) or 1.5 (stomach pH), electrostatic interactions
became weaker, which led to increased diameter of nanoparticles and release of more than 90% of
anticancer agent. The release of a smaller amount of DOX in the blood prevents side eects and
increases its concentration in tumor cells. This pH-dependent release profile makes intravenous
injection the best route for administration of these nanoparticles. Furthermore, using fucoidan that
possesses the ability to interact with P-selectin present in the breast cancer cells (MDA-MB-231),
resulted in the improved cell internalization and better inhibitory eect of designed nanoparticles
than free DOX [52]. The polyelectrolyte complexing method (PEC) was used to combine fucoidan
with polyethyleneimine (PEI). Similarly to the previously described study, when the medium pH was
decreased, the increased release rate of DOX was noted. In vivo studies were performed using BALB/c
mice with induced breast tumor. The results demonstrated that intravenously injected nanoparticles
with DOX were characterized by stronger antiproliferative properties and higher tumor inhibitory
activity than free DOX. The beneficial eects on the antitumor activity of the obtained nanoparticles
were caused probably by immunomodulatory properties of fucoidan, which were confirmed in the
in vitro studies [53]. In addition, fucoidan is characterized by the ability to improve the stability
of metallic nanoparticles thereby contributing to the reduction of their toxicity. Fucoidan-coated
gold nanoparticles with DOX caused a significant decrease in rabbit squamous carcinoma cells (VX2)
viability. The most significant decrease in viability of the examined cells was observed after using both
the discussed nanoparticles and laser irradiation. Similar conclusions were drawn from the in vivo
studies conducted in rabbits with eye tumor. Only in the group treated with nanoparticles and laser,
complete eradication of the tumor was observed after 6 days, and most importantly, there was no
relapse after 14 days [42]. In turn, Jang et al. used the layer-by-layer technique for the synthesis of CuS
nanoparticles alternately coated with fucoidan and polyallylamine hydrochloride (PAH). Based on
the obtained results, it was found that the applied nanoparticles showed stronger anticancer eect
than their components used separately. Obtained nanoparticles not only improved the intracellular
transport of fucoidan, which possesses the ability to induce apoptosis, but it also provided favorable
photothermal features. This was confirmed in the in vitro studies using human cervical cancer cells
(HeLa) and human lung adenocarcinoma cells (A549), and in thein vivo studies in mice injected with
these cells [54]. The same ingredients (fucoidan and PAH) were used to create nanoparticles with
MTX via self-assembly method. MTX was released according to the first order kinetics and it showed
stronger in vitro inhibition of HeLa and MCF-7 cell proliferation than unbounded drug. It should be
noted that fucoidan and PAH complex was biocompatible and did not aect cells viability [55].
In nanoparticles the combination of fucoidan and chitosan or its derivatives is very commonly
used, which is related to the properties of both polysaccharides. The positive charge of chitosan
allows electrostatic interactions with sulfate groups of fucoidan, leading to the formation of complexes.
In addition, chitosan is characterized by antifungal and antibacterial activity. Due to its ability to
muco-adhesion and overcoming epithelial barriers, it facilitates the transport of active substances.
Importantly, similar to fucoidan, it is biocompatible and its acquisition costs are relatively low [40,56,57].
In 2012, Silva et al. compared the activity of fucoidan–chitosan nanoparticles with the eect of fucoidan
solution. Nanoparticles obtained by coacervation method were biocompatible with human epithelial
cells (Caco-2) from colon adenocarcinoma and had the ability to open tight junctions between them.
Mar. Drugs 2019, 17, 458 9 of 20
As a result, nanoparticles showed better permeability and stronger anticoagulant eect than the
fucoidan solution per se [56]. Applying self-assembly method, Barbosa et al. produced nanoparticles
and as the positive charge provider, chitosan was used. MTX loaded nanoparticles were nontoxic for
both fibroblasts and keratinocytes when drug concentration did not exceed 50 g/mL. The strongest
anti-inflammatory activity manifested by the inhibition of Il1-, Il-6, and TNF- production and the
greatest penetration of the active substance through the pig’s ear skin provided formulation with the
highest fucoidan:chitosan ratio (5:1) [57]. Using the same electrostatic interactions between fucoidan
and chitosan, Huang et al. created pH-sensitive nanoparticles for oral administration of CUR, which
is characterized by limited bioavailability due to its poor solubility and sensitivity to environmental
conditions of the gastrointestinal tract. Whereas at pH 1.2 CUR release was inhibited, at pH 7.0
significant increase in drug release was observed. Hence, the discussed nanoparticles can be considered
as a carrier enabling oral administration of CUR, providing protection against degradation in the
stomach and adequate bioavailability after absorption in the intestine [58]. Similar conclusions were
also drawn from the studies in which CUR was placed in nanoparticles obtained from fucoidan and
o-carboxymethylchitosan by ionic crosslinking. Controlled release of CUR in the gastrointestinal tract,
its reduced cytotoxicity to mouse fibroblasts cells (L929), and increased cellular uptake by Caco-2
cells constitute the positive eects resulting from CUR encapsulation in nanoparticles [59]. By using
thiolated fucoidan and arginine-modified chitosan it was also possible to obtain nanoparticles with
dextran (DTX) or CUR. Due to the fact that utilized polysaccharides possess the ability to open
tight junctions in intestinal Caco-2 cell monolayer, and thiolated fucoidan inhibits the activity of the
P-glycoprotein pump, the penetration of both drugs were significantly better. Encapsulation of CUR as
model hydrophobic compound into nanoparticles improved its water solubility and stability [60].
To deliver antibiotics to the lungs, fucoidan and chitosan composed nanoparticles via simple
self-assembly method were designed. Antioxidant activity of the obtained nanoparticles (confirmed
by the test of reactive oxygen species with lipopolysaccharides and by the method of scavenging
1,1-diphenyl-2-picrylhydrazyl radicals), stability in phosphate buered saline, no negative eect on
the viability of A549 cells at a concentration from 0.37 mg/mL to 3 mg/mL and controlled release of
gentamicin make them an auspicious pulmonary drug delivery system [41]. Similar conclusions were
drawn from the study in which nanoparticles with gentamycin were obtained by ionotropic crosslinking
and applied intratracheally. The biphasic antibiotic release profile, as well as the antibacterial and
antioxidant properties of fucoidan, resulted in stronger growth inhibition of Klebsiella pneumoniae,
simultaneously limiting the potential nephrotoxic and ototoxic eects of gentamicin [50].
The protective properties of fucoidan were used to provide stability for silver nanoparticles. The
fucoidan–chitosan complex coated silver nanoparticles, produced by self-assembly method, showed
antibacterial activity both against Gram-positive ( Staphylococcus aureus) and Gram-negative ( Escherichia
coli) bacteria. Moreover, the use of nanoparticles at a concentration of 50 g/mL showed considerable
cytotoxicity to HeLa cells [61]. In turn, Elbi et al. investigated the activity of ciprofloxacin loaded into
chitosan nanoparticles obtained by ionic crosslinking using tripolyphosphate and subsequently coated
with fucoidan. As fucoidan reacts with scavenger macrophages receptors, the obtained nanoparticles
were characterized by better cellular uptake and thus led to more eective eradication of Salmonella.
Research conducted on infected by Salmonella Drosophila melanogaster confirmed that ciprofloxacin
(in the form of free drug, chitosan nanoparticles, or fucoidan coated chitosan nanoparticles) at
concentrations four times higher than its minimum inhibitory concentration (MIC) did not aect
the survival of the flies. However, the use of antibiotics at a concentration four times higher than
minimum bactericidal concentration (MBC) caused the greatest survival rate of flies treated with the
drug in the form of particles coated with fucoidan. Moreover, in this group the greatest decrease in
bacterial load was noted, which confirmed the stronger antibacterial activity of ciprofloxacin enclosed
in fucoidan coated chitosan nanoparticles, both in comparison to the free drug and to the form of
chitosan nanoparticles [40]. By using the layer-by-layer method, Pinheiro et al. received nanoparticles
made of alternating layers of chitosan and fucoidan deposited on polystyrene core. After removing
Mar. Drugs 2019, 17, 458 10 of 20
the polystyrene element, nanoparticles were filled with poly-l-lysine (PLL), which is characterized
by antibacterial properties. The satisfactory encapsulation eciency (about 45%) obtained by the
adsorption of PLL on the core before its removal, as well as the pH-dependent release of the active
ingredient from the nanoparticles, make them a promising drug carrier [62].
The peptide therapy is becoming increasingly important in medicine and pharmacy. However, due
to poor bioavailability, which results from peptide degradation in digestive tract and poor absorption,
oral administration is limited. Tsai et al. have recently developed nanoparticles with trimethyl chitosan
and fucoidan as carrier for insulin. Greater insulin protection against pH changes and better penetration
across the intestinal epithelium was observed. It was also shown that fucoidan possesses the ability to
inhibit -glucosidase activity. Obtained nanoparticles at concentration of 2 mg/mL inhibited activity of
this enzyme at ratio of 33.2% [63]. Additionally, the combination of fucoidan and chitosan provided
protection for the basic fibroblast growth factor (bFGF). Nanoparticles were obtained by the ionotropic
cross-linking. The eectiveness of encapsulation (EE), due to the ability of fucoidan to bind amino acids
on the surface of bFGF, increased with increasing fucoidan content (for nanoparticles prepared from
chitosan and fucoidan in a ratio of 10:1 and 1:10, EE was 80.9% and 90.4%, respectively). Moreover, the
same relationship regarding the contribution of fucoidan was observed with respect to the protection
of bFGF by enzymatic degradation and temperature inactivation. Nanoparticles at a concentration of
125 ng/mL showed no toxicity to pheochromocytoma PC12 cells, and due to the fucoidan and chitosan
content therein, the amount of bFGF required to neurite extension was significantly lower than free
bFGF [44].
It is worth noting that biological activity of nanoparticles built from fucoidan depends on the
properties of this polysaccharide and the form in which it was used. In 2017, Juenet et al. obtained
by redox radical emulsion polymerization (RREP) fucoidan-based nanoparticles. Due to the use of
fucoidan, which has an anity for P-selectin present on thrombocytes, the ability of nanoparticles
to bind to the activated plates was greater. The fluorescence intensity observed after perfusion of
both the fucoidan coated nanoparticles without recombinant tissue plasminogen activator (rt-PA) and
with it was significantly higher than those in which no fucoidan was used. In addition, nanoparticles
containing rt-PA and fucoidan showed the strongest thrombolytic activity in studies conducted in
C57BL/6 mice. The thrombus density after their application decreased to about 30%, whereas after
injecting nanoparticles without fucoidan, the value of this parameter was about 66%. It was found
that nanoparticles not only protected rt-PA, but because of the ability of fucoidan to imitate P-selectin
glycoprotein ligand-1, they improved its thrombolytic activity [64].
In turn, Lira et al. compared the properties of isobutylcyanoacrylate (IBCA) nanoparticles coated
with fucoidan, which were obtained by two dierent methods. Anionic emulsion polymerization (AEP)
allows the creation of stable formulations without the addition of DXT, whereas in the case of REEP
only those containing up to 25% of fucoidan in the polysaccharide mixture were stable. Cytotoxicity
was assessed after 48 h of nanoparticles incubation with macrophages J774 or fibroblasts NIH-3T3.
The method of obtaining nanoparticles had a major impact on the cytotoxicity of nanoparticles only
in relation to macrophages J774, because the IC50 value for nanoparticles formulated by AEP was
1.3 0.2 g/mL, and by REEP 9.6 0.5 g/mL. Interestingly, distribution of nanoparticles in these cells
was dependent not on the method used for their production, but on fucoidan presence. Fucoidan did
not aect the interaction of nanoparticles with fibroblast cells, but it also modulated nanoparticles
uptake and distribution within macrophages [65]. The safety of using nanoparticles was assessed
by Cai et al. Core of nanoparticles was made of poly(lactic-co-glycolic) acid (PLGA) and then it was
covered with alternating layers of poly-L-ornithine (PLO) and fucoidan using the layer-by-layer (LbL)
technique. Even at concentration of 100 g/mL, they did not aect the morphology and proliferation
ratio of breast epithelium (MCF-10A) cells. In addition, the hemolysis test on fresh blood from rabbits
proved that by closing the PLGA in the capsule made of fucoidan and PLO, its biocompatibility
increased (hemolysis rate for LbL and PLGA nanoparticles at concentration 100 g/mL was 2.67%
and 3.85%, respectively). Destructive eects on the liver, changes in nutrition and weight were not
Mar. Drugs 2019, 17, 458 11 of 20
observed in mice after intraperitoneal injection of these nanoparticles at concentration of 10 mg/mL.
The obtained results confirmed that such connection of fucoidan and PLO, not only protected active
ingredient, but also improved its biocompatibility [66].
Interestingly, Hwang et al. prepared nanoparticles for colonic drug delivery by combining CSN
and pure fucoidan. In order to obtain nanoparticles, two methods were used, diering in the order of
dissolution of the components. Appropriate amounts of fucoidan were dissolved in 2 mg/mL CSN
solution, or appropriate amounts of CSN were dissolved in 10 mg/mL fucoidan solution. Fucoidan
nanoparticles tested as CSN carrier not only decreased toxicity of CSN against RAW264.7 macrophage
cells, but they also improved its cytotoxicity in HCT-8 tumor cells [67].
Fucoidan was used to form shell of nanostructures dispersible in aqueous media. The source
of polymer composed of (1-3)-glucuronic acid substituted at the C4 position with fucose was the
brown seaweed Spatoglossum schroederi. Chemical modification with hexadecylamine allowed to
produce amphiphilic nanoparticles stable in water for up to 70 days. In order to determine the use
of nanoparticles in the treatment of tumors, their eect on the proliferation and viability of various
cell lines at 50, 100, and 500 g/mL was evaluated by MTT test. It is based on the reduction of the
water-soluble MTT dye, which is a triazole salt (3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium
bromide) to an insoluble dark blue formazan, the amount of which is proportional to the number of
viable cells. This study showed that the produced nanoparticles did not aect Chinese hamster ovary
(CHO) and mouse monocyte macrophage (RAW) cells. Antiproliferative activity was noted against
rabbit aorta endothelial cells (RAEC) and all tested tumor cells: human renal cell carcinoma (786),
human hepatocellular carcinoma cells (HepG2) and human marrow stromal cells (HS-5) [68].
4.2. Liposomes
Liposomes are specific type of structures, which size usually does not exceed 100 nm. The core
formed by the aqueous environment is surrounded by the phospholipid bilayer. The main reason for the
use of liposomes is the possibility of targeted therapy, which in addition to the improved eectiveness
of the treatment, allows to reduce drug side eects [69]. The antitumor eect of LMWF encapsulated in
liposomes was compared to the activity of fucoidan with molecular weight 2–10 kDa and 80 kDa using
human osteosarcoma cells (143B). It was shown that apoptosis induction by activating caspase pathway
was significantly stronger than by native fucoidan (95% and 25%, respectively). Similar conclusions
were observed in studies in C3H mice that were previously injected with murine osteosarcoma cells
(LM8). In the group receiving orally for 28 days liposomes with fucoidan or unprocessed fucoidan in
the amount of 100 mg/kg per day, there was a significant decrease in both volume and weight of the
tumor compared to mice receiving only water. However, better results were noted when fucoidan
liposomes were applied. Furthermore, liposomes were characterized by better penetration through the
Caco-2 cell layer. In turn, native fucoidan more strongly inhibited spontaneous metastases to the lungs.
None of the forms (unprocessed fucoidan and fucoidan liposomes) led to negative changes in animal
weight [70].
4.3. Microparticles
The term microparticles refers to particles with sizes ranging from 1 m to 1000 m. Analogously,
as in the nanoparticles discussed above, among these group of particles microspheres and microcapsules
can be distinguished. Microspheres prepared with fucoidan utilization are termed ‘fucospheres’.
The morphology of fucospheres developed by our research team is shown in Figure 3. They were
successfully obtained by the spray drying of 3% aqueous fucoidan solution (Table 1) using the Mini
Spray Dryer B-290 (Büchi, Flawil, Switzerland) with experimental parameters set as follows: inlet
temperature 122 C, outlet temperature 62 C, pressure 60 mmHg, aspirator blower capacity 100%,
feed rate 2.1 mL/min.
Mar. Drugs 2019, 17, 458 12 of 20
Mar. Drugs 2019, 17, x 12 of 21
(a)
(b)
Figure 3. Scanning electron microscope (SEM) pictures of fucospheres: (a) magnification °ø5000; (b)
magnification °ø20,000.
According to the literature, preparation of fucoidan‐based microparticles is usually performed
with additional copolymers or positive charge donors (Table 3). In 2006, Sezer et al. proposed the use
of fucospheres obtained by interaction between the negatively charged fucoidan and positively
charged chitosan and, as a model protein, bovine serum albumin (BSA) was used. It was noted that
with increasing fucoidan content in the fucospheres, BSA encapsulation capacity increased and the
value of zeta potential decreased. Formulated microspheres provided three‐phasic BSA release
profile [45]. Interactions between fucoidan and chitosan were also used to create fucospheres for the
treatment of dermal burns in rabbits. As in the previous study, the size of the fucospheres obtained
by polyion complexation increased with the increase in the polymer concentration. Wounds that were
treated with fucospheres, were characterized by the highest epithelial thickness. The most relevant
difference was observed on day 7th, when the value of epithelial thickness was 193, 121, 118, and 111
μm, for fucospheres, fucoidan solution, chitosan microspheres and untreated group, respectively.
Similarly, epithelial lengths increased with time, and on the 14th day of therapy the highest values
were observed in the group treated with fucospheres (2733, 2086, 2316, and 1950 μm, as above). The
fastest skin regeneration after burns was noted in the group treated with fucospheres due to the effect
of fucoidan on the migration of fibroblasts, release of growth hormones and cytokines involved in reepithelization.
In addition, based on this study, it was concluded that fucoidan and chitosan have a
synergistic effect on the treatment of skin burns [48].
Li et al. designed poly(alkylcyanoacrylate) (PACA) microcapsules with fucoidan layer on their
surface. The use of fucoidan was due to its ability to detect thrombosis by binding to P‐selectin. The
microparticles produced by emulsion–evaporation polymerization process were filled with contrast
agent (perfluorooctylbromide—PFOB) used in various imaging diagnostics (ultrasonography, The safety of microparticles application was
determined 3T3 mouse fibroblast cells. Even at concentration of 5 g/L, there was
no difference in cell viability when using microcapsules with and without fucoidan. Additionally,
two in vitro studies were performed to evaluate their binding capacity to P‐selectin. Both in the static
binding on human activated platelets and in the flow chamber assay, microparticles with fucoidan
showed significant adhesive properties. Moreover, designed microcapsules were administered
intravenously to healthy Wistar rats and rats with abdominal aortic aneurysm. The strongest
fluorescence was observed in the animals treated with microcapsules with fucoidan but only in the
affected area. Weaker bonds of both types of microparticles were noted in healthy blood vessels. It
was concluded that fucoidan microcapsules seem to be valuable targeting carrier candidate for drug
Figure 3. Scanning electron microscope (SEM) pictures of fucospheres: (a) magnification 5000;
(b) magnification 20,000.
According to the literature, preparation of fucoidan-based microparticles is usually performed
with additional copolymers or positive charge donors (Table 3). In 2006, Sezer et al. proposed the use of
fucospheres obtained by interaction between the negatively charged fucoidan and positively charged
chitosan and, as a model protein, bovine serum albumin (BSA) was used. It was noted that with
increasing fucoidan content in the fucospheres, BSA encapsulation capacity increased and the value of
zeta potential decreased. Formulated microspheres provided three-phasic BSA release profile [45].
Interactions between fucoidan and chitosan were also used to create fucospheres for the treatment of
dermal burns in rabbits. As in the previous study, the size of the fucospheres obtained by polyion
complexation increased with the increase in the polymer concentration. Wounds that were treated
with fucospheres, were characterized by the highest epithelial thickness. The most relevant dierence
was observed on day 7th, when the value of epithelial thickness was 193, 121, 118, and 111 m, for
fucospheres, fucoidan solution, chitosan microspheres and untreated group, respectively. Similarly,
epithelial lengths increased with time, and on the 14th day of therapy the highest values were observed
in the group treated with fucospheres (2733, 2086, 2316, and 1950 m, as above). The fastest skin
regeneration after burns was noted in the group treated with fucospheres due to the eect of fucoidan
on the migration of fibroblasts, release of growth hormones and cytokines involved in re-epithelization.
In addition, based on this study, it was concluded that fucoidan and chitosan have a synergistic eect
on the treatment of skin burns [48].
Li et al. designed poly(alkylcyanoacrylate) (PACA) microcapsules with fucoidan layer on their
surface. The use of fucoidan was due to its ability to detect thrombosis by binding to P-selectin.
The microparticles produced by emulsion–evaporation polymerization process were filled with
contrast agent (perfluorooctylbromide—PFOB) used in various imaging diagnostics techniques
(ultrasonography, magnetic resonance imaging). The safety of microparticles application was
determined by MTT test using 3T3 mouse fibroblast cells. Even at concentration of 5 g/L, there was no
dierence in cell viability when using microcapsules with and without fucoidan. Additionally, two
in vitro studies were performed to evaluate their binding capacity to P-selectin. Both in the static binding
on human activated platelets and in the flow chamber assay, microparticles with fucoidan showed
significant adhesive properties. Moreover, designed microcapsules were administered intravenously to
healthyWistar rats and rats with abdominal aortic aneurysm. The strongest fluorescence was observed
in the animals treated with microcapsules with fucoidan but only in the aected area. Weaker bonds
of both types of microparticles were noted in healthy blood vessels. It was concluded that fucoidan
Mar. Drugs 2019, 17, 458 13 of 20
microcapsules seem to be valuable targeting carrier candidate for drug substances or contrast agents
for the treatment or imaging of diseases that are accompanied by overexpression of P-selectin [71].
Fucoidan microparticles are also examined as potential drug carriers in the cancer treatment.
In 2017,Wang et al. used the layer-by-layer self-assembly technique to develop microparticles with
DOX. After coating the core made of calcium carbonate with PLO and fucoidan, they were loaded
with DOX at a high, nearly 70% encapsulation eciency. Designed microparticles were characterized
by biocompatibility—they did not aect the survival of C2C12 mouse myoblasts (at concentration of
400 g/mL cell viability was about 90%). In addition, the hemolysis rate at 200 g/mL was lower than
3%. A study conducted at pH 7.4 showed prolonged DOX release, and antitumor tests on breast cancer
cells (MCF-7) confirmed its eective release and ability to inhibit cells more than free DOX [72].
Fucoidan microparticles are also tested as antibiotics carriers. Sezer et al. compared the properties
of fucospheres and chitosan microspheres, obtained by polyion complexation and precipitation
methods, respectively. Ofloxacin (OFL) encapsulation eciency (EE) was greater in fucospheres than
chitosan microparticles (EE values 63.9%–94.8% and 48.8%–89.2%, respectively). The use of fucoidan
caused a slower OFL release, consistent with the Higuchi kinetics model [73]. Recently, Cunha et al.
conducted studies on the use of fucoidan-based microparticles in the treatment of tuberculosis. This is
the result of fucoidan ability to recognize macrophages in the alveoli, which allows the delivery of
drugs to the aected areas and leads to an increase in the eectiveness of the therapy. The microparticles
containing isoniazid (INH) or rifabutin (RFB) produced by the spray-drying with high yield (75%–85%)
were characterized by the appropriate aerodynamic properties measured using the Andersen cascade
impactor (ACI). The mass median aerodynamic diameter of particles with isoniazid was about 3.78 m,
and with rifampicin 1.99 m. The fraction of fine particles tested with the dry powder inhaler type
RS01, showing the most eective penetration to the lungs, was 39% and 55%, respectively. Due to the
wrinkled surface of the microparticles and the good solubility of fucoidan, the total release of both
drugs occurred in a short time (about 15 minutes), regardless of the medium pH and the solubility of
the free drug. It is worth noting that MTT tests proved that microencapsulation reduces the toxicity
of loaded RFB, both in relation to human alveolar epithelium (A549) and human monocytic (THP-1)
cells. The viability of A549 and THP-1 cells after 24 h incubation with RFB at a concentration of 0.05
mg/mL was 50% and 49%, respectively. In turn, the use of the microparticles with RFB increased cells
viability after 24 h incubation even up to 90% (A549) and to around 70% (THP-1). However, unloaded
fucoidan-based microparticles, free fucoidan, and free INH did not show cytotoxicity [43]. Very similar
results were noted with reference to fucoidan-based microparticles being a combination of INF and
RFB. The spray drying technique allowed to obtain product with a yield of over 80%. By using the
RS01 inhaler in the ACI, it was found that product exhibited proper aerodynamic properties. In studies
in vitro it was reported that microparticles did not aect the viability of A549 cells and their viability
was classified above 70%, regardless the incubation time. THP-1 cells showed greater sensitivity and
at concentration 1.0 mg/mL after 24 h of exposure, the viability decreased to 65%. Viability of A549
and THP-1 cells after 3 h incubation with free RFB (0.05 mg/mL) was 68% and 56%, whereas with
microparticles (1.0 mg/mL) containing INH and RFB at mass ratio 1:0.5, 84% and 77%, respectively.
It is worth noting that the significant uptake of microparticles by macrophage-like cells (up to 87%)
along with their simultaneous stimulation for the production of cytokines involved in the fight against
pathogen was associated with the presence of fucoidan [74].
4.4. Semi-Solid Formulations
Hydrogels are three-dimensional formulations made of hydrophilic polymers with appropriate
structure and properties. They are widely used in biomedicine, for example as wound dressings,
implants, drug delivery systems, or tissue regeneration materials [75].
Mar. Drugs 2019, 17, 458 14 of 20
Table 3. Characteristic of selected fucoidan-based microparticles.
Fucoidan (Source/Molecular
Weight)
Copolymer/Positive
Charge Donor Drug Method of Obtaining Application Route of
Administration Ref.
Fucoidan
(Fucus vesiculosus, 80 kDa) Chitosan Bovine serum albumin Ionotropic cross-linking Peptide and protein
delivery NA 1 45
Fucoidan
(Fucus vesiculosus, 80 kDa) Chitosan - Polyion complexation Treatment of dermal burns Topical 48
Fucoidan Poly(alkylcyanoacrylate)
and dextran Perfluorooctylbromide Emulsion-evaporation
polymerization Targeting carrier Intravenous 71
Fucoidan (200–400 kDa) Poly-l-ornithine (shell);
calcium carbonate (core) Doxorubicin Layer-by-layer
self-assembly Anticancer therapy NA 1 72
Fucoidan
(Fucus vesiculosus, 80 kDa) Chitosan Ofloxacin Polyion complexation Antibiotics carriers NA 1 73
Fucoidan
(Laminaria japonica,
598.4 Da–0.598 kDa)
- Isoniazid or rifabutin Spray-drying Tuberculosis therapy Pulmonary 43
Fucoidan (Laminaria japonica) - Isoniazid and rifabutin Spray-drying Tuberculosis therapy Pulmonary 74
1 No data available.
Mar. Drugs 2019, 17, 458 15 of 20
Unique properties, such as anticoagulant and anti-inflammatory activity make fucoidan a useful
component in the formation of hydrogels. Sezer et al. proposed a hydrogel obtained by combining
fucoidan with an oppositely charged chitosan. An important feature of hydrogels is the ability to
absorb exudate and to provide adequate moisture. It turns out that formulations with fucoidan
addition were characterized by greater possibility of water absorption and higher swelling ratio than
pure chitosan gels. Electrostatic interactions between polymers significantly aected the hardness of
the gels, which increased with the increase of fucoidan concentration. Similarly to the parameters
discussed above, formulations containing the highest concentrations of polymers (0.75% fucoidan
and 2% chitosan) showed the greatest cohesiveness and adhesion values. Stronger mucoadhesive
properties were also associated with the content of fucoidan. Hydrogels containing only chitosan
and chitosan with the addition of 0.25% or 0.75% fucoidan showed the following values of work of
adhesion 23, 62, and 142 J/cm2, respectively. The eectiveness of wound healing using the designed
hydrogels has also been evaluated in vivo. In a study conducted on New Zealand rabbits statistically
significantly higher values of the length and thickness of the epithelium were observed in wounds
treated with fucoidan–chitosan gel. In addition, the number of rete pegs, responsible for fixing the
epidermis in the dermis and organized nuclear regions (NOR), which confirmed cell proliferation,
were also the highest in this group. Complete cure within 21 days was observed only with the use of a
complex hydrogel, arming the synergistic eect of fucoidan and chitosan on the healing process [76].
In turn, Murakami et al. proposed hydrogels made of chitosan/chitin, fucoidan and alginate.
Hydrogels obtained by crosslinking with ethylene glycol diglycidyl ether were safe and showed no
cytotoxicity to human dermal fibroblast (DFC) and dermal microvascular endothelial (DMVEC) cells.
In comparison to the commercially available product (Kaltostat—calcium alginate fiber), they were
characterized by better—gradually increasing exudate absorption capacity, without maceration for 18 h.
In vivo studies in rats with mitomycin C-induced, impaired wound healing confirmed the positive
eects of fucoidan—its ability to interact with growth factors (FGF-1 and FGF-2) and with cytokines
involved in the reconstruction of the epidermis and angiogenesis processes. The stimulation of the
granulation and capillary formation observed after 7 days of treatment with hydrogel contributed
to the best results after 18 days of application, including eective division of epidermal stem cells
located in the intact adjacent epidermis and the strongest wound closure. Interestingly, the eect of the
hydrogel on the course of wound healing without the factor inhibiting cell proliferation (mitomycin
C) was insignificant [77]. Hydrosheets made of the same polymers (fucoidan, chitin/chitosan, and
alginate) were compared with two dierent hydrogel dressings (Kaltostat and DuoACTIVE). The
obtained results indicated an advantage in the treatment of mitomycin C-impaired wounds with the
proposed dressings than with the currently available products or plastic wrap used as a control (in
the study group within 14 days there was a reconstruction of the epidermis with significantly higher
wound closure rate). It should be added that the use of all these dressings had no significant eect on
the wounds which were not treated with mitomycin C [78].
As fucoidan is characterized by the ability to interact with growth factors and possesses lower
than heparin anticoagulant activity, it might be utilized to create drug carrier for tissue regeneration.
The main limitation in designed carriers for protein compounds, is their short half-life in vivo, resulting
from sensitivity to temperature and enzymes. Nakamura et al. formed a micro complex-hydrogel
composed of chitosan and fucoidan loaded with fibroblast growth factor-2 (FGF-2). The gradual release
of FGF-2 along with progressive degradation of the hydrogel contributed to statistically significantly
stronger neovascularization in the test group compared to mice injected with growth factor alone
or hydrogel placebo [79]. In turn, vascular endothelial growth factor (VEGF) and the influence of
fucoidan molecular weight on its activity in micro- and macroporous 3D scaolds were evaluated by
Purnama et al. Hydrogels obtained by crosslinking with sodium trimetaphosphate (STMP) pullulan
and DXT were control group, and those with fucoidan (with low, medium, and high molecular
weight—LMWF/MMWF/HMWF) addition were test groups. It was shown that significant decrease
in the release rate of VEGF compared to the control, regardless of the pore size in the hydrogel,
Mar. Drugs 2019, 17, 458 16 of 20
guaranteed the addition of fucoidan with medium molecular weight (39 kDa). A positive aspect is
also the significant increase in the number of human endothelial progenitor cells and their extent of
proliferation. The synergism of MMWF and VEGF was confirmed by observations after subcutaneous
injection of hydrogels to C57/BL6J mice. Both the neovessel area and density in the group treated with
hydrogels obtained from MMWF and VEGF were statistically higher compared with control group or
group treated scaolds with single substance [80].
In order to obtain hydrogel to ocular applications made of poly(2-hydroxyethyl methacrylate)
(pHEMA), fucoidan and methacrylic acid (MAA), Lee at al. used ethylene glycol dimethacrylate
(EGDMA) as crosslinking agent. Developed hydrogels were tested in terms of water absorption
capacity, adsorption of proteins and antibacterial properties. It was noted that water content of
hydrogels and adsorption of proteins (especially lysozyme) did not depend on the concentration of
fucoidan and increased only with MAA increase. The predicted antibacterial activity of fucoidan with
regard to S. aureus and E. coli turned out to be infinitesimal, and its potentiation was caused by the
addition of MAA [81].
5. Conclusions
Fucoidan is the important ingredient in various pharmaceutical formulations. It can be used
either as a substance with a specific therapeutic eect or as an excipient, allowing to obtain a drug
form with appropriate properties. However, utilization of fucoidan in the pharmaceutical industry
involves many challenges. Diculties appear already at the acquisition stage. Dierences in the
growth conditions of algae, pollution of the marine environment, as well as a wide range of species
that are the source of fucoidan contribute to the heterogeneity of its properties. This, in turn, leads to
the necessity of matching extraction and purification methods to allow obtaining the raw material with
desired characteristics [82–84]. Fucoidan features are also influenced by sugar composition and sulfate
content, and by dierential molecular weight (10–2000 kDa). Other disadvantage is lack of gelation
ability, important feature in designing drug dosage forms. This limitation can be overcome by fucoidan
combining with polymers (chitosan and its derivatives) or other compounds that provide a positive
charge (protamine, polyethyleneimine, polyallyamine hydrochloride, poly(isobutylcyanoacrylate),
poly(lactide-co-glycolide), poly-l-orithine, and hexadecylamine, poly(alkylcyanoacrylate)). The broad
spectrum of biological activity of this polysaccharide per se contributes to the fact that application
of designed drug carriers, like nanoparticles, liposomes, microparticles or semi-solid forms is very
dierential and using of fucoidan not only provides protection of the loaded active substance, but can
also increase its potency and improve the eectiveness of the treatment. Due to diversified properties
and potential broad area of application, the interest in fucoidan utilization in the biomedicine and
pharmaceutical industry will be probably extended.
Author Contributions: Conceptualization, A.C. and K.W.; designing and performing experiments, A.C. and M.S.;
writing—review and editing, A.C. and K.W.; visualization, A.C.; supervision, K.W.; funding acquisition, K.W.
Funding: This research was funded by Medical University of Bialystok grant number SUB/2/DN/19/004/2215.
Acknowledgments: We gratefully acknowledge A. Basa for SEM pictures made using the scanning electron
microscope(Inspect.S50, FEI Company, Hillsboro, OR, USA), which is a part of Center BioNano Techno equipment,
supported partly by EU founds (projectnumber POPW.O1.03.00-20-004/11).
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Kylin, H. Zur biochemie der meeresalgen. Z. Physiol. Chem. 1913, 83, 171–197. [CrossRef]
2. Chollet, L.; Saboural, P.; Chauvierre, C.; Villemin, J.N.; Letourneur, D.; Chaubet, F. Fucoidans in nanomedicine.
Mar. Drugs 2016, 14, 145. [CrossRef] [PubMed]
3. Fitton, J.H.; Stringer, D.N.; Karpiniec, S.S. Therapies from fucoidan: An update. Mar. Drugs 2015, 13,
5920–5946. [CrossRef] [PubMed]
Mar. Drugs 2019, 17, 458 17 of 20
4. Cunha, L.; Grenha, A. Sulfated seaweed polysaccharides as multifunctional materials in drug delivery
applications. Mar. Drugs 2016, 14, 42. [CrossRef] [PubMed]
5. Cardoso, M.J.; Costa, R.R.; Mano, J.F. Marine origin polysaccharides in drug delivery systems. Mar. Drugs
2016, 14, 34. [CrossRef] [PubMed]
6. Garcia-Vaquero, M.; Rajauria, G.; O’Doherty, J.V.; Sweeney, T. Polysaccharides from macroalgae: Recent
advances, innovative technologies and challenges in extraction and purification. Food Res. Int. 2017, 99,
1011–1020. [CrossRef] [PubMed]
7. Zhao, D.; Xu, J.; Xu, X. Bioactivity of fucoidan extracted from Laminaria Japonica using a novel procedure
with high yield. Food Chem. 2018, 245, 911–918. [CrossRef] [PubMed]
8. Jiao, G.; Yu, G.; Zhang, J.; Ewart, H.S. Chemical structures and bioactivities of sulfated polysaccharides from
marine algae. Mar. Drugs 2011, 9, 199–223. [CrossRef]
9. Hifney, A.F.; Fawzy, M.A.; Abdel-Gawad, K.M.; Gomaa, M. Upgrading the antioxidant properties of fucoidan
and alginate from Cystoseira trinodis by fungal fermentation or enzymatic pretreatment of the seaweed
biomass. Food Chem. 2018, 269, 387–395. [CrossRef]
10. Mansour, M.B.; Balti, R.; Yacoubi, L.; Ollivier, V.; Chaubet, F.; Maaroufi, R.M. Primary structure and
anticoagulant activity of fucoidan from the sea cucumber Holothuria polii. Int. J. Biol. Macromol. 2019, 121,
1145–1153. [CrossRef]
11. Lee, K.Y.; Jeong, M.R.; Choi, S.M.; Na, S.S.; Cha, J.D. Synergistic eect of fucoidan with antibiotics against
oral pathogenic bacteria. Arch. Oral Biol. 2013, 58, 482–492. [Cros
|