Colcom D 21 00172 R2
Colcom D 21 00172 R2
Colcom D 21 00172 R2
Prasher, P., Sharma, M., Mehta, M., Satija, S., Aljabali, A. A., Tambuwala, M. M., Anand, K., Sharma, N.,
Dureja, H., Jha, N. K., Gupta, G., Gulati, M., Singh, S. K., Chellappan, D. K., Paudel, K. R., Hansbro, P. M., &
Dua, K. (2021). Current-status and applications of polysaccharides in drug delivery systems. Colloid and
Interface Science Communications, 42, 1-17. [100418]. https://doi.org/10.1016/j.colcom.2021.100418
Published in:
Colloid and Interface Science Communications
Publication Status:
Published (in print/issue): 31/05/2021
DOI:
10.1016/j.colcom.2021.100418
Document Version
Peer reviewed version
General rights
Copyright for the publications made accessible via Ulster University's Research Portal is retained by the author(s) and / or other copyright
owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these
rights.
Highlights
Parteek Prasher1*, Mousmee Sharma2, Meenu Mehta3, Saurabh Satija3, 4, Alaa A. Aljabali5,
Murtaza M. Tambuwala6, Krishnan Anand7, Nitin Sharma8, Harish Dureja9, Niraj Kumar Jha10,
Gaurav Gupta11, Monica Gulati12, Sachin Kumar Singh12, Dinesh K. Chellappan13, Keshav
Paudel14, 15, Philip M. Hansbro14, 15, Kamal Dua3, 14*
1. Department of Chemistry, University of Petroleum & Energy Studies, Dehradun
248007, India
2. Department of Chemistry, Uttaranchal University, Dehradun 248007, India
3. Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney,
NSW 2007, Australia.
4. School of Pharmaceutical Sciences, Lovely Professional University, Phagwara,
144411, Punjab, India
5. Yarmouk University, Faculty of Pharmacy, Department of Pharmaceutics and
Pharmaceutical Technology, Irbid 566, Jordan
6. School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, County
Londonderry, Northern Ireland BT52 1SA, United Kingdom
7. Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences
and National Health Laboratory Service, University of the Free State, Bloemfontein,
South Africa
8. Department of Pharmacy, Merrut Institute of Engineering and Technology 250005, UP,
India.
9. Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak
124001, India.
10. Department of Biotechnology, School of Engineering and Technology (SET), Sharda
University, Greater Noida, 201310, UP, India.
11. School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan, India
12. School of Pharmaceutical sciences, Lovely Professional University, Jalandhar-Delhi
G.T Road, Phagwara, Punjab, India
13. Department of Life Sciences, School of Pharmacy, International Medical University,
Bukit Jalil 57000, Kuala Lumpur, Malaysia
14. Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia
15. School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo,
NSW, 2007, Australia
*Corresponding author email: [email protected], [email protected]
Revised Manuscript (Text Unmarked) Click here to view linked References
O O
O O
HH
N N HN NH
R R R R
PS-based Therapeutic agents & Type of Cell types Model of study Polymer Advantages References
polymer targeting
Dextran COX-2 siRNA/ Passive Breast cancer cell In vitro and in vivo Biocompatible, pH stimuli- [215]
lines (MDA- responsive, down-regulate
MB231) Cyclooxygenase COX-2 genes in
tumor cells
Dextran- folic DOX/Active Breast cancer cell In vitro and in vivo Reducing the side effect of DOX, [216]
acid line (4T1) enhanced tumor growth
inhibition, and prolonged survival
rate
Dextran- Paclitaxel/ Passive Breast cancer cell In vitro and in vivo Enhanced cellular uptake, [216]
Indomethacin line (MCF-7) prolonged pharmacokinetics
(prolonged circulation time and
slower elimination), enhanced
tumor growth inhibition
Dextran- Paclitaxel/ Passive Colorectal cell line In vitro and in vivo Cellular proliferation enhanced [217]
albumin (CT-26) inhibition, inducing apoptosis,
Enhanced drug circulation times
Dextran-folic Resveratrol/ Active Lung cell line In vivo Enhanced cellular apoptosis and [218]
acid (A549) drug intake
Dextran-DOX Doxorubicin/ Passive Lymphoma cell In vitro and in vivo Reduced cytotoxicity (cardiac), [219]
lines enhanced apoptosis, enhanced
intracellular intake leading to
increased DOX concentration that
inhibits tumor growth
Hyaluronic 5-Fluorouracil/ Active Lung cell line In vitro Enhanced cell apoptosis and [220]
acid- Chitosan (A549) & cellular drug accumulation
liver cell line
(HepG2)
Hyaluronic Paclitaxel & curcumin /Active Breast cancer cell In vitro and in vivo Prolonged pharmacokinetics, [221]
acid- Poly- line (MCF-7) Enhanced cellular accumulation
lactic-co-
glycolic acid;
Hyaluronic Paclitaxel/Active Breast cancer cell In vitro Decreased the IC50 of paclitaxel [73]
acid- Poly- line significantly
lactic-co- (MDA-MB-231)
glycolic acid;
Hyaluronic Naringenin/Active Lung cancer cell In vitro and in vivo Enhanced cell apoptosis, [222]
acid- Poly-ε- line (A549) increased drug intake resulting in
caprolactone- inhibiting cancer growth
Chitosan
Hyaluronic Paclitaxel/Active Breast cancer cell In vitro and in vivo Enhanced cancer growth [223]
acid-Silica line (MCF-7) suppression, increased intake and
drug accumulation
Hyaluronic Doxorubicin & Gemcitabine Breast cancer cell In vitro and in vivo Dual modality drug delivery [224]
acid /Active lines system, enhanced inhibition to
(MDA-MB-231 cancer growth
& 4T1)
Hyaluronic Doxorubicin & Cisplatin /Active Breast cancer cell In vitro and in vivo Tumor growth inhibitor at low [225]
acid- lines drug concentrations
Polyamidoamin (MDA-MB231
e & MCF-7)
Hyaluronic TWIST- siRNAs (Basic helix- Ovarian cancer cell In vitro and in vivo Reduced cancer growth and cell [226]
acid-Silica- loop-helix transcription factors)/ line survival through selective
polyethyleneim Active (Ovcar-8) targeting
ine
Hyaluronic 5-Fluorouracil /Active Colon cancer cell In vitro and in vivo Tumor repression, high [227]
acid-Silica line (HT29) cytotoxicity and high
accumulation in the tumor
through selective targeting
Chitosan- N- DOX/Passive Breast cancer cell In vitro Enhanced cytotoxicity and [228]
acetyl histidine lines MCF-7 cellular uptake
and arginine
Chitosan- DOX & IL-2/Active Lung cancer cell In vitro and in vivo Increase IgG expression levels, [229]
Trimethyl and line (A549), Increased cytotoxic T
folate Liver cell line lymphocytes
(SMMC-7721)
and hepatoma cell
line
(H22)
Chitosan Suramin and Breast cancer cell In vitro and in vivo Inhibit Breast and lung metastasis [230]
DOX/ Passive line
(MDA-MB231)
Chitosan- DOX and IL17RB Breast cancer cell In vitro Decrease tumor cellular viability, [231]
Carboxymethyl siRNA/ Passive line inhibits growth, tumor cells
dextran (MDA-MB361) migration, and proliferation
Chitosan- Curcumin/Passive Colon cancer cell In vitro Enhanced cellular uptake, [232]
Hyaluronic line (HT29) reduced cellular proliferation, and
acid- high toxicity
sulfobutyl-
ether-β-
cyclodextrin
Alginate DOX/Passive Melanoma cell line In vitro and in vivo Inhibit tumor growth, higher [233]
(B16) cellular uptake, high cytotoxicity
Alginate Gold NPs (AuNPs) & cisplatin/ Colon cancer cell In vitro and in vivo Tumor growth high inhibition, [234]
Passive line (CT26) enhanced survival
Alginate Paclitaxel/Passive Different breast In vitro and in vivo Promote cell cycle arrest, induce [235]
cancer cell lines apoptosis and reduce the
bioavailability
Alginate- DOX/Active Breast cancer cell In vitro Inhibit tumor progression and [236]
chitosan line (MCF-7) cellular growth
Pullulan Floate & Cervical (Hela) In vitro and in vivo Tumor growth inhibition [237]
Polyethylenimine/Active and liver
(HepG2)
Pullulan- Paclitaxel/Active Liver cancer cell In vitro and in vivo Lower cytotoxicity due to [238]
Folate line selective targeting and prolonged
(SMMC-7721) release
Therefore, the polysaccharide offer a fascinating choice as polymeric material in drug delivery
due to biodegradability and biocompatibility. The application of polysaccharide in drug
delivery is growing very fast as it provides a safe, nontoxic and tunable alternates of synthetic
polymers for the same purpose. The diverse chemical structures, functional groups and
physicochemical properties are making polysaccharides a suitable candidate in drug delivery
[239, 240].
Polysaccharide based controlled release and targeted formulations
Unlike the synthetic hydrophilic polymers, polysaccharide possess a variety of hydrophilic
functional groups on its structure like –OH, -COOH, and –NH2 group which are responsible
for water absorption and further swelling of polysaccharide. These groups grant a variety of
functional capability to the polysaccharide-based systems such as bioadhesion and control
release property. Controlled release property of polysaccharide-based drug delivery systems
has been explored extensively for achieving a prolonged delivery of variety of therapeutics
such as chemotherapeutics, proteins, peptides, nucleic acid and many more [1]. The control
release of therapeutics from the swelled polysaccharide depends on the porosity or degree of
swelling, further regulated by certain parameters like pH, temperature, ionic strength and
electric fields [241]. Several polysaccharides present controlled release and target specific
delivery applications.
1. Chitosan: It is a linear polysaccharide obtained from the exoskeleton of arthropods such as
crabs, lobster, and shrimp and cell walls of fungi. It is available in variety of molecular weight
and degree of acetylation, which control its physical and biological property. Hydroxy and
amino groups on the chitosan are involved in making intra- and intermolecular hydrogen bonds.
The pH dependent protonation of amino group leads to swelling of the Chitosan to form three-
dimension structure gel at pH lower than the pKa [242]. Various chitosan salts such as chitosan
palmitate and chitosan laurate exhibit different degrees of swelling, and thus drug release from
the variety of dosage form. The presence of primary amines in chitosan is responsible for its
biological activity such as antimicrobial, anticancer. Chitosan nanoparticles showed greater
antimicrobial activity against Staphylococcus aureus, Klebsiella pneumoniae, Pseudomonas
aeruginosa, and Escherichia coli compared to parent chitosan and chitin [243]. Chitosan-based
controlled release and targeted formulations developed so far are compiled in Table 2.
2. Alginate: It is an extensively studied linear polysaccharide derived from brown seaweed
(Macrocystis pyrifera, Laminaria hyperborean) and from some certain bacteria belonging to
the family Pseudomonas and Azotobacter. Similar to chitosan, aginate also form a thick gel at
low pH, which served as a barrier for controlled release of therapeutics. Additionally, alginate
also form thick gel with certain polyvalent cations (Ca2+ and Na+) due to crosslinking of
carboxylic groups of polysaccharide structure. Due to the bioadhesion property of alginate, it
offers targeted drug delivery applications. So far, this polysaccharide has been explored for
variety of formulations including matrix tablet [244], microspheres [245], pellets [246] and
nanoparticles for targeted and functionalized delivery [247] (Table 2). The combination of S-
nitroso-mercaptosuccinate (a nitric oxide donor) and green tea synthesized silver nanoparticles
incorporated into alginate hydrogel showed synergistic antimicrobial activity against
Staphylococcus aureus, Escherichia coli and Streptococcus mutants compared to individual
compounds [248].
3. Pectin: Pectin is a complex polysaccharide present almost one third of the dry cell wall
material of most of the plants. These are higher molecular weight (50,000 and 150, 000 Da)
polysaccharides consist α-(1-4) linked esterified D-galacturonic moiety. Various
physicochemical parameters of pectin like solubility, viscosity and gelling properties depends
upon the degree of methyl esterification of D-galacturonic moiety. Depending upon the degree
of esterification two types of pectin are commercially available; highmethoxyl pectins (degree
of esterification > 50) and low methoxyl pectins (degree of esterification < 50) [249]. Various
plant pectins reportedly exhibit applications as controlled release excipients in tablet, hydrogels
and pellets. Additionally, plant pectins offer applications in colon targeted drug delivery due
to their biodegradation potential by colonic microflora [250]. The pectin polysaccharide from
Ulmus pumila L. (PPU) possess potent anti-inflammatory activity. It was observed that the
selenized-PPU inhibit lipopolysaccharide (LPS)-stimulated nitric oxide release by targeting the
protein expression of inducible nitric oxide synthase in murine macrophage cell line RAW
264.7 [251].
4. Guar Gum: It is a high molecular weight linear polysaccharide obtained from the seeds of
the plant Cyamopsis tetragonoloba. The polysaccharide consist of linear chain of (1 → 4)-
linked β-D-mannopyranosyl units with (1 → 6)-linked α-D-galactopyranosyl residues as side
chains with the ratio of mannose to galactose units as 2:1. This polysaccharide swells in polar
solvent and thus has been widely explored for various pharmaceutical applications. Addition
of ionic and non-ionic additives (sodium chloride and glycerin) could change the swelling and
erosion property of polysaccharide for offering a variety of applications [14]. This
polysaccharide serves as tablet binder sustain release polymer and viscosity enhancers. This
polysaccharide has become the primary choice as excipient in colon specific oral product as it
remain indigestible in upper GIT and completely degrades by the colonic microflora [252].
Gamal-Eldeen prepared guar gum C-glycosylated derivative (GG) and its sulphated derivative
(SGG) to investigate its anti-cancer and anti-inflammatory potency. It was observed that GG
inhibits cytochrome P450 1A (a carcinogen activator enzyme) and stimulated the carcinogen-
neutralizing enzymes such as glutathione-S-transferases. Furthermore, both GG and SGG were
able to exert anti-inflammatory by reducing the release of nitric oxide and tumour necrosis
factor-alpha in LPS induced RAW264.7. GG and SGG also inhibited the proliferation of human
hepatocellular carcinoma cells (Hep G2), while only SGG was particularly toxic for MCF-7
[253]. Table 2 presents the selected applications of guar gum as pharmaceutical excipients.
5. Starch: Starch is a naturally available polysaccharide obtained from plants. Its characteristic
properties such as biotolerance and non-immunogenicity for human use, and easy availability
make it a commonly used polymers/pharmaceutical excipient [254, 255]. In comparison to
other polymers such as gum and cellulose, starch does not need to undergo excessive
purification, as they are relatively pure. Structurally, starch has two different glucan chains a.
amylose (linear polymer) and b. amylopectin (branched polymer) representing 98-99% of the
dry weight. In the production of capsules and tablets, starch is widely utilized as a lubricant,
diluent, binder and disintegrant whereas it is also commonly employed for variety of
specialized drug delivery applications, for example delivery of specialized drug and targeting
specific parts of human body [256]. The microsphere of oregano essential oil prepared by
starch-based material utilizing supercritical fluid technology showed superior antioxidant
capacity (in terms of oxygen radical absorbing capacity) and stability. Starch-based drug carrier
systems present high efficacy in the oral delivery of insoluble drugs [257]. Luo et al. (2020)
encapsulated curcumin into starch microparticles and the formulation showed enhanced
stability against photodegradation and oxidative modification. Furthermore, the size of
curcumin starch microparticles was possible to control accurately from 0.3-2µm by altering the
rate of debranching reaction that caused alteration of release characteristic of curcumin. These
observations suggested the management of the release site by modifying the crystallinity or
size of microparticles [258]. Alp et al. 2019, formulated starch nanoparticles for the delivery
of epigenetic drugs CG-1521 for breast cancer that inhibits histone deacetylase. The
nanoparticles formulation showed decrease release rate of CG-1521 allowing remarkable
cytotoxicity against MCF-7 breast cancer cell as compared to free CG-1521 [259].
Table 2: Different polysaccharides explored for the sustained release of pharmaceuticals
Folic acid Methotrexate Enhanced uptake of delivery systems by colorectal cancer [266]
conjugated cells
nanoparticles
Nanoaggregates Isoniazid and Formulations shows enhanced local action due to sustained [267]
Rifampicin
release
Hydrogel Rifampicin Enhance mucoadhesion thus better efficacy [268]
Starch Microspheres Oregano essential oil High antioxidant activity values and stability [257]
Nanoparticles CG-1521 (histone Decrease release rate of CG-1521 allowing remarkable [259]
deacetylase inhibitor)
cytotoxicity against MCF-7 breast cancer
Polysaccharides as emulsion stabilizers
The amphiphilic nature of chemically modified polysaccharides causes their adsorption at the
interface of oil and aqueous solvents that results in the stabilization of the emulsion [269]. This
property of polysaccharides proved highly beneficial for the delivery of lipophilic or non-polar
therapeutics at their target site [270]. As such, the native cellulose does not offer emulsion
stabilization due to its non-solubility in water. The chemically modified cellulose not only
improves its aqueous solubility but also affords the stabilization of the emulsion [271]. The
large size of polysaccharides slows down their adsorption process compared to the small sized
surface-active agents. Mainly, the polysaccharide-based emulsifiers mainly base on pectins,
gum arabic, galactomannans, and cellulose. Cellulose and starch promote the emulsion
stabilization only after their chemical modification however; the polysaccharides such as gum
arabic and pectins endowed with covalently linked glycoproteins possess intrinsic ability for
the stabilization of the emulsion. In addition, the polysaccharides such as xanthan gum,
carrageenan, alginates, hyaluronan, chitosan, and alginates that display trivial interfacial
activity offer the eulsion stabilization via thickening, gelling, and structuring [272]. The
structural relationship between chitin-based nanomaterials to that of cellulose results in a
similar amphiphilic behavior for potential applications as stabilizers of oil-in-water Pickering
emulsion. Starch microgranules demonstrate similar applications however; the nanocrystals
and nanospheres of starch exhibit a limited stabilization of the emulsion [273]. Yan et al. 2019
reported the stabilization of Pickering emulsions droplets by nanocrystals of bacterial cellulose
for hydrophobic drug delivery of Alfacalcidol. The irreversible adsorption of bacterial cellulose
nanocrystals at the oil-water interface of Pickering emulsions prevented the coalescence of the
droplets. The emulsion showed Ostwald ripening in the alginate solution. The interfacial
assembly of the amphiphilic bacterial cellulose nanocrystals and the hydrogel shells of the
alginate beads generated by the external gelation led to the achievement of efficient loading
and a controlled release of Alfacalcidol. The release mechanism of Alfacalcidol from the
composite beads followed non-Fickian transport. The alginate composite beads demonstrated
minimal toxicity that further proved beneficial for drug delivery applications. Koshani et al.
2021 presented the lipophilic drug delivery applications via natural emulgel obtained from
dialdehyde cellulose crosslinked with chitosan. The delivery of the lipophilic compound β-
carotene occurred using an embedded oil-in-water emulsion. The lipophilic β-carotene loaded
in the oil-phase showed 20% release in stomach after passing safely through the oral cavity.
Furthermore, 50% of β-carotene released in the intestines after 4h in the presence of emulgel
thereby indicating its application as oral delivery vehicle [274].
Clinical trials on Polysaccharide based drug delivery systems
Riedl et al. (1993) performed phase I clinical trials on the dextran-conjugated doxorubicin on
13 patients at a starting dose of 40 mg/m2 that led to the development of WHO grade IV
thrombocytopenia in 2/2 patients. Whereas, the WHO grade IV hepatotoxicity, and WHO grade
III cardiotoxicity appeared in patients with preexisting heart ailments. Lowering of the dose to
20 mg/m2 further decreased the existence of thrombocytopenia. However, the hepatotoxicity
persisted. Further reducing the dose to 12.5 mg/m2 caused marked reduction in the malignancy
fibrous histiocytoma for up to 4 months [275]. Soepenberg et al. (2005) performed phase I
clinical trials on DE-310, a camptothecin analog, and a carboxymethyldextran polyalcohol
carrier, in the patients with advanced solid tumors. The DE-301 drug delivery system
reportedly released DX-8951 drug slowly, while maintaining a sustained release. This
prolonged the drug exposure at its deliberated tumor site. Some patients showed partial-to-
complete remission of metastatic adenocarcinoma at dose 7.5 mg/ m2 [276]. Kim et al. (2006)
conducted phase II clinical trials on holmium 166/ chitosan complex for the effective treatment
of hepatocellular carcinoma. The percutaneous administration of the nanosystem caused a
complete tumor necrosis in 31/40 patients after 2 months period. However, a long-term follow-
up period indicated recurring of tumor in 28 patients. The polysaccharide based drug delivery
system proved beneficial for the treatment of small hepatocellular carcinoma by performing
local ablative procedure [277]. Pinnix et al. (2012) performed single-blind randomized phase
III clinical trials on topical hyaluronic acid after adjuvant radiotherapy for breast cancer. The
topical hyaluronic based gel did not proved effective for the treatment of grade 2 dermatitis
following radiotherapy [278]. Pritchard et al. (2016) reported phase II clinical trials on
oligosaccharide polymer therapy for modification of mucus barrier of COPD. The binding of
the oligosaccharide caused alteration in the surface charge, porosity, and 3-D mucin networks
in sputum of patients with cystic fibrosis, and its inhalation in patients caused effective
deposition in lungs and altered the viscoelasticity of the cystic fibrosis sputum [279].
Conclusion and Future perspectives
The polysaccharide based drug-delivery vehicles traversed a long journey for the controlled
release of pharmaceuticals at the target site with minimized ensuing side effects caused by the
customary delivery vectors. The biodegradability and trivial immunogenicity of the
polysaccharide-based drug delivery vehicles makes them the material of the future. The
controlled release profile offers improved drug pharmacokinetics thereby leading to
ameliorated local action, and effectivity. As such, the polysaccharides present the first-in-class
drug delivery system ‘Novochizol’ for the delivery of COVID-19 drugs. The system comprises
of biodegradable and biocompatible chitosan nanoparticles that strongly adhere to the lung
epithelium and offers a sustained drug-release. Similarly, the recent discovery of helical V-
amylose as advanced drug delivery system further validated the precedence of polysaccharides
as drug delivery vehicles. The helical morphology of V-amylose offers drug encapsulation in
the helix groves while interacting with the functional head groups. This system proved highly
advantageous for the delivery of indomethacin, diclofenac, and aspirin with enhanced gastric
tolerance. The conjugation of polysaccharides with metal nanoparticles with unique
physicochemical profile in the form of optical, electronic, magnetic, and surface properties
provide nanoprobes that present a sturdy candidature in bioimaging of the effected site, while
delivery the cargo pharmaceutical at the same time. The development of multicompartmental
microspheres based on polysaccharides revolutionized bioengineering and multi-drug delivery
with high precision. The coaxial electrospun fiber membranes based on polysaccharides
transformed the plant grafting techniques and effectively managed the plant tissue injury due
to their high mechanical and tensile strength. The biological phenomenon such as wood
degradation by brown-rot fungi led to the development of bioinspired, core shell cellulose
microparticles for the light-triggered release of anticancer drugs. Furthermore, the stabilization
of Pickering emulsions by chemically modified polysaccharides play a significant role in the
lipophilic drug delivery applications, and targeted delivery of various pharmaceuticals and
bioactive ingredients.
Despite several benefits, the polysaccharide-based formulations suffer limitations such as
susceptibility towards microbial contamination, uncontrolled hydration rate, and reduced
viscosity during storage. To overcome these limitations, it mandates the modification of natural
polysaccharides via cross-linking, grafting, and blending with natural and synthetic polymers
to improve their physicochemical profile. Similarly, the limited knowledge about the
mechanism of drug release by polysaccharides, and permeation enhancement decelerates the
progressive developments in polysaccharide-based drug delivery systems.
References
[1] Z. Liu, Y. Jiao, Y. Wang, C. Zhou and Z.J. Zhang, Polysaccharides-based nanoparticles as
drug delivery systems, Adv. Drug Deliv. Rev. 60 (2008) 1650-1662.
[2] K. Janes, P. Calvo, M.J. Alonso, Polysaccharide colloidal particles as delivery systems for
macromolecules, Adv. Drug Deliv. Rev. 47 (2001) 83-97.
[3] A.F. Metaxa, E. Vrontaki, E.K. Efthimiadou, T. Mavromoustakos (2021) Drug Delivery
Systems Based on Modified Polysaccharides: Synthesis and Characterization. In: T.
Mavromoustakos, A.G. Tzakos, S. Durdagi (eds) Supramolecules in Drug Discovery and Drug
Delivery. Methods in Molecular Biology, vol 2207. Humana, New York, NY.
https://doi.org/10.1007/978-1-0716-0920-0_12.
[5] Y. Zheng, Q. Xie, H. Wang, Y. Hu, B. Ren, X. Li, Recent advances in plant polysaccharide-
mediated nano drug delivery systems, Int. J. Biol. Macromol. 165 (2020) 2668-2683.
[6] Y. Sun, X. Jing, X. Ma, Y. Feng, H. Hu, Versatile types of polysaccharide-based drug
delivery systems: From strategic design to cancer therapy, Int. J. Mol. Sci. (MDPI). 21 (2020)
Article 9159.
[7] A.K. Nayak, M.S. Hasnain, A.K. Dhara D. Pal (2021) Plant Polysaccharides in
Pharmaceutical Applications. In: D. Pal, A.K. Nayak (eds) Bioactive Natural Products for
Pharmaceutical Applications. Advanced Structured Materials, vol 140. Springer, Cham.
https://doi.org/10.1007/978-3-030-54027-2_3.
[8] K. Park, J.-H. Kim, Y.S. Nam, et al., Effect of polymer molecular weight on the tumor
targeting characteristics of self-assembled glycol chitosan nanoparticles, J. Control Release.
122 (2007) 305-314.
[12] S.H. Lee, R. Bajracharya, J.Y. Min, J-W Han, B.J. Park, H-K Han, Strategic approaches
for colon targeted drug delivery: An overview of recent advancements, Pharmaceutics (MDPI).
12 (2020) Article 68.
[13] M. Naeem, U.A. Awan, F. Subhan, J. Cao, S.P. Hlaing, J. Lee, E. Im, Y. Jung, J-W Yoo,
Advances in colon-targeted nano-drug delivery systems: challenges and solutions, Arch.
Pharmacal. Res. 43 (2020) 153-169.
[14] B. Layek, S. Mandal, Natural polysaccharides for controlled delivery of oral therapeutics:
a recent update, Carbohyd. Polym. 230 (2020) 115617.
[15] M. Thanou, J. Verhoef and H.J. Junginger, Oral drug absorption enhancement by chitosan
and its derivatives, Adv. Drug Deliv. Rev. 52 (2001) 117-126.
[16] N.G. Schipper, S. Olsson, J.A. Hoogstraate, A.G. deBoer, K.M. Vårum and P. Artursson,
Chitosans as absorption enhancers for poorly absorbable drugs 2: mechanism of absorption
enhancement, J. Pharm. Res. 14 (1997) 923-929.
[17] A. Almeida, M. Araujo, R.N. Carballal, F. Andrade, H. Goncalves, R.L. Reis, M. Lucio,
S. Schwartz, B. Sarmento, Novel amphiphillic chitosan micelles as carriers for hydrophobic
anticancer drugs, Mater. Sci. Eng. C. 112 (2020) Article 110920.
[19] S. Peers, A. Montembault, C. Ladaviere, Chitosan hydrogels for sustained drug delivery,
J. Control. Release. 326 (2020) 150-163.
[20] Z.A. Raza, S. Khalil, A. Ayub, I.M. Banat, Recent developments in chitosan encapsulation
of various active ingredients for multifunctional applications, Carbohyd. Res. 492 (2020)
Article 108004.
[21] V.M. Platt and F.C. Szoka Jr, Anticancer therapeutics: targeting macromolecules and
nanocarriers to hyaluronan or CD44, a hyaluronan receptor, Mol. Pharm. 5 (2008) 474-486.
[23] R. Liu, R. Zuo, G.A. Hudalla, Harnessing molecular recognition for localized drug
delivery, . Adv. Drug Deliv. Rev. 170 (2021) 238-260.
[24] A. Bianchera, R. Bettini, Polysaccharide nanoparticles for oral controlled drug delivery:
the role of drug-polymer and interpolymer interactions, Expert Opin. Drug Deliv. 17 (2020)
1345-1359.
[26] H.Q. Song, Y. Fan, Y. Hu, G. Cheng, F-J Xu, Polysaccharide-peptide conjugates: A
versatile material platform for biomedical applications, Adv. Funct. Mater. 31 (2021) Article
2005978.
[28] D. Wu, L. Zhu, Y. Li, X. Zhang, S. Xu, G. Yang, T. Delair, Chitosan-based colloidal
polyelectrolyte complexes for drug delivery: A review, Carbohyd. Polym. 238 (2020) Article
116126.
[30] V.M. Leitner, G.F. Walker, A. Bernkop-Schnürch, Thiolated polymers: evidence for the
formation of disulphide bonds with mucus glycoproteins, Eur. J. Pharmaceutic
Biopharmaceutic. 56 (2003), pp. 207-214.
[31] R.H. Marchessault, F. Ravenelle and X.X. Zhu, Polysaccharides for drug delivery and
pharmaceutical applications, ACS Symposium Series (2006).
[32] A.K. Sharma, P. Prasher, A.A. Aljabali, V. Mishra, H. Gandhi, S. Kumar, S. Mutalik, D.K.
Chellappan, M.M. Tambuwala, K. Dua, D.N. Kapoor, Emerging era of “somes”:
polymersomes as versatile drug delivery carrier for cancer diagnostics and therapy. Drug Deliv.
Transl. Res. 10 (2020) 1171-1190.
[33] J.-H. Kim, Y.-S. Kim, S. Kim, et al., Hydrophobically modified glycol chitosan
nanoparticles as carriers for paclitaxel, J. Control. Release. 111 (2006), pp. 228-234.
[35] X. Du, S. Yin, L. Xu, J. Ma, H. Yu, G. Wang, J. Li, Polylysine and cysteine functionalized
chitosan nanoparticle as an efficient platform for oral delivery of paclitaxel, Carbohyd. Polym.
229 (2020) 115484.
[36] N. Feng, H. Wu, Y. Xie, Q. Wu, A novel drug delivery system obtained from hydrophobic
modified amphiphilic polymers by Maillard reaction, Int. J. Biol. Macromol. 157 (2020) 146-
150.
[37] M. Zembala, Electrokinetics of heterogeneous interfaces, Adv. Colloid Interface Sci. 112
(2004) 59-92.
[38] A.A. Kamel, C.M. Ma, M.S. El-Aasser, F.J. Micale, S.A. Vanderhoff, Concerning the
origin of charge at the polystyrene particle/water interface, J. Disp. Sci. Technol. 2 (1981) 315-
330.
[40] S. Bhattacharjee, DLS and zeta potential–what they are and what they are not?, J. Control.
Release. 235 (2016) 337-351.
[41] R.N. Wijesena, N.D. Tissera, V.W.S.G. Rathnayaka, R.M. Silva, K.M.N. Silva, Colloidal
stability of chitin nanofibers in aqueous systems: Effect of pH, ionic strength, temperature and
concentration, Carbohyd. Polym. 235 (2020) Article 116024.
[42] F. Gambinossi, S.E. Mylon, J.K. Ferri, Aggregation kinetics and colloidal stability of
functionalized nanoparticles, Adv. Colloid Interface Sci. 222 (2015) 332-349.
[43] P. Vera, V. Gallardo, J. Salcedo, M.A. Ruiz and A.V. Delgado, Electrokinetics and
stability of a cellulose acetate phthalate latex, J. Appl. Polymer Sci. 65 (1997) 2721-2726.
[45] X-Y Zhang, Q-S Wang, Z-X Wu, D-P Tao, An experimental study on size distribution
and zeta potential of bulk cavitation nanobubbles, Int. J. Mineral Metallurgy mater. 27 (2020)
152-161.
[46] B-L Vinh, N.M.N. Le, I. Nazir, B. Matuszczak, A.B. Schnurch, Chitosan based micelle
with zeta potential changing property for effective mucosal drug delivery, Int. J. Biol.
Macromol. 133 (2019) 647-655.
[48] G. Han, S. Liu, Z. Pan, Y. Lin, S. Ding, L. Li, B. Luo, Y. Jiao, C. Zhou, Sulfonated chitosan
and phosphorylated chitosan coated polylactide membrane by polydopamine-assisting for the
growth and osteogenic differentiation of MC3T3-E1s, Carbohyd. Polym. 229 (2020) Article
115517.
[49] A.J. Bernkop-Schnürch, Strategies to overcome the polycation dilemma in drug delivery,
Adv. Drug Deliv. Rev. 136 (2018) 62-72.
[50] L. Wu, W. Shan, Z. Zhang, Y.J. Huang, Engineering nanomaterials to overcome the
mucosal barrier by modulating surface properties, Adv. Drug Deliv. Rev. 124 (2018) 150-163.
[51] A. George, P.A. Shah, P.S. Srivastava, Natural biodegradable polymers based nano-
formulations for drug delivery: A review, Int. J. Pharm. 561 (2019) 244-264.
[52] A. Gagliardi, E. Giuliano, E. Venkateshwararao, M. Fresta, S. Bulotta, V. Awasthi, D.
Cosco, Biodegradable polymeric nanoparticles for drug delivery to solid tumors, Front.
Pharmacol. 12 (2021) Article 601626.
[53] M.S. Hasnain, et al. (2020) Biopolymers for Drug Delivery. In: A. Nayak, M. Hasnain
(eds) Advanced Biopolymeric Systems for Drug Delivery. Advances in Material Research and
Technology. Springer, Cham. https://doi.org/10.1007/978-3-030-46923-8_1.
[55] T.G. Barclay, C.M. Day, N. Petrovsky, S. Garg, Review of polysaccharide particle-based
functional drug delivery, Carbohyd. Polym. 221 (2019) 94-112.
[57] Y. Yu, TT-Ergas, Y. Zhu, G. Fittolani, V. Bardoni, A. Singhal, R.J. Fair, A. Grafmuller,
P.H. Seeberger, M. Delbianco, Sysematic hydrogen-bond manipulations to establish
polysaccharide structure-property relationship, Angew Chem. 58 (2019) 13127-13132.
[59] G. Machell, G.N. Richards, 906. The alkaline degradation of polysaccharides. Part II. The
alkali-stable residue from the action of sodium hydroxide on cellulose, J. Chem. Soc. 1957,
4500-4506.
[61] Y. Ding, Y. Yan, Y. Peng, D. Chen, J. Mi, L. Lu, Q. Luo, X. Li, X. Zeng, Y. Cao, In
vitro digestion under simulated saliva, gastric and small intestinal conditions and fermentation
by human gut microbiota of polysaccharides from the fruits of Lycium barbarum. Int. J. Biol.
Macromol. 125 (2019) 751-760.
[62] T. Di, G. Chen, Y. Sun, S. Ou, X. Zeng, H. Ye, In vitro digestion by saliva, simulated
gastric and small intestinal juices and fermentation by human fecal microbiota of sulfated
polysaccharides from Gracilaria rubra, J. Funct. Food. 40 (2018) 18-27.
[63] X. Li, R. Guo, X. Wu, X. Liu, L. Ai, Y. Sheng, Z. Song, Y. Wu, Dynamic digestion of
tamarind seed polysaccharide: Indigestibility in gastrointestinal simulations and gut microbiota
changes in vitro, Carbohyd. Polym. 239 (2020) Article 116194.
[64] J.M. Krochta, J.S. Hudson, S.J. Tillin (1988) Kinetics of alkaline thermochemical
degradation of polysaccharides to organic acids, ACS Publications.
[65] N.G. Kotla, S. Rana, G. Sivaraman, O. Sunnapu, P.K. Vemula, A. Pandit, Y. Rochev,
Bioresponsive drug delivery systems in intestinal inflammation: State-of-the-art and future
perspectives, Adv. Drug Deliv. Rev. 146 (2019) 248-266.
[66] P. Prasher, R. Fatima, M. Sharma, Therapeutic delivery with V-amylose, Drug Dev. Res.
(2021) DOI: 10.1002/ddr.21804.
[67] B. Stone, B. Svensson, M. Collins, R. Rastall (2008) Polysaccharide Degradation. In: B.O.
Fraser-Reid, K. Tatsuta, J. Thiem (eds) Glycoscience. Springer, Berlin, Heidelberg.
https://doi.org/10.1007/978-3-540-30429-6_60.
[69] X. Lu, N. Li, X. Qiao, Z. Qiu, P. Liu, Effects of thermal treatment on polysaccharide
degradation during black garlic processing, LWT, 95 (2018) 223-229.
[71] J. Fu, J. Ji, W. Yuan, J.J.B. Shen, Construction of anti-adhesive and antibacterial
multilayer films via layer-by-layer assembly of heparin and chitosan, Biomater. 26 (2005)
6684-6692.
[72] M.G. Carneiro-da-Cunha, M.A. Cerqueira, B.W. Souza, S. Carvalho, M.A. Quintas, J.A.
Teixeira, A.A. Vicente, Physical and thermal properties of a chitosan/alginate nanolayered PET
film, Carbohyd. Polymer, 82 (2010) 153-159.
[75] F. Shi, X. Tian, D.J. McClements, Y. Chang, J. Shen, C. Xue, Influence of molecular
weight of an anionic marine polysaccharide (sulfated fucan) on the stability and digestibility
of multilayer emulsions: Establishment of structure-function relationships. Food Hydrocolloid.
113 (2021) Article 106418.
[76] S. Iqbal, Z. Xu, H. Huang, X.D. Chen, Structuring of water-in-oil emulsions using
controlled aggregation of polysaccharide in aqueous phases, J. Food Eng. 258 (2019) 34-44.
[77] M. Farid-Ul-Haq, M.A. Hussain, M.T. Haseeb, M.U. Ashraf, S.Z. Hussain, T. Tabassum,
I. Hussain, M. Sher, S.N.A. Bukhari, M.N. Hassan, A stimuli-responsive, superporous and non-
toxic smart hydrogel from seeds of mugwort (Artemisia vulgaris): stimuli responsive
swelling/deswelling, intelligent drug delivery and enhanced aceclofenac bioavailability. RSC
Adv. 10 (2020) 19832-19843.
[78] M.A. Hussain, A.I. Rana, M.T. Haseeb, G. Muhammad, L. Kiran, Citric acid cross-linked
glucuronoxylans: A pH-sensitive polysaccharide material for responsive swelling-
deswelling vs various biomimetic stimuli and zero-order drug release. J. Drug Deliv. Sci.
Technol. 55 (2020) Article 101470.
[81] H-G Fu, Y. Chen, Q. Yu, Y. Liu, Polysaccharide-based nanoparticles for two-step
responsive release of antitumor drug, ACS Med. Chem. Lett. 11 (2020) 1191-1195.
[84] B. Sasa, P. Odon, S. Stane, K. Julijana, Analysis of surface properties of cellulose ethers
and drug release from their matrix tablets, Eur. J. Pharm. Sci. 27 (2006) 375-383.
[85] N. Raghav, N. Mor, R.D. Gupta, R. Kaur, M.R. Sharma, P. Arya, Some
cetyltrimethylammonium bromide modified polysaccharide supports as sustained release
systems for curcumin. Int. J. Biol. Macromol. 154 (2020) 361-370.
[86] R. Zhang, M. Tang, A. Bowyer, R. Eisenthal, J.J.B. Hubble, A novel pH-and ionic-
strength-sensitive carboxy methyl dextran hydrogel, Biomater. 26 (2005) 4677-4683.
[88] Y. Guo, J. Sun, S. Bai, X. Wu, pH-Sensitive performance of dextran-poly (acrylic acid)
copolymer and its application in controlled in vitro release of ibuprofen, Int. J. Polym. Mater.
Polym. Biomater. 66 (2017) 900-906.
[90] N. Lin, A. Dufresne, Nanocellulose in biomedicine: Current status and future prospect,
Eur. Polym. J. 59 (2014) 302-325.
[91] H. Ni, S. Zeng, J. Wu, X. Cheng, T. Luo, W. Wang, W. Zeng, Y. Chen, Cellulose
nanowhiskers: preparation, characterization and cytotoxicity evaluation, Biomed. Mater. Eng.
22 (2012) 121-127.
[98] P.J. Eggenhuizen, B.H. Ng, J.D. Ooi, Treg enhancing therapies to treat autoimmune
diseases, Int. J. Mol. Sci. (MDPI) 21 (2020) Article 7015.
[99] E.B. Okeke, J.E. Uzonna, Th epivotal role of regulatory T cells in the regulation of innate
immune cells, Front. Immunol. 10 (2019) Article 680.
[100] N. Gupta, A.K. Jangid, D. Pooja, H. Kulhari, Inulin: A novel and stretchy polysaccharide
tool for biomedical and nutritional applications, Int. J. Biol. Macromol. 132 (2019) 852-863.
[101] K. Sampathkumar, S.C.J. Loo, Targeted gastrointestinal delivery of nutraceuticals with
polysaccharide-based coatings, Macromol. Biosci. 18 (2018) Article 1700363.
[102] E.V. Lopez, A.L. Alvarez, J.B. Mendez, F.J.O. Espinar, Cellulose-polysaccharide film-
coating of cyclodextrin based pellets for controlled drug release, J. Drug Deliv. Sci. Technol.
42 (2017) 273-283.
[103] S. Gao, G. Tang, D. Hua, R. Xiong, J. Han, S. Jiang, Q. Zhang, C. Huang, Stimuli-
responsive bio-based polymeric systems and their applications, J. Mater. Chem. B. 7 (2019)
709-729.
[104] T. Thambi, D.G. You, H.S. Han, V.G. Deepagan, S.M. Jeon, Y.D. Suh, K.Y. Choi, K.
Kim, I.C. Kwon, G-R Yi, J.Y. Lee, D.S. Lee, J.H. Park, Bioreducible carboxymethyl dextran
nanoparticles for tumor-targeted drug delivery, Adv. Healthcare Med. 11 (2014) 1829-1838.
[106] J.M. Shin, S-H Kim, T. Thambi, D.G. You, J. Jeon, J.O. Lee, B.Y. Chung, D-G Jo, J.H.
Park, A hyaluronic acid–methotrexate conjugate for targeted therapy of rheumatoid arthritis,
Chem. Commun. 50 (2014) 7632-7635.
[107] H. Jung, K.M. Park, J-A Yang, E.J. Oh, D-W Lee, K. Park, S.H. Ryu, S.K. Hahn, K.
Kim, Theranostic systems assembled in situ on demand by host-guest chemistry, Biomater. 32
(2011) 7687-7694.
[108] Q. Qu, J. Zhang, X. Chen, H. Ravanbakhsh, G. Tang, R. Xiong, B.B. Manshian, S.J.
Soenen, F. Sauvage, K. Braeckmans, S.C.D. Smedt, C. Huang, Triggered release from cellulose
microparticles inspired by wood degradation by fungi, ACS Sustain. Chem. Eng. 9 (2021) 387-
397.
[109] G. Tang, R. Xiong, D. Lv, R.X. Xu, K. Braeckmans, C. Huang, S.C.D. Smedt, Gas-
shearing fabrication of multicompartmental-microspheres: A one-step and oil-free approach,
Adv. Sci. 6 (2019) Article 1802342.
[110] Z. Guo, G. Tang, Y. Zhou, L. Shuwu, H. Hou, Z. Chen, J. Chen, C. Hu, F. Wang, S.C.D.
Smedt, R. Xiong, C. Huang, Fabrication of sustained-release CA-PU coaxial electrospun fiber
membranes for plant grafting application, Carbohyd. Polym. 169 (2017) 198-205.
[111] S. Zhao, X. Yu, Y. Qian, W. Chen, J. Shen, Multifunctional magnetic iron oxide
nanoparticles: an advanced platform for cancer theranostics, Theranostics. 10 (2020) 6278-
6309.
[112] H-W Cheng, H-Y Tsao, C-S Chiang, S-Y Chen, Advances in nanoparticle-mediated
cancer immune-theranostics, Adv. Healthcare Mater. 10 (2021) Article 2001451.
[113] R.G. Thomas, M. Muthiah, M.J. Moon, I-K Park, Y.Y. Jeong, SPION loaded poly(L-
lysine)/hyaluronic acid micelles as MR contrast agent and gene delivery vehicle for cancer
theranostics. Macromol. Res. 25 (2017) 446-451.
[114] M.H. El-Dakdouki, D.C. Zhu, K. El-Boubbou, M. Kamat, J. Chen, W. Li, X. Huang,
Development of Multifunctional Hyaluronan-Coated Nanoparticles for Imaging and Drug
Delivery to Cancer Cells, Biomacromol. 13 (2017) 1144-1151.
[115] R. Choi, J. Yang, J. Choi, E-K Lim, E. Kim, J-S Suh, Y-M Huh, S. Haam, Thiolated
Dextran-Coated Gold Nanorods for Photothermal Ablation of Inflammatory Macrophages,
Langmuir. 26 (2010) 17520-17527.
[116] C. Yue, P. Liu, M. Zheng, P. Zhao, Y. Wang, Y. Ma, L. Cai, IR-780 dye loaded tumor
targeting theranostic nanoparticles for NIR imaging and photothermal therapy, Biomater. 34
(2013) 6853-6861.
[120] M.A. Otache, R.U. Duru, O. Achugasim, O.J. Abayeh, Advances in the modification of
starch via esterification for enhanced properties, J. Polymer Environ. 29 (2021) 1365-1379.
[122] A.S. Volokhova, K.J. Edgar, J.B. Matson, Polysaccharide-containing block copolymers:
synthesis and applications, Mater. Chem. Front. 4 (2020) 99-112.
[124] A.J. Kerchove, M. Elimelech, Formation of Polysaccharide Gel Layers in the Presence
of Ca2+ and K+ Ions: Measurements and Mechanisms, Biomacromol. 8 (2007) 113-121.
[125] A. Chowhan, T.K. Giri, Polysaccharide as renewable responsive biopolymer for in situ
gel in the delivery of drug through ocular route, Int. J. Biol. Macromol. 150 (2020) 559-572.
[126] H. Zhong, X. Gao, C. Cheng, C. Liu, Q. Wang, X. Han, The structural characteristics of
seaweed polysaccharides and their applications in gel drug delivery systems, Marine Drug
MDPI). 18 (2020) Article 658.
[132] H.H. Tonnesen, J. Karlsen, Alginate in drug delivery systems, Drug Dev. Indus. Pharm.
28 (2002) 621-630.
[133] J. Li, C. Cai, J. Li, T. Sun, L. Wang, H. Wu, G. Yu, Chitosan-based nanomaterials for
drug delivery, Molecules (MDPI). 23 (2018) Article 2661.
[134] L. Szabo, S.G. Lemaire, C. Wandrey, Strategies to functionalize the anionic biopolymer
Na-alginate without restricting its polyelectrolyte properties, Polymers (MDPI). 12 (2020)
Article 919.
[135] E.M.P. Quito, R.R. Caro, M.D. Veiga, Carrageenan: Drug delivery systems and other
biomedical applications, Marine Drug (MDPI). 18 (2020) Article 583.
[136] J. Guan, L. Li, S. Mao, Applications of carrageenan in advanced drug delivery. Seaweed
polysaccharides. Elsevier. 2017.
[137] J.L. Jiang, W-Z Zhang, W-X Ni, J-W Shao, Insight on structure-property relationships
of carrageenan from marine red algal: A review, Carbohyd. Polym. 257 (2021) Article 117642.
[143] A. Aguilera-Garrido, T.C. Santaella, Y. Yang, F.G. Gonzalez, M.J.G. Ruiz, J.A.M.
Bolivar, J.A.H. Terriza, M.A.C. Vilchez, J.M. Valderrama, Aplications of serum albumins in
delivery systems: Differences in interfacial behavior and interacting abilities with
polysaccharides, Adv. Colloid. Interface Sci. 290 (2021) Article 102365.
[146] J.N. Putro, S. Ismadji, C. Gunarto, F.E. Soetaredjo, Y.H. Ju, Effect of natural and
synthetic surfactants on polysaccharide nanoparticles: Hydrophobic drug loading, release , and
cytotoxic studies, Colloid Surf A. 578 (2019) Article 123618.
[148] P.F. Cui, W-R Zhuang, X. Hu, L. Xing, R-Y Yu, J-B Qiao, Y-J He, F. Li, D. Ling, H-L
Jiang, A new strategy for hydrophobic drug delivery using a hydrophilic polymer equipped
with stacking units, Chem. Commun. 54 (2018) 8218-8221.
[149] C.A. Lorenzo, B.B. Fernandez, A.M. Puga, A. Concheiro, Crosslinked ionic
polysaccharides for stimuli-sensitive drug delivery, Adv. Drug. Dev. Rev. 65 (2013) 1148-
1171.
[150] Y. Wu, X. Zhang, H. Li, P. Deng, H. Li, T. He, J. Rong, J. Zhao, Z. Liu, A core/ shell
stabilized polysaccharide-based nanoparticle with intracellular environment-sensitive drug
delivery for breast cancer therapy, J. Mater. Chem. B. 6 (2018) 6646-6659.
[151] Y. Yu, T.T. Ergas, Y. Zhu, G. Fittolani, V. Bordoni, A. Singhal, R.J. Fair, A. Grafmuller,
P.H. Seeberger, M. Delbianco, Systematic Hydrogen‐ Bond Manipulations To Establish
Polysaccharide Structure–Property Correlations, Angew. Chem. 58 (2019) 13127-13132.
[152] F. Zhu, J. Rodriguez, T. Yang, I. Kevlishvili, E. Miller, D. Yi, S. Neill, M.J. Rourke, P.
Liu, M.A. Walczak, Glycosyl Cross-Coupling of Anomeric Nucleophiles: Scope, Mechanism,
and Applications in the Synthesis of Aryl C-Glycosides, J. Am. Chem. Soc. 139 (2017) 17908-
17922.
[153] P.O. Adero, H. Amarasekara, P. Wen, L. Bohe, D. Crich, The Experimental Evidence in
Support of Glycosylation Mechanisms at the SN1-SN2 Interface, Chem. Rev. 118 (2018) 8242-
8284.
[154] T. Hansen, L. Lebedel, et al., Defining the SN1 Side of Glycosylation Reactions:
Stereoselectivity of Glycopyranosyl Cations, ACS Cent. Sci. 5 (2019) 781-788.
[156] J.A.C. Romero, S.A. Tobbacco, K.A. Woerpel, Stereochemical Reversal of Nucleophilic
Substitution Reactions Depending upon Substituent: Reactions of Heteroatom-Substituted
Six-Membered-Ring Oxocarbenium Ions through Pseudoaxial Conformers, J. Am. Chem. Soc.
122 (2000) 168-169.
[157] E.C. Rodriguez, L.A. Marcaurelle, C.R. Bertozzi, Aminooxy-, Hydrazide-, and
Thiosemicarbazide-Functionalized Saccharides: Versatile Reagents for Glycoconjugate
Synthesis. J. Org. Chem. 63 (1998) 7134-7135.
[158] K.C.K. Swamy, N.N.B. Kumar, E. Balaraman, K.V.P.P. Kumar, Mitsunobu and related
reactions: Advances and applications, Chem. Rev. 109 (2009) 2551-2651.
[159] S. Fletcher, The Mitsunobu reaction in the 21st century, Org. Chem. Front. 2 (2015) 739-
752.
[164] T. Breton, G. Bashiardes, J-M Leger, K.B. Kokoh, Selective Oxidation of Unprotected
Carbohydrates to Aldehyde Analogues by Using TEMPO Salts, Eur. J. Org. Chem. 10 (2007)
1567-1570.
[166] P.S. Chang, J.F. Robyt, Oxidation of Primary Alcohol Groups of Naturally Occurring
Polysaccharides with 2,2,6,6-Tetramethyl-1-Piperidine Oxoammonium Ion, J. Carbohyd.
Chem. 15 (1996) 819-830.
[171] C. Chen, J-L Zhou, X. Han, F. Song, X-L Wang, Y-Z Wang, A prodrug strategy based
on chitosan for efficient intracellular anticancer drug delivery, Nanotechol. 25 (2014) 255101.
[172] C.K. Chen, P.K. Huang, W.C. Law, C.H. Chu, N.T. Chen, L.W. Lo, Biodegradable
Polymers for Gene-Delivery Applications, Int J Nanomedicine. 15 (2020) 2131-2150.
[173] N.N. Siddiqui, A. Aman, A. Silipo, S.A.U. Qader, A. Molinaro, Structural analysis and
characterization of dextran produced by wild and mutant strains of Leuconostoc mesenteroides,
Carbohyd. Polym. 99 (2014) 331-338.
[176] C. Longinotti, The use of hyaluronic acid based dressings to treat burns: A review, Burns
Trauma. 2 (2014) 162-168.
[182] O.P. Varghese, J. Liu, K. Sundaram, J. Hilborn, O.P. Oommen, Chondroitin sulfate
derived theranostic nanoparticles for targeted drug delivery, Biomater. Sci. 4 (2016) 1310-
1313.
[183] S. Arpicco, P. Milla, B. Stella, F. Dosio, Hyaluronic acid conjugates as vectors for the
active targeting of drugs, genes and nanocomposites in cancer treatment, Molecules (MDPI).
19 (2014) 3193-3230.
[184] C.I.C. Crucho, MT. Barros, Surfactant-free polymeric nanoparticles for biomedical
applications, Microsc. Microanal. 21 (2015) 74-75.
[185] X. Shen, J.L. Shamshina, P. Berton, G. Gurau, R.D. Rogers, Hydrogels based on
cellulose and chitin: fabrication, properties, and applications, Green Chem. 18 (2016) 53-75.
[186] R. Jayakumar, A. Nair, N.S. Rejinold, S. Maya, S.J.C.P. Nair, Doxorubicin-loaded pH-
responsive chitin nanogels for drug delivery to cancer cells, Carbohyd. Polym. 87 (2012) 2352-
2356.
[187] N.S. Rejinold, A. Nair, M. Sabitha, K. Chennazhi, H. Tamura, S.V. Nair, R. Jayakumar,
Synthesis, characterization and in vitro cytocompatibility studies of chitin nanogels for
biomedical applications, Carbohyd. Polym. 87 (2012) 943-949.
[192] W-L Song, K. Song, L-Z Fan, A Versatile Strategy toward Binary Three-Dimensional
Architectures Based on Engineering Graphene Aerogels with Porous Carbon Fabrics for
Supercapacitors, ACS Appl. Mater. Interface, 7 (2015) 4257-4264.
[193] J. Muller, A.C. Jakel, D. Schwarz, L. Aufinger, F.C. Simmel, Programming Diffusion
and Localization of DNA Signals in 3D-Printed DNA-Functionalized Hydrogels, Small. 16
(2020) Article 2001815.
[194] S.G. Kelmanovich, R. Parke-Houben, C.W. Frank, Competitive swelling forces and
interpolymer complexation in pH- and temperature-sensitive interpenetrating network
hydrogels, Soft Matter. 8 (2012) 8137-8148.
[195] D.G. Lim, E. Kang, S.H. Jeong, pH-dependent nanodiamonds enhance the mechanical
properties of 3D-printed hyaluronic acid nanocomposite hydrogels, J. Nanobiotechnol. 18
(2020) Article 88.
[197] L. Li, J. Ge, P.X. Ma, B. Guo, Injectable conducting interpenetrating polymer network
hydrogels from gelatin-graft-polyaniline and oxidized dextran with enhanced mechanical
properties, RSC Adv. 5 (2015) 92490-92498.
[198] T.T. Khong, O.A. Aarstad, G. Skjåk-Bræk, K.I. Draget, K.M. Vårum, Gelling Concept
Combining Chitosan and Alginate-Proof of Principle, Biomacromol. 14 (2013) 2765-2771.
[203] S. Kaity, A. Ghosh, Comparative bio-safety and in vivo evaluation of native or modified
locust bean gum-PVA IPN microspheres, Int. J. Biol. Macromol. 72 (2015) 883-893.
[204] V.K. Thakur, M.K. Thakur, Recent trends in hydrogels based on psyllium
polysaccharide: a review, J. Clean. Product. 82 (2014) 1-15.
[205] S.K. Nitta, K. Numata, Biopolymer-based nanoparticles for drug/gene delivery and tissue
engineering, Int. J. Mol. Sci. 14 (2013) 1629-1654.
[206] D.G. Fatouros, D.A. Lamprou, A.J. Urquhart, S.N. Yannopoulos, I.S. Vizirianakis, S.
Zhang, S. Koutsopoulos, Lipid-like self-assembling peptide nanovesicles for drug delivery,
ACS Appl. Mater. Interface. 6 (2014) 8184-8189.
[207] S.V. Lale, A. Kumar, S. Prasad, A.C. Bharti, V. Koul, Folic Acid and Trastuzumab
Functionalized Redox Responsive Polymersomes for Intracellular Doxorubicin Delivery in
Breast Cancer, Biomacromol. 16 (2015) 1736-1752.
[208] Y. Lu, K. Park, Polymeric micelles and alternative nanonized delivery vehicles for poorly
soluble drugs, Int. J. Pharm. 453 (2013) 198-214.
[210] M.R. Saboktakin, N.A. Tabar, R.M. Tabatabaie, A. Maharramov, M.A. Ramazanov,
Intelligent drug delivery systems based on modified chitosan nanoparticles, Lett. Org. Chem.
9 (2012) 56-70.
[211] D. Yadav, S. Suri, A. Chaudhary, M. Beg, V. Garg, M. Asif, A. Ahmad, Stimuli
responsive polymeric nanoparticles in regulated drug delivery for cancer, Polish J. Chem.
Technol. 14 (2012) 57-64.
[213] L. Li, K.M. Huh, Y-K Lee, S.Y. Kim, Biofunctional self-assembled nanoparticles of
folate-PEG-heparin/PBLA copolymers for targeted delivery of doxorubicin, J. Mater. Chem.
21 (2011) 15288-15297.
[216] Y. Tang, Y. Li, R. Xu, S. Li, H. Hu, C. Xiao, H. Wu, L. Zhu, J. Ming, Z.J.N. Chu, H.
Xu, X. Yang, Z. Li, Self-assembly of folic acid dextran conjugates for cancer chemotherapy,
Nanoscale. 10 (2018) 17265-17274.
[217] X. Zhang, R. Zhang, J. Huang, M. Luo, X. Chen, Y. Kang, J. Wu, Albumin enhances
PTX delivery ability of dextran NPs and therapeutic efficacy of PTX for colorectal cancer, J.
Mater. Chem. B. 7 (2019) 3537-3545.
[218] Q.S. Zhao, L.I. Hu, Z.D. Wang, Z.P. Li, W.A. Wen, J. Liu, Resveratrol-loaded folic acid-
grafted dextran stearate submicron particles exhibits enhanced antitumor efficacy in non-small
cell lung cancers, Mater. Sci. Eng. C. 72 (2017) 185-191.
[219] Y. Fang, H. Wang, H-J Dou, X. Fan, X-C Fei, L. Wang, S. Cheng, A. Janin, L. Wang,
W-L Zhao, Doxorubicin-loaded dextran-based nano-carriers for highly efficient inhibition of
lymphoma cell growth and synchronous reduction of cardiac toxicity, Int. J. Nanomed. 13
(2018) 5673-5683.
[220] T. Wang, J. Hou, C. Su, L. Zhao, Y. Shi, Hyaluronic acid-coated chitosan nanoparticles
induce ROS-mediated tumor cell apoptosis and enhance antitumor efficiency by targeted drug
delivery via CD44, J. Nanobiotechnol. 15 (2017) Article 7.
[221] Z. Yang, N. Sun, R. Cheng, C. Zhao, J. Liu, Z. Tian, Hybrid nanoparticles coated with
hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically
eliminate breast cancer stem cells, J. Mater Chem. B. 5 (2017) 6762-6775.
[222] P. Parashar, M. Rathor, M. Dwivedi, S.A. Saraf, Hyaluronic acid decorated naringenin
nanoparticles: appraisal of chemopreventive and curative potential for lung cancer,
Pharmaceutics. 10 (2018) Article 33.
[223] J. Li, X. Yang, P. Yang, F.J.I. Gao, Hyaluronic acid-conjugated silica nanoparticles for
breast cancer therapy, Inorg. Nanometal Chem. 47 (2017) 777-782.
[225] X-L Guo, X-X Kang, Y-Q Wang, X-J Zhang, C-J Li, Y. Liu, L-B Du, Co-delivery of
cisplatin and doxorubicin by covalently conjugating with polyamidoamine dendrimer for
enhanced synergistic cancer therapy, Acta Biomater. 84 (2019) 367-377.
[226] S.A. Shahin, R. Wang, S.I. Simargi, A. Contreras, L.P. Echavarria, L. Qu, W. Wen, T.
Dellinger, J. Unternaehrer, F.J.N.N. Tamanoi, Hyaluronic acid conjugated nanoparticle
delivery of siRNA against TWIST reduces tumor burden and enhances sensitivity to cisplatin
in ovarian cancer, Biol. Med. 14 (2018) 1381-1394.
[227] H. Jiang, X. Shi, X. Yu, X. He, Y. An, H. Lu, Hyaluronidase enzyme-responsive targeted
nanoparticles for effective delivery of 5-fluorouracil in colon cancer, Pharm. Res. 35 (2018)
73.
[228] M.A. Raja, M. Arif, C. Feng, S. Zeenat, C-G Liu, Synthesis and evaluation of pH-
sensitive, self-assembled chitosan-based nanoparticles as efficient doxorubicin carriers, J.
Biomater. Appl. 31 (2017) 1182-1195.
[229] J. Wu, C. Tang, C. Yin, Co-delivery of doxorubicin and interleukin-2 via chitosan based
nanoparticles for enhanced antitumor efficacy, Acta Biomater. 47 (2017) 81-90.
[230] B. Cheng, F. Gao, E. Maissy, P. Xu, Repurposing suramin for the treatment of breast
cancer lung metastasis with glycol chitosan-based nanoparticles, Acta Biomater. 84 (2019)
378-390.
[233] C. Zhang, G. Shi, J. Zhang, J. Niu, P. Huang, Z. Wang, Y. Wang, W. Wang, C. Li, D.
Kong, Redox-and light-responsive alginate nanoparticles as effective drug carriers for
combinational anticancer therapy, Nanoscale. 9 (2017) 3304-3314.
[235] A.A. Markeb, N.A. El-Maali, D.M. Sayed, A. Osama, M.A.Y. Abdel-Malek, A.H. Zaki,
M.E.A. Elwanis, J.J. Driscoll, Synthesis, Structural Characterization, and Preclinical Efficacy
of a Novel Paclitaxel-Loaded Alginate Nanoparticle for Breast Cancer Treatment, Int. J. Breast
Cancer. 2016 (2016) Article 7549372.
[236] N.P. Katuwavila, A.D.L.C. Perera, S.R. Samarakoon, P. Soysa, V. Karunaratne, G.A.J.
Amaratunga, D.N. Karunaratne, Chitosan-Alginate Nanoparticle System Efficiently Delivers
Doxorubicin to MCF-7 Cells, Journal of Nanomater. 2016 (2016) Article 3178904.
[237] L. Chen, M. Qian, L. Zhang, J. Xia, Y. Bao, J. Wang, L. Guo, Y. Li, Co-delivery of
doxorubicin and shRNA of Beclin1 by folate receptor targeted pullulan-based multifunctional
nanomicelles for combinational cancer therapy, RSC Advances. 8 (2018) 17710-17722.
[238] L. Huang, B. Chaurasiya, D. Wu, H. Wang, Y. Du, J. Tu, T. J. Webster, C. Sun, Versatile
redox-sensitive pullulan nanoparticles for enhanced liver targeting and efficient cancer therapy,
Nanomed. Nanotechnol. Biol. Med. 14 (2018) 1005-1017.
[242] Y. Cao, Y.F. Tan, Y.S. Wong, M.W.J. Liew, S. Venkatraman, Recent Advances in
Chitosan-Based Carriers for Gene Delivery, Marine Drug. 17 (2019) 381.
[247] M. Xie, Y. Xu, J. Liu, T. Zhang, H. Zhang, Preparation and Characterization of Folate
Targeting Magnetic Nanomedicine Loaded with Cisplatin, Journal of Nanomater. 2012 (2012)
1-9.
[248] A.L. Urzedo, M.C. Goncalves, M.H.M. Nascimento, C.B. Lombello, G. Nakazato, A.B.
Seabra, Cytotoxicity and Antibacterial Activity of Alginate Hydrogel Containing Nitric Oxide
Donor and Silver Nanoparticles for Topical Applications, ACS Biomater. Sci. Eng. 6 (2020)
2117-2134.
[250] T.W. Wong, G. Colombo, F. Sonvico, Pectin Matrix as Oral Drug Delivery Vehicle for
Colon Cancer Treatment, AAPS PharmSciTech. 12 (2011) 201-214.
[251] J-H Lee, Y-K Lee, Y-R Choi, J. Park, S.K. Jung, Y.H. Chang, The characterization,
selenylation and anti-inflammatory activity of pectic polysaccharides extracted from Ulmus
pumila L, Int. J. Biol. Macromol. 111 (2018) 311-318.
[252] R.C. Mundargi, S.A. Patil, S.A. Agnihotri, T.M. Aminabhavi, Development of
Polysaccharide-Based Colon Targeted Drug Delivery Systems for the Treatment of
Amoebiasis, Drug Dev. Indus. Pharm. 33 (2007) 255-264.
[253] A.M.G. Eldeen, H. Amer, W.A. Helmy, Cancer chemopreventive and anti-inflammatory
activities of chemically modified guar gum, Chem. Biol. Interact. 161 (2006) 229-240.
[256] R.M. Daudt, I.C.K. Guerreiro, F.C. Olivera, R.C.S. Thys, L.D.F. Marczak,
Determination of properties of pinhão starch: Analysis of its applicability as pharmaceutical
excipient, Industrial Crop Product. 52 (2014) 420-429.
[257] A.P. Almeida, S.R. Rojo, A.T. Serra, H-V Real, A-L Simplicio, I. Delgadilho, S.V.
Costa, L.B. Costa, Microencapsulation of oregano essential oil in starch-based materials using
supercritical fluid technology, Innovative Food Sci. Innovative Technol. 20 (2013) 140-145.
[258] K. Luo, H.J. Adra, Y-R Kim, Preparation of starch-based drug delivery system through
the self-assembly of short chain glucans and control of its release property, Carbohyd. Polym.
243 (2020) Article 116385.
[259] E. Alp, F. Dmkaci, E. Guven, M. Tenniswood, Starch nanoparticles for delivery of the
histone deacetylase inhibitor CG-1521 in breast cancer treatment. Int. J. Nanomed. 14 (2019)
1335-1346.
[260] Y. Shao, L. Li, X. Gu, L. Wang, S. Mao, Evaluation of chitosan-anionic polymers based
tablets for extended-release of highly water-soluble drugs, Asian J. Pharm. Sci. 10 (2015) 24-
30.
[262] R.A. Fitaihi, F.S. Aleanizy, S. Elsamaligy, H.A. Mahmoud, M.A. Bayomi, Role of
chitosan on controlling the characteristics and antifungal activity of bioadhesive fluconazole
vaginal tablets, Saudi Pharm. J. 26 (2018) 151–161.
[263] R.V. Pai, R.R. Jain, A.S. Bannalikar, M.D. Menon, Development and Evaluation of
Chitosan Microparticles Based Dry Powder Inhalation Formulations of Rifampicin and
Rifabutin. J. Aerosol Med. Pulmon. Drug Deliv. 29 (2016) 179-195.
[264] R.K. Dutta, S. Sahu, Development of diclofenac sodium loaded magnetic nanocarriers
of pectin interacted with chitosan for targeted and sustained drug delivery, Colloid. Surf. B. 97
(2012) 19–26.
[265] W. Zhou, L. Zou, C. Liu, R. Liang, J. Chen, Storage stability and skin permeation of
vitamin C liposomes improved by pectin coating, Colloid. Surf. B. 117 (2014) 330-337.
[266] M. Sharma, R. Malik, A. Verma, P. Dwivedi, G.S. Banoth, N. Pandey, J. Sarkar, P.R.
Mishra, A.K. Dwivedi, Folic Acid Conjugated Guar Gum Nanoparticles for Targeting
Methotrexate to Colon Cancer, J. Biomed. Nanotechnol. 9 (2013) 96-106.
[267] R. Kaur, T. Garg, B. Malik, U.D. Gupta, P. Gupta, G. Rath, A.K. Goyal, Development
and characterization of spray-dried porous nanoaggregates for pulmonary delivery of anti-
tubercular drugs, Drug Deliv. 23 (2016) 872–877.
[268] X. Yuan, R. Amarnath Praphakar, M.A. Munusamy, A.A. Alarfaj, S. Kumar, M. Rajan,
Mucoadhesive guargum hydrogel inter-connected chitosan-g-polycaprolactone micelles for
Rifampicin delivery, Carbohyd. Polym. 206 (2019) 1-10.
[270] K.S. Mikkonen, Strategies for structuring diverse emulsion systems by using wood
lignocellulose-derived stabilizers, Green Chem. 22 (2020) 1019-1037.
[272] B. Wang, H. Tian, D. Xiang, Stabilizing the oil-in-water emulsion using the mixtures of
Dendrobium Officinale polysaccharides and gum arabic or propylene glycol alginate,
Molecules (MDPI) 25 (2020) Article 759.
[274] R. Koshani, M. Tavakolian, T.G.M. Ven, Natural emulgel from dialdehyde cellulose for
lipophilic drug delivery, ACS Sustain. Chem. Eng. 9 (2021) 4487-4497.
[275] S.D. Riedl, E. Hausmann, H.D. Schick, R. Bender, et al., Phase I clinical and
pharmacokinetic trial of dextran conjugated doxorubicin (AD-70, DOX-OXD), Invest. New
Drug. 11 (1993) 187-195.
[277] J.K. Kim, K-H Han, J-T Lee, Y-H Paik, S.H. Ahn, J.D. Lee, K.S. Lee, C.Y. Chon, Y.M.
Moon, Long-term clinical outcome of phase IIb clinical trial of percutaneous injection with
holmium-166/chitosan complex (Milican) for the treatment of small hepatocellular carcinoma,
Clin. Cancer Res. 12 (2006) 543-548.
[278] C. Pinnix, G.H. Perkins, E.A. Strom, W. Tereffe, W. Woodward, J.L. Oh, L. Arriaga,
M.F. Munsll, P. Kelly, K.E. Hoffmann, B.D. Smith, T.A. Buchholz, T.K. Yu, Topical
hyaluronic acid vs. standard of care for the prevention of radiation dermatitis after adjuvant
radiotherapy for breast cancer: single-blind randomized phase III clinical trial, Int. J. Radiat.
Oncol. Biol. Phys. 83 (2012) 1089-1094.
[279] M.F. Pritchard, L.C. Powell, G.E. Menzies, P.D. Lewis, K. Hawkins, C. Wright, I. Doull,
T.R. Walsh, E. Onsoyen, E. Dessen, R. Myrvold, P.D. Rye, A.H. Myrset, H.N.E. Stevens, L.A.
Hodges, G.M. Gregor, J.B. Neilly, K.E. Hill, D.W. Thomas, A New Class of Safe
Oligosaccharide Polymer Therapy To Modify the Mucus Barrier of Chronic Respiratory
Disease, Mol. Pharm. 13 (2016) 863-872.
Revised Manuscript (Text with Changes marked) Click here to view linked References
O O
O O
HH
N N HN NH
R R R R
PS-based Therapeutic agents & Type of Cell types Model of study Polymer Advantages References
polymer targeting
Dextran COX-2 siRNA/ Passive Breast cancer cell In vitro and in vivo Biocompatible, pH stimuli- [215]
lines (MDA- responsive, down-regulate
MB231) Cyclooxygenase COX-2 genes in
tumor cells
Dextran- folic DOX/Active Breast cancer cell In vitro and in vivo Reducing the side effect of DOX, [216]
acid line (4T1) enhanced tumor growth
inhibition, and prolonged survival
rate
Dextran- Paclitaxel/ Passive Breast cancer cell In vitro and in vivo Enhanced cellular uptake, [216]
Indomethacin line (MCF-7) prolonged pharmacokinetics
(prolonged circulation time and
slower elimination), enhanced
tumor growth inhibition
Dextran- Paclitaxel/ Passive Colorectal cell line In vitro and in vivo Cellular proliferation enhanced [217]
albumin (CT-26) inhibition, inducing apoptosis,
Enhanced drug circulation times
Dextran-folic Resveratrol/ Active Lung cell line In vivo Enhanced cellular apoptosis and [218]
acid (A549) drug intake
Dextran-DOX Doxorubicin/ Passive Lymphoma cell In vitro and in vivo Reduced cytotoxicity (cardiac), [219]
lines enhanced apoptosis, enhanced
intracellular intake leading to
increased DOX concentration that
inhibits tumor growth
Hyaluronic 5-Fluorouracil/ Active Lung cell line In vitro Enhanced cell apoptosis and [220]
acid- Chitosan (A549) & cellular drug accumulation
liver cell line
(HepG2)
Hyaluronic Paclitaxel & curcumin /Active Breast cancer cell In vitro and in vivo Prolonged pharmacokinetics, [221]
acid- Poly- line (MCF-7) Enhanced cellular accumulation
lactic-co-
glycolic acid;
Hyaluronic Paclitaxel/Active Breast cancer cell In vitro Decreased the IC50 of paclitaxel [73]
acid- Poly- line significantly
lactic-co- (MDA-MB-231)
glycolic acid;
Hyaluronic Naringenin/Active Lung cancer cell In vitro and in vivo Enhanced cell apoptosis, [222]
acid- Poly-ε- line (A549) increased drug intake resulting in
caprolactone- inhibiting cancer growth
Chitosan
Hyaluronic Paclitaxel/Active Breast cancer cell In vitro and in vivo Enhanced cancer growth [223]
acid-Silica line (MCF-7) suppression, increased intake and
drug accumulation
Hyaluronic Doxorubicin & Gemcitabine Breast cancer cell In vitro and in vivo Dual modality drug delivery [224]
acid /Active lines system, enhanced inhibition to
(MDA-MB-231 cancer growth
& 4T1)
Hyaluronic Doxorubicin & Cisplatin /Active Breast cancer cell In vitro and in vivo Tumor growth inhibitor at low [225]
acid- lines drug concentrations
Polyamidoamin (MDA-MB231
e & MCF-7)
Hyaluronic TWIST- siRNAs (Basic helix- Ovarian cancer cell In vitro and in vivo Reduced cancer growth and cell [226]
acid-Silica- loop-helix transcription factors)/ line survival through selective
polyethyleneim Active (Ovcar-8) targeting
ine
Hyaluronic 5-Fluorouracil /Active Colon cancer cell In vitro and in vivo Tumor repression, high [227]
acid-Silica line (HT29) cytotoxicity and high
accumulation in the tumor
through selective targeting
Chitosan- N- DOX/Passive Breast cancer cell In vitro Enhanced cytotoxicity and [228]
acetyl histidine lines MCF-7 cellular uptake
and arginine
Chitosan- DOX & IL-2/Active Lung cancer cell In vitro and in vivo Increase IgG expression levels, [229]
Trimethyl and line (A549), Increased cytotoxic T
folate Liver cell line lymphocytes
(SMMC-7721)
and hepatoma cell
line
(H22)
Chitosan Suramin and Breast cancer cell In vitro and in vivo Inhibit Breast and lung metastasis [230]
DOX/ Passive line
(MDA-MB231)
Chitosan- DOX and IL17RB Breast cancer cell In vitro Decrease tumor cellular viability, [231]
Carboxymethyl siRNA/ Passive line inhibits growth, tumor cells
dextran (MDA-MB361) migration, and proliferation
Chitosan- Curcumin/Passive Colon cancer cell In vitro Enhanced cellular uptake, [232]
Hyaluronic line (HT29) reduced cellular proliferation, and
acid- high toxicity
sulfobutyl-
ether-β-
cyclodextrin
Alginate DOX/Passive Melanoma cell line In vitro and in vivo Inhibit tumor growth, higher [233]
(B16) cellular uptake, high cytotoxicity
Alginate Gold NPs (AuNPs) & cisplatin/ Colon cancer cell In vitro and in vivo Tumor growth high inhibition, [234]
Passive line (CT26) enhanced survival
Alginate Paclitaxel/Passive Different breast In vitro and in vivo Promote cell cycle arrest, induce [235]
cancer cell lines apoptosis and reduce the
bioavailability
Alginate- DOX/Active Breast cancer cell In vitro Inhibit tumor progression and [236]
chitosan line (MCF-7) cellular growth
Pullulan Floate & Cervical (Hela) In vitro and in vivo Tumor growth inhibition [237]
Polyethylenimine/Active and liver
(HepG2)
Pullulan- Paclitaxel/Active Liver cancer cell In vitro and in vivo Lower cytotoxicity due to [238]
Folate line selective targeting and prolonged
(SMMC-7721) release
Therefore, the polysaccharide offer a fascinating choice as polymeric material in drug delivery
due to biodegradability and biocompatibility. The application of polysaccharide in drug
delivery is growing very fast as it provides a safe, nontoxic and tunable alternates of synthetic
polymers for the same purpose. The diverse chemical structures, functional groups and
physicochemical properties are making polysaccharides a suitable candidate in drug delivery
[239, 240].
Polysaccharide based controlled release and targeted formulations
Unlike the synthetic hydrophilic polymers, polysaccharide possess a variety of hydrophilic
functional groups on its structure like –OH, -COOH, and –NH2 group which are responsible
for water absorption and further swelling of polysaccharide. These groups grant a variety of
functional capability to the polysaccharide-based systems such as bioadhesion and control
release property. Controlled release property of polysaccharide-based drug delivery systems
has been explored extensively for achieving a prolonged delivery of variety of therapeutics
such as chemotherapeutics, proteins, peptides, nucleic acid and many more [1]. The control
release of therapeutics from the swelled polysaccharide depends on the porosity or degree of
swelling, further regulated by certain parameters like pH, temperature, ionic strength and
electric fields [241]. Several polysaccharides present controlled release and target specific
delivery applications.
1. Chitosan: It is a linear polysaccharide obtained from the exoskeleton of arthropods such as
crabs, lobster, and shrimp and cell walls of fungi. It is available in variety of molecular weight
and degree of acetylation, which control its physical and biological property. Hydroxy and
amino groups on the chitosan are involved in making intra- and intermolecular hydrogen bonds.
The pH dependent protonation of amino group leads to swelling of the Chitosan to form three-
dimension structure gel at pH lower than the pKa [242]. Various chitosan salts such as chitosan
palmitate and chitosan laurate exhibit different degrees of swelling, and thus drug release from
the variety of dosage form. The presence of primary amines in chitosan is responsible for its
biological activity such as antimicrobial, anticancer. Chitosan nanoparticles showed greater
antimicrobial activity against Staphylococcus aureus, Klebsiella pneumoniae, Pseudomonas
aeruginosa, and Escherichia coli compared to parent chitosan and chitin [243]. Chitosan-based
controlled release and targeted formulations developed so far are compiled in Table 2.
2. Alginate: It is an extensively studied linear polysaccharide derived from brown seaweed
(Macrocystis pyrifera, Laminaria hyperborean) and from some certain bacteria belonging to
the family Pseudomonas and Azotobacter. Similar to chitosan, aginate also form a thick gel at
low pH, which served as a barrier for controlled release of therapeutics. Additionally, alginate
also form thick gel with certain polyvalent cations (Ca2+ and Na+) due to crosslinking of
carboxylic groups of polysaccharide structure. Due to the bioadhesion property of alginate, it
offers targeted drug delivery applications. So far, this polysaccharide has been explored for
variety of formulations including matrix tablet [244], microspheres [245], pellets [246] and
nanoparticles for targeted and functionalized delivery [247] (Table 2). The combination of S-
nitroso-mercaptosuccinate (a nitric oxide donor) and green tea synthesized silver nanoparticles
incorporated into alginate hydrogel showed synergistic antimicrobial activity against
Staphylococcus aureus, Escherichia coli and Streptococcus mutants compared to individual
compounds [248].
3. Pectin: Pectin is a complex polysaccharide present almost one third of the dry cell wall
material of most of the plants. These are higher molecular weight (50,000 and 150, 000 Da)
polysaccharides consist α-(1-4) linked esterified D-galacturonic moiety. Various
physicochemical parameters of pectin like solubility, viscosity and gelling properties depends
upon the degree of methyl esterification of D-galacturonic moiety. Depending upon the degree
of esterification two types of pectin are commercially available; highmethoxyl pectins (degree
of esterification > 50) and low methoxyl pectins (degree of esterification < 50) [249]. Various
plant pectins reportedly exhibit applications as controlled release excipients in tablet, hydrogels
and pellets. Additionally, plant pectins offer applications in colon targeted drug delivery due
to their biodegradation potential by colonic microflora [250]. The pectin polysaccharide from
Ulmus pumila L. (PPU) possess potent anti-inflammatory activity. It was observed that the
selenized-PPU inhibit lipopolysaccharide (LPS)-stimulated nitric oxide release by targeting the
protein expression of inducible nitric oxide synthase in murine macrophage cell line RAW
264.7 [251].
4. Guar Gum: It is a high molecular weight linear polysaccharide obtained from the seeds of
the plant Cyamopsis tetragonoloba. The polysaccharide consist of linear chain of (1 → 4)-
linked β-D-mannopyranosyl units with (1 → 6)-linked α-D-galactopyranosyl residues as side
chains with the ratio of mannose to galactose units as 2:1. This polysaccharide swells in polar
solvent and thus has been widely explored for various pharmaceutical applications. Addition
of ionic and non-ionic additives (sodium chloride and glycerin) could change the swelling and
erosion property of polysaccharide for offering a variety of applications [14]. This
polysaccharide serves as tablet binder sustain release polymer and viscosity enhancers. This
polysaccharide has become the primary choice as excipient in colon specific oral product as it
remain indigestible in upper GIT and completely degrades by the colonic microflora [252].
Gamal-Eldeen prepared guar gum C-glycosylated derivative (GG) and its sulphated derivative
(SGG) to investigate its anti-cancer and anti-inflammatory potency. It was observed that GG
inhibits cytochrome P450 1A (a carcinogen activator enzyme) and stimulated the carcinogen-
neutralizing enzymes such as glutathione-S-transferases. Furthermore, both GG and SGG were
able to exert anti-inflammatory by reducing the release of nitric oxide and tumour necrosis
factor-alpha in LPS induced RAW264.7. GG and SGG also inhibited the proliferation of human
hepatocellular carcinoma cells (Hep G2), while only SGG was particularly toxic for MCF-7
[253]. Table 2 presents the selected applications of guar gum as pharmaceutical excipients.
5. Starch: Starch is a naturally available polysaccharide obtained from plants. Its characteristic
properties such as biotolerance and non-immunogenicity for human use, and easy availability
make it a commonly used polymers/pharmaceutical excipient [254, 255]. In comparison to
other polymers such as gum and cellulose, starch does not need to undergo excessive
purification, as they are relatively pure. Structurally, starch has two different glucan chains a.
amylose (linear polymer) and b. amylopectin (branched polymer) representing 98-99% of the
dry weight. In the production of capsules and tablets, starch is widely utilized as a lubricant,
diluent, binder and disintegrant whereas it is also commonly employed for variety of
specialized drug delivery applications, for example delivery of specialized drug and targeting
specific parts of human body [256]. The microsphere of oregano essential oil prepared by
starch-based material utilizing supercritical fluid technology showed superior antioxidant
capacity (in terms of oxygen radical absorbing capacity) and stability. Starch-based drug carrier
systems present high efficacy in the oral delivery of insoluble drugs [257]. Luo et al. (2020)
encapsulated curcumin into starch microparticles and the formulation showed enhanced
stability against photodegradation and oxidative modification. Furthermore, the size of
curcumin starch microparticles was possible to control accurately from 0.3-2µm by altering the
rate of debranching reaction that caused alteration of release characteristic of curcumin. These
observations suggested the management of the release site by modifying the crystallinity or
size of microparticles [258]. Alp et al. 2019, formulated starch nanoparticles for the delivery
of epigenetic drugs CG-1521 for breast cancer that inhibits histone deacetylase. The
nanoparticles formulation showed decrease release rate of CG-1521 allowing remarkable
cytotoxicity against MCF-7 breast cancer cell as compared to free CG-1521 [259].
Table 2: Different polysaccharides explored for the sustained release of pharmaceuticals
Folic acid Methotrexate Enhanced uptake of delivery systems by colorectal cancer [266]
conjugated cells
nanoparticles
Nanoaggregates Isoniazid and Formulations shows enhanced local action due to sustained [267]
Rifampicin
release
Hydrogel Rifampicin Enhance mucoadhesion thus better efficacy [268]
Starch Microspheres Oregano essential oil High antioxidant activity values and stability [257]
Nanoparticles CG-1521 (histone Decrease release rate of CG-1521 allowing remarkable [259]
deacetylase inhibitor)
cytotoxicity against MCF-7 breast cancer
Polysaccharides as emulsion stabilizers
The amphiphilic nature of chemically modified polysaccharides causes their adsorption at the
interface of oil and aqueous solvents that results in the stabilization of the emulsion [269]. This
property of polysaccharides proved highly beneficial for the delivery of lipophilic or non-polar
therapeutics at their target site [270]. As such, the native cellulose does not offer emulsion
stabilization due to its non-solubility in water. The chemically modified cellulose not only
improves its aqueous solubility but also affords the stabilization of the emulsion [271]. The
large size of polysaccharides slows down their adsorption process compared to the small sized
surface-active agents. Mainly, the polysaccharide-based emulsifiers mainly base on pectins,
gum arabic, galactomannans, and cellulose. Cellulose and starch promote the emulsion
stabilization only after their chemical modification however; the polysaccharides such as gum
arabic and pectins endowed with covalently linked glycoproteins possess intrinsic ability for
the stabilization of the emulsion. In addition, the polysaccharides such as xanthan gum,
carrageenan, alginates, hyaluronan, chitosan, and alginates that display trivial interfacial
activity offer the eulsion stabilization via thickening, gelling, and structuring [272]. The
structural relationship between chitin-based nanomaterials to that of cellulose results in a
similar amphiphilic behavior for potential applications as stabilizers of oil-in-water Pickering
emulsion. Starch microgranules demonstrate similar applications however; the nanocrystals
and nanospheres of starch exhibit a limited stabilization of the emulsion [273]. Yan et al. 2019
reported the stabilization of Pickering emulsions droplets by nanocrystals of bacterial cellulose
for hydrophobic drug delivery of Alfacalcidol. The irreversible adsorption of bacterial cellulose
nanocrystals at the oil-water interface of Pickering emulsions prevented the coalescence of the
droplets. The emulsion showed Ostwald ripening in the alginate solution. The interfacial
assembly of the amphiphilic bacterial cellulose nanocrystals and the hydrogel shells of the
alginate beads generated by the external gelation led to the achievement of efficient loading
and a controlled release of Alfacalcidol. The release mechanism of Alfacalcidol from the
composite beads followed non-Fickian transport. The alginate composite beads demonstrated
minimal toxicity that further proved beneficial for drug delivery applications. Koshani et al.
2021 presented the lipophilic drug delivery applications via natural emulgel obtained from
dialdehyde cellulose crosslinked with chitosan. The delivery of the lipophilic compound β-
carotene occurred using an embedded oil-in-water emulsion. The lipophilic β-carotene loaded
in the oil-phase showed 20% release in stomach after passing safely through the oral cavity.
Furthermore, 50% of β-carotene released in the intestines after 4h in the presence of emulgel
thereby indicating its application as oral delivery vehicle [274].
Clinical trials on Polysaccharide based drug delivery systems
Riedl et al. (1993) performed phase I clinical trials on the dextran-conjugated doxorubicin on
13 patients at a starting dose of 40 mg/m2 that led to the development of WHO grade IV
thrombocytopenia in 2/2 patients. Whereas, the WHO grade IV hepatotoxicity, and WHO grade
III cardiotoxicity appeared in patients with preexisting heart ailments. Lowering of the dose to
20 mg/m2 further decreased the existence of thrombocytopenia. However, the hepatotoxicity
persisted. Further reducing the dose to 12.5 mg/m2 caused marked reduction in the malignancy
fibrous histiocytoma for up to 4 months [275]. Soepenberg et al. (2005) performed phase I
clinical trials on DE-310, a camptothecin analog, and a carboxymethyldextran polyalcohol
carrier, in the patients with advanced solid tumors. The DE-301 drug delivery system
reportedly released DX-8951 drug slowly, while maintaining a sustained release. This
prolonged the drug exposure at its deliberated tumor site. Some patients showed partial-to-
complete remission of metastatic adenocarcinoma at dose 7.5 mg/ m2 [276]. Kim et al. (2006)
conducted phase II clinical trials on holmium 166/ chitosan complex for the effective treatment
of hepatocellular carcinoma. The percutaneous administration of the nanosystem caused a
complete tumor necrosis in 31/40 patients after 2 months period. However, a long-term follow-
up period indicated recurring of tumor in 28 patients. The polysaccharide based drug delivery
system proved beneficial for the treatment of small hepatocellular carcinoma by performing
local ablative procedure [277]. Pinnix et al. (2012) performed single-blind randomized phase
III clinical trials on topical hyaluronic acid after adjuvant radiotherapy for breast cancer. The
topical hyaluronic based gel did not proved effective for the treatment of grade 2 dermatitis
following radiotherapy [278]. Pritchard et al. (2016) reported phase II clinical trials on
oligosaccharide polymer therapy for modification of mucus barrier of COPD. The binding of
the oligosaccharide caused alteration in the surface charge, porosity, and 3-D mucin networks
in sputum of patients with cystic fibrosis, and its inhalation in patients caused effective
deposition in lungs and altered the viscoelasticity of the cystic fibrosis sputum [279].
Conclusion and Future perspectives
The polysaccharide based drug-delivery vehicles traversed a long journey for the controlled
release of pharmaceuticals at the target site with minimized ensuing side effects caused by the
customary delivery vectors. The biodegradability and trivial immunogenicity of the
polysaccharide-based drug delivery vehicles makes them the material of the future. The
controlled release profile offers improved drug pharmacokinetics thereby leading to
ameliorated local action, and effectivity. As such, the polysaccharides present the first-in-class
drug delivery system ‘Novochizol’ for the delivery of COVID-19 drugs. The system comprises
of biodegradable and biocompatible chitosan nanoparticles that strongly adhere to the lung
epithelium and offers a sustained drug-release. Similarly, the recent discovery of helical V-
amylose as advanced drug delivery system further validated the precedence of polysaccharides
as drug delivery vehicles. The helical morphology of V-amylose offers drug encapsulation in
the helix groves while interacting with the functional head groups. This system proved highly
advantageous for the delivery of indomethacin, diclofenac, and aspirin with enhanced gastric
tolerance. The conjugation of polysaccharides with metal nanoparticles with unique
physicochemical profile in the form of optical, electronic, magnetic, and surface properties
provide nanoprobes that present a sturdy candidature in bioimaging of the effected site, while
delivery the cargo pharmaceutical at the same time. The development of multicompartmental
microspheres based on polysaccharides revolutionized bioengineering and multi-drug delivery
with high precision. The coaxial electrospun fiber membranes based on polysaccharides
transformed the plant grafting techniques and effectively managed the plant tissue injury due
to their high mechanical and tensile strength. The biological phenomenon such as wood
degradation by brown-rot fungi led to the development of bioinspired, core shell cellulose
microparticles for the light-triggered release of anticancer drugs. Furthermore, the stabilization
of Pickering emulsions by chemically modified polysaccharides play a significant role in the
lipophilic drug delivery applications, and targeted delivery of various pharmaceuticals and
bioactive ingredients.
Despite several benefits, the polysaccharide-based formulations suffer limitations such as
susceptibility towards microbial contamination, uncontrolled hydration rate, and reduced
viscosity during storage. To overcome these limitations, it mandates the modification of natural
polysaccharides via cross-linking, grafting, and blending with natural and synthetic polymers
to improve their physicochemical profile. Similarly, the limited knowledge about the
mechanism of drug release by polysaccharides, and permeation enhancement decelerates the
progressive developments in polysaccharide-based drug delivery systems.
References
[1] Z. Liu, Y. Jiao, Y. Wang, C. Zhou and Z.J. Zhang, Polysaccharides-based nanoparticles as
drug delivery systems, Adv. Drug Deliv. Rev. 60 (2008) 1650-1662.
[2] K. Janes, P. Calvo, M.J. Alonso, Polysaccharide colloidal particles as delivery systems for
macromolecules, Adv. Drug Deliv. Rev. 47 (2001) 83-97.
[3] A.F. Metaxa, E. Vrontaki, E.K. Efthimiadou, T. Mavromoustakos (2021) Drug Delivery
Systems Based on Modified Polysaccharides: Synthesis and Characterization. In: T.
Mavromoustakos, A.G. Tzakos, S. Durdagi (eds) Supramolecules in Drug Discovery and Drug
Delivery. Methods in Molecular Biology, vol 2207. Humana, New York, NY.
https://doi.org/10.1007/978-1-0716-0920-0_12.
[5] Y. Zheng, Q. Xie, H. Wang, Y. Hu, B. Ren, X. Li, Recent advances in plant polysaccharide-
mediated nano drug delivery systems, Int. J. Biol. Macromol. 165 (2020) 2668-2683.
[6] Y. Sun, X. Jing, X. Ma, Y. Feng, H. Hu, Versatile types of polysaccharide-based drug
delivery systems: From strategic design to cancer therapy, Int. J. Mol. Sci. (MDPI). 21 (2020)
Article 9159.
[7] A.K. Nayak, M.S. Hasnain, A.K. Dhara D. Pal (2021) Plant Polysaccharides in
Pharmaceutical Applications. In: D. Pal, A.K. Nayak (eds) Bioactive Natural Products for
Pharmaceutical Applications. Advanced Structured Materials, vol 140. Springer, Cham.
https://doi.org/10.1007/978-3-030-54027-2_3.
[8] K. Park, J.-H. Kim, Y.S. Nam, et al., Effect of polymer molecular weight on the tumor
targeting characteristics of self-assembled glycol chitosan nanoparticles, J. Control Release.
122 (2007) 305-314.
[12] S.H. Lee, R. Bajracharya, J.Y. Min, J-W Han, B.J. Park, H-K Han, Strategic approaches
for colon targeted drug delivery: An overview of recent advancements, Pharmaceutics (MDPI).
12 (2020) Article 68.
[13] M. Naeem, U.A. Awan, F. Subhan, J. Cao, S.P. Hlaing, J. Lee, E. Im, Y. Jung, J-W Yoo,
Advances in colon-targeted nano-drug delivery systems: challenges and solutions, Arch.
Pharmacal. Res. 43 (2020) 153-169.
[14] B. Layek, S. Mandal, Natural polysaccharides for controlled delivery of oral therapeutics:
a recent update, Carbohyd. Polym. 230 (2020) 115617.
[15] M. Thanou, J. Verhoef and H.J. Junginger, Oral drug absorption enhancement by chitosan
and its derivatives, Adv. Drug Deliv. Rev. 52 (2001) 117-126.
[16] N.G. Schipper, S. Olsson, J.A. Hoogstraate, A.G. deBoer, K.M. Vårum and P. Artursson,
Chitosans as absorption enhancers for poorly absorbable drugs 2: mechanism of absorption
enhancement, J. Pharm. Res. 14 (1997) 923-929.
[17] A. Almeida, M. Araujo, R.N. Carballal, F. Andrade, H. Goncalves, R.L. Reis, M. Lucio,
S. Schwartz, B. Sarmento, Novel amphiphillic chitosan micelles as carriers for hydrophobic
anticancer drugs, Mater. Sci. Eng. C. 112 (2020) Article 110920.
[19] S. Peers, A. Montembault, C. Ladaviere, Chitosan hydrogels for sustained drug delivery,
J. Control. Release. 326 (2020) 150-163.
[20] Z.A. Raza, S. Khalil, A. Ayub, I.M. Banat, Recent developments in chitosan encapsulation
of various active ingredients for multifunctional applications, Carbohyd. Res. 492 (2020)
Article 108004.
[21] V.M. Platt and F.C. Szoka Jr, Anticancer therapeutics: targeting macromolecules and
nanocarriers to hyaluronan or CD44, a hyaluronan receptor, Mol. Pharm. 5 (2008) 474-486.
[23] R. Liu, R. Zuo, G.A. Hudalla, Harnessing molecular recognition for localized drug
delivery, . Adv. Drug Deliv. Rev. 170 (2021) 238-260.
[24] A. Bianchera, R. Bettini, Polysaccharide nanoparticles for oral controlled drug delivery:
the role of drug-polymer and interpolymer interactions, Expert Opin. Drug Deliv. 17 (2020)
1345-1359.
[26] H.Q. Song, Y. Fan, Y. Hu, G. Cheng, F-J Xu, Polysaccharide-peptide conjugates: A
versatile material platform for biomedical applications, Adv. Funct. Mater. 31 (2021) Article
2005978.
[28] D. Wu, L. Zhu, Y. Li, X. Zhang, S. Xu, G. Yang, T. Delair, Chitosan-based colloidal
polyelectrolyte complexes for drug delivery: A review, Carbohyd. Polym. 238 (2020) Article
116126.
[30] V.M. Leitner, G.F. Walker, A. Bernkop-Schnürch, Thiolated polymers: evidence for the
formation of disulphide bonds with mucus glycoproteins, Eur. J. Pharmaceutic
Biopharmaceutic. 56 (2003), pp. 207-214.
[31] R.H. Marchessault, F. Ravenelle and X.X. Zhu, Polysaccharides for drug delivery and
pharmaceutical applications, ACS Symposium Series (2006).
[32] A.K. Sharma, P. Prasher, A.A. Aljabali, V. Mishra, H. Gandhi, S. Kumar, S. Mutalik, D.K.
Chellappan, M.M. Tambuwala, K. Dua, D.N. Kapoor, Emerging era of “somes”:
polymersomes as versatile drug delivery carrier for cancer diagnostics and therapy. Drug Deliv.
Transl. Res. 10 (2020) 1171-1190.
[33] J.-H. Kim, Y.-S. Kim, S. Kim, et al., Hydrophobically modified glycol chitosan
nanoparticles as carriers for paclitaxel, J. Control. Release. 111 (2006), pp. 228-234.
[35] X. Du, S. Yin, L. Xu, J. Ma, H. Yu, G. Wang, J. Li, Polylysine and cysteine functionalized
chitosan nanoparticle as an efficient platform for oral delivery of paclitaxel, Carbohyd. Polym.
229 (2020) 115484.
[36] N. Feng, H. Wu, Y. Xie, Q. Wu, A novel drug delivery system obtained from hydrophobic
modified amphiphilic polymers by Maillard reaction, Int. J. Biol. Macromol. 157 (2020) 146-
150.
[37] M. Zembala, Electrokinetics of heterogeneous interfaces, Adv. Colloid Interface Sci. 112
(2004) 59-92.
[38] A.A. Kamel, C.M. Ma, M.S. El-Aasser, F.J. Micale, S.A. Vanderhoff, Concerning the
origin of charge at the polystyrene particle/water interface, J. Disp. Sci. Technol. 2 (1981) 315-
330.
[40] S. Bhattacharjee, DLS and zeta potential–what they are and what they are not?, J. Control.
Release. 235 (2016) 337-351.
[41] R.N. Wijesena, N.D. Tissera, V.W.S.G. Rathnayaka, R.M. Silva, K.M.N. Silva, Colloidal
stability of chitin nanofibers in aqueous systems: Effect of pH, ionic strength, temperature and
concentration, Carbohyd. Polym. 235 (2020) Article 116024.
[42] F. Gambinossi, S.E. Mylon, J.K. Ferri, Aggregation kinetics and colloidal stability of
functionalized nanoparticles, Adv. Colloid Interface Sci. 222 (2015) 332-349.
[43] P. Vera, V. Gallardo, J. Salcedo, M.A. Ruiz and A.V. Delgado, Electrokinetics and
stability of a cellulose acetate phthalate latex, J. Appl. Polymer Sci. 65 (1997) 2721-2726.
[45] X-Y Zhang, Q-S Wang, Z-X Wu, D-P Tao, An experimental study on size distribution
and zeta potential of bulk cavitation nanobubbles, Int. J. Mineral Metallurgy mater. 27 (2020)
152-161.
[46] B-L Vinh, N.M.N. Le, I. Nazir, B. Matuszczak, A.B. Schnurch, Chitosan based micelle
with zeta potential changing property for effective mucosal drug delivery, Int. J. Biol.
Macromol. 133 (2019) 647-655.
[48] G. Han, S. Liu, Z. Pan, Y. Lin, S. Ding, L. Li, B. Luo, Y. Jiao, C. Zhou, Sulfonated chitosan
and phosphorylated chitosan coated polylactide membrane by polydopamine-assisting for the
growth and osteogenic differentiation of MC3T3-E1s, Carbohyd. Polym. 229 (2020) Article
115517.
[49] A.J. Bernkop-Schnürch, Strategies to overcome the polycation dilemma in drug delivery,
Adv. Drug Deliv. Rev. 136 (2018) 62-72.
[50] L. Wu, W. Shan, Z. Zhang, Y.J. Huang, Engineering nanomaterials to overcome the
mucosal barrier by modulating surface properties, Adv. Drug Deliv. Rev. 124 (2018) 150-163.
[51] A. George, P.A. Shah, P.S. Srivastava, Natural biodegradable polymers based nano-
formulations for drug delivery: A review, Int. J. Pharm. 561 (2019) 244-264.
[52] A. Gagliardi, E. Giuliano, E. Venkateshwararao, M. Fresta, S. Bulotta, V. Awasthi, D.
Cosco, Biodegradable polymeric nanoparticles for drug delivery to solid tumors, Front.
Pharmacol. 12 (2021) Article 601626.
[53] M.S. Hasnain, et al. (2020) Biopolymers for Drug Delivery. In: A. Nayak, M. Hasnain
(eds) Advanced Biopolymeric Systems for Drug Delivery. Advances in Material Research and
Technology. Springer, Cham. https://doi.org/10.1007/978-3-030-46923-8_1.
[55] T.G. Barclay, C.M. Day, N. Petrovsky, S. Garg, Review of polysaccharide particle-based
functional drug delivery, Carbohyd. Polym. 221 (2019) 94-112.
[57] Y. Yu, TT-Ergas, Y. Zhu, G. Fittolani, V. Bardoni, A. Singhal, R.J. Fair, A. Grafmuller,
P.H. Seeberger, M. Delbianco, Sysematic hydrogen-bond manipulations to establish
polysaccharide structure-property relationship, Angew Chem. 58 (2019) 13127-13132.
[59] G. Machell, G.N. Richards, 906. The alkaline degradation of polysaccharides. Part II. The
alkali-stable residue from the action of sodium hydroxide on cellulose, J. Chem. Soc. 1957,
4500-4506.
[61] Y. Ding, Y. Yan, Y. Peng, D. Chen, J. Mi, L. Lu, Q. Luo, X. Li, X. Zeng, Y. Cao, In
vitro digestion under simulated saliva, gastric and small intestinal conditions and fermentation
by human gut microbiota of polysaccharides from the fruits of Lycium barbarum. Int. J. Biol.
Macromol. 125 (2019) 751-760.
[62] T. Di, G. Chen, Y. Sun, S. Ou, X. Zeng, H. Ye, In vitro digestion by saliva, simulated
gastric and small intestinal juices and fermentation by human fecal microbiota of sulfated
polysaccharides from Gracilaria rubra, J. Funct. Food. 40 (2018) 18-27.
[63] X. Li, R. Guo, X. Wu, X. Liu, L. Ai, Y. Sheng, Z. Song, Y. Wu, Dynamic digestion of
tamarind seed polysaccharide: Indigestibility in gastrointestinal simulations and gut microbiota
changes in vitro, Carbohyd. Polym. 239 (2020) Article 116194.
[64] J.M. Krochta, J.S. Hudson, S.J. Tillin (1988) Kinetics of alkaline thermochemical
degradation of polysaccharides to organic acids, ACS Publications.
[65] N.G. Kotla, S. Rana, G. Sivaraman, O. Sunnapu, P.K. Vemula, A. Pandit, Y. Rochev,
Bioresponsive drug delivery systems in intestinal inflammation: State-of-the-art and future
perspectives, Adv. Drug Deliv. Rev. 146 (2019) 248-266.
[66] P. Prasher, R. Fatima, M. Sharma, Therapeutic delivery with V-amylose, Drug Dev. Res.
(2021) DOI: 10.1002/ddr.21804.
[67] B. Stone, B. Svensson, M. Collins, R. Rastall (2008) Polysaccharide Degradation. In: B.O.
Fraser-Reid, K. Tatsuta, J. Thiem (eds) Glycoscience. Springer, Berlin, Heidelberg.
https://doi.org/10.1007/978-3-540-30429-6_60.
[69] X. Lu, N. Li, X. Qiao, Z. Qiu, P. Liu, Effects of thermal treatment on polysaccharide
degradation during black garlic processing, LWT, 95 (2018) 223-229.
[71] J. Fu, J. Ji, W. Yuan, J.J.B. Shen, Construction of anti-adhesive and antibacterial
multilayer films via layer-by-layer assembly of heparin and chitosan, Biomater. 26 (2005)
6684-6692.
[72] M.G. Carneiro-da-Cunha, M.A. Cerqueira, B.W. Souza, S. Carvalho, M.A. Quintas, J.A.
Teixeira, A.A. Vicente, Physical and thermal properties of a chitosan/alginate nanolayered PET
film, Carbohyd. Polymer, 82 (2010) 153-159.
[75] F. Shi, X. Tian, D.J. McClements, Y. Chang, J. Shen, C. Xue, Influence of molecular
weight of an anionic marine polysaccharide (sulfated fucan) on the stability and digestibility
of multilayer emulsions: Establishment of structure-function relationships. Food Hydrocolloid.
113 (2021) Article 106418.
[76] S. Iqbal, Z. Xu, H. Huang, X.D. Chen, Structuring of water-in-oil emulsions using
controlled aggregation of polysaccharide in aqueous phases, J. Food Eng. 258 (2019) 34-44.
[77] M. Farid-Ul-Haq, M.A. Hussain, M.T. Haseeb, M.U. Ashraf, S.Z. Hussain, T. Tabassum,
I. Hussain, M. Sher, S.N.A. Bukhari, M.N. Hassan, A stimuli-responsive, superporous and non-
toxic smart hydrogel from seeds of mugwort (Artemisia vulgaris): stimuli responsive
swelling/deswelling, intelligent drug delivery and enhanced aceclofenac bioavailability. RSC
Adv. 10 (2020) 19832-19843.
[78] M.A. Hussain, A.I. Rana, M.T. Haseeb, G. Muhammad, L. Kiran, Citric acid cross-linked
glucuronoxylans: A pH-sensitive polysaccharide material for responsive swelling-
deswelling vs various biomimetic stimuli and zero-order drug release. J. Drug Deliv. Sci.
Technol. 55 (2020) Article 101470.
[81] H-G Fu, Y. Chen, Q. Yu, Y. Liu, Polysaccharide-based nanoparticles for two-step
responsive release of antitumor drug, ACS Med. Chem. Lett. 11 (2020) 1191-1195.
[84] B. Sasa, P. Odon, S. Stane, K. Julijana, Analysis of surface properties of cellulose ethers
and drug release from their matrix tablets, Eur. J. Pharm. Sci. 27 (2006) 375-383.
[85] N. Raghav, N. Mor, R.D. Gupta, R. Kaur, M.R. Sharma, P. Arya, Some
cetyltrimethylammonium bromide modified polysaccharide supports as sustained release
systems for curcumin. Int. J. Biol. Macromol. 154 (2020) 361-370.
[86] R. Zhang, M. Tang, A. Bowyer, R. Eisenthal, J.J.B. Hubble, A novel pH-and ionic-
strength-sensitive carboxy methyl dextran hydrogel, Biomater. 26 (2005) 4677-4683.
[88] Y. Guo, J. Sun, S. Bai, X. Wu, pH-Sensitive performance of dextran-poly (acrylic acid)
copolymer and its application in controlled in vitro release of ibuprofen, Int. J. Polym. Mater.
Polym. Biomater. 66 (2017) 900-906.
[90] N. Lin, A. Dufresne, Nanocellulose in biomedicine: Current status and future prospect,
Eur. Polym. J. 59 (2014) 302-325.
[91] H. Ni, S. Zeng, J. Wu, X. Cheng, T. Luo, W. Wang, W. Zeng, Y. Chen, Cellulose
nanowhiskers: preparation, characterization and cytotoxicity evaluation, Biomed. Mater. Eng.
22 (2012) 121-127.
[98] P.J. Eggenhuizen, B.H. Ng, J.D. Ooi, Treg enhancing therapies to treat autoimmune
diseases, Int. J. Mol. Sci. (MDPI) 21 (2020) Article 7015.
[99] E.B. Okeke, J.E. Uzonna, Th epivotal role of regulatory T cells in the regulation of innate
immune cells, Front. Immunol. 10 (2019) Article 680.
[100] N. Gupta, A.K. Jangid, D. Pooja, H. Kulhari, Inulin: A novel and stretchy polysaccharide
tool for biomedical and nutritional applications, Int. J. Biol. Macromol. 132 (2019) 852-863.
[101] K. Sampathkumar, S.C.J. Loo, Targeted gastrointestinal delivery of nutraceuticals with
polysaccharide-based coatings, Macromol. Biosci. 18 (2018) Article 1700363.
[102] E.V. Lopez, A.L. Alvarez, J.B. Mendez, F.J.O. Espinar, Cellulose-polysaccharide film-
coating of cyclodextrin based pellets for controlled drug release, J. Drug Deliv. Sci. Technol.
42 (2017) 273-283.
[103] S. Gao, G. Tang, D. Hua, R. Xiong, J. Han, S. Jiang, Q. Zhang, C. Huang, Stimuli-
responsive bio-based polymeric systems and their applications, J. Mater. Chem. B. 7 (2019)
709-729.
[104] T. Thambi, D.G. You, H.S. Han, V.G. Deepagan, S.M. Jeon, Y.D. Suh, K.Y. Choi, K.
Kim, I.C. Kwon, G-R Yi, J.Y. Lee, D.S. Lee, J.H. Park, Bioreducible carboxymethyl dextran
nanoparticles for tumor-targeted drug delivery, Adv. Healthcare Med. 11 (2014) 1829-1838.
[106] J.M. Shin, S-H Kim, T. Thambi, D.G. You, J. Jeon, J.O. Lee, B.Y. Chung, D-G Jo, J.H.
Park, A hyaluronic acid–methotrexate conjugate for targeted therapy of rheumatoid arthritis,
Chem. Commun. 50 (2014) 7632-7635.
[107] H. Jung, K.M. Park, J-A Yang, E.J. Oh, D-W Lee, K. Park, S.H. Ryu, S.K. Hahn, K.
Kim, Theranostic systems assembled in situ on demand by host-guest chemistry, Biomater. 32
(2011) 7687-7694.
[108] Q. Qu, J. Zhang, X. Chen, H. Ravanbakhsh, G. Tang, R. Xiong, B.B. Manshian, S.J.
Soenen, F. Sauvage, K. Braeckmans, S.C.D. Smedt, C. Huang, Triggered release from cellulose
microparticles inspired by wood degradation by fungi, ACS Sustain. Chem. Eng. 9 (2021) 387-
397.
[109] G. Tang, R. Xiong, D. Lv, R.X. Xu, K. Braeckmans, C. Huang, S.C.D. Smedt, Gas-
shearing fabrication of multicompartmental-microspheres: A one-step and oil-free approach,
Adv. Sci. 6 (2019) Article 1802342.
[110] Z. Guo, G. Tang, Y. Zhou, L. Shuwu, H. Hou, Z. Chen, J. Chen, C. Hu, F. Wang, S.C.D.
Smedt, R. Xiong, C. Huang, Fabrication of sustained-release CA-PU coaxial electrospun fiber
membranes for plant grafting application, Carbohyd. Polym. 169 (2017) 198-205.
[111] S. Zhao, X. Yu, Y. Qian, W. Chen, J. Shen, Multifunctional magnetic iron oxide
nanoparticles: an advanced platform for cancer theranostics, Theranostics. 10 (2020) 6278-
6309.
[112] H-W Cheng, H-Y Tsao, C-S Chiang, S-Y Chen, Advances in nanoparticle-mediated
cancer immune-theranostics, Adv. Healthcare Mater. 10 (2021) Article 2001451.
[113] R.G. Thomas, M. Muthiah, M.J. Moon, I-K Park, Y.Y. Jeong, SPION loaded poly(L-
lysine)/hyaluronic acid micelles as MR contrast agent and gene delivery vehicle for cancer
theranostics. Macromol. Res. 25 (2017) 446-451.
[114] M.H. El-Dakdouki, D.C. Zhu, K. El-Boubbou, M. Kamat, J. Chen, W. Li, X. Huang,
Development of Multifunctional Hyaluronan-Coated Nanoparticles for Imaging and Drug
Delivery to Cancer Cells, Biomacromol. 13 (2017) 1144-1151.
[115] R. Choi, J. Yang, J. Choi, E-K Lim, E. Kim, J-S Suh, Y-M Huh, S. Haam, Thiolated
Dextran-Coated Gold Nanorods for Photothermal Ablation of Inflammatory Macrophages,
Langmuir. 26 (2010) 17520-17527.
[116] C. Yue, P. Liu, M. Zheng, P. Zhao, Y. Wang, Y. Ma, L. Cai, IR-780 dye loaded tumor
targeting theranostic nanoparticles for NIR imaging and photothermal therapy, Biomater. 34
(2013) 6853-6861.
[120] M.A. Otache, R.U. Duru, O. Achugasim, O.J. Abayeh, Advances in the modification of
starch via esterification for enhanced properties, J. Polymer Environ. 29 (2021) 1365-1379.
[122] A.S. Volokhova, K.J. Edgar, J.B. Matson, Polysaccharide-containing block copolymers:
synthesis and applications, Mater. Chem. Front. 4 (2020) 99-112.
[124] A.J. Kerchove, M. Elimelech, Formation of Polysaccharide Gel Layers in the Presence
of Ca2+ and K+ Ions: Measurements and Mechanisms, Biomacromol. 8 (2007) 113-121.
[125] A. Chowhan, T.K. Giri, Polysaccharide as renewable responsive biopolymer for in situ
gel in the delivery of drug through ocular route, Int. J. Biol. Macromol. 150 (2020) 559-572.
[126] H. Zhong, X. Gao, C. Cheng, C. Liu, Q. Wang, X. Han, The structural characteristics of
seaweed polysaccharides and their applications in gel drug delivery systems, Marine Drug
MDPI). 18 (2020) Article 658.
[132] H.H. Tonnesen, J. Karlsen, Alginate in drug delivery systems, Drug Dev. Indus. Pharm.
28 (2002) 621-630.
[133] J. Li, C. Cai, J. Li, T. Sun, L. Wang, H. Wu, G. Yu, Chitosan-based nanomaterials for
drug delivery, Molecules (MDPI). 23 (2018) Article 2661.
[134] L. Szabo, S.G. Lemaire, C. Wandrey, Strategies to functionalize the anionic biopolymer
Na-alginate without restricting its polyelectrolyte properties, Polymers (MDPI). 12 (2020)
Article 919.
[135] E.M.P. Quito, R.R. Caro, M.D. Veiga, Carrageenan: Drug delivery systems and other
biomedical applications, Marine Drug (MDPI). 18 (2020) Article 583.
[136] J. Guan, L. Li, S. Mao, Applications of carrageenan in advanced drug delivery. Seaweed
polysaccharides. Elsevier. 2017.
[137] J.L. Jiang, W-Z Zhang, W-X Ni, J-W Shao, Insight on structure-property relationships
of carrageenan from marine red algal: A review, Carbohyd. Polym. 257 (2021) Article 117642.
[143] A. Aguilera-Garrido, T.C. Santaella, Y. Yang, F.G. Gonzalez, M.J.G. Ruiz, J.A.M.
Bolivar, J.A.H. Terriza, M.A.C. Vilchez, J.M. Valderrama, Aplications of serum albumins in
delivery systems: Differences in interfacial behavior and interacting abilities with
polysaccharides, Adv. Colloid. Interface Sci. 290 (2021) Article 102365.
[146] J.N. Putro, S. Ismadji, C. Gunarto, F.E. Soetaredjo, Y.H. Ju, Effect of natural and
synthetic surfactants on polysaccharide nanoparticles: Hydrophobic drug loading, release , and
cytotoxic studies, Colloid Surf A. 578 (2019) Article 123618.
[148] P.F. Cui, W-R Zhuang, X. Hu, L. Xing, R-Y Yu, J-B Qiao, Y-J He, F. Li, D. Ling, H-L
Jiang, A new strategy for hydrophobic drug delivery using a hydrophilic polymer equipped
with stacking units, Chem. Commun. 54 (2018) 8218-8221.
[149] C.A. Lorenzo, B.B. Fernandez, A.M. Puga, A. Concheiro, Crosslinked ionic
polysaccharides for stimuli-sensitive drug delivery, Adv. Drug. Dev. Rev. 65 (2013) 1148-
1171.
[150] Y. Wu, X. Zhang, H. Li, P. Deng, H. Li, T. He, J. Rong, J. Zhao, Z. Liu, A core/ shell
stabilized polysaccharide-based nanoparticle with intracellular environment-sensitive drug
delivery for breast cancer therapy, J. Mater. Chem. B. 6 (2018) 6646-6659.
[151] Y. Yu, T.T. Ergas, Y. Zhu, G. Fittolani, V. Bordoni, A. Singhal, R.J. Fair, A. Grafmuller,
P.H. Seeberger, M. Delbianco, Systematic Hydrogen‐ Bond Manipulations To Establish
Polysaccharide Structure–Property Correlations, Angew. Chem. 58 (2019) 13127-13132.
[152] F. Zhu, J. Rodriguez, T. Yang, I. Kevlishvili, E. Miller, D. Yi, S. Neill, M.J. Rourke, P.
Liu, M.A. Walczak, Glycosyl Cross-Coupling of Anomeric Nucleophiles: Scope, Mechanism,
and Applications in the Synthesis of Aryl C-Glycosides, J. Am. Chem. Soc. 139 (2017) 17908-
17922.
[153] P.O. Adero, H. Amarasekara, P. Wen, L. Bohe, D. Crich, The Experimental Evidence in
Support of Glycosylation Mechanisms at the SN1-SN2 Interface, Chem. Rev. 118 (2018) 8242-
8284.
[154] T. Hansen, L. Lebedel, et al., Defining the SN1 Side of Glycosylation Reactions:
Stereoselectivity of Glycopyranosyl Cations, ACS Cent. Sci. 5 (2019) 781-788.
[156] J.A.C. Romero, S.A. Tobbacco, K.A. Woerpel, Stereochemical Reversal of Nucleophilic
Substitution Reactions Depending upon Substituent: Reactions of Heteroatom-Substituted
Six-Membered-Ring Oxocarbenium Ions through Pseudoaxial Conformers, J. Am. Chem. Soc.
122 (2000) 168-169.
[157] E.C. Rodriguez, L.A. Marcaurelle, C.R. Bertozzi, Aminooxy-, Hydrazide-, and
Thiosemicarbazide-Functionalized Saccharides: Versatile Reagents for Glycoconjugate
Synthesis. J. Org. Chem. 63 (1998) 7134-7135.
[158] K.C.K. Swamy, N.N.B. Kumar, E. Balaraman, K.V.P.P. Kumar, Mitsunobu and related
reactions: Advances and applications, Chem. Rev. 109 (2009) 2551-2651.
[159] S. Fletcher, The Mitsunobu reaction in the 21st century, Org. Chem. Front. 2 (2015) 739-
752.
[164] T. Breton, G. Bashiardes, J-M Leger, K.B. Kokoh, Selective Oxidation of Unprotected
Carbohydrates to Aldehyde Analogues by Using TEMPO Salts, Eur. J. Org. Chem. 10 (2007)
1567-1570.
[166] P.S. Chang, J.F. Robyt, Oxidation of Primary Alcohol Groups of Naturally Occurring
Polysaccharides with 2,2,6,6-Tetramethyl-1-Piperidine Oxoammonium Ion, J. Carbohyd.
Chem. 15 (1996) 819-830.
[171] C. Chen, J-L Zhou, X. Han, F. Song, X-L Wang, Y-Z Wang, A prodrug strategy based
on chitosan for efficient intracellular anticancer drug delivery, Nanotechol. 25 (2014) 255101.
[172] C.K. Chen, P.K. Huang, W.C. Law, C.H. Chu, N.T. Chen, L.W. Lo, Biodegradable
Polymers for Gene-Delivery Applications, Int J Nanomedicine. 15 (2020) 2131-2150.
[173] N.N. Siddiqui, A. Aman, A. Silipo, S.A.U. Qader, A. Molinaro, Structural analysis and
characterization of dextran produced by wild and mutant strains of Leuconostoc mesenteroides,
Carbohyd. Polym. 99 (2014) 331-338.
[176] C. Longinotti, The use of hyaluronic acid based dressings to treat burns: A review, Burns
Trauma. 2 (2014) 162-168.
[182] O.P. Varghese, J. Liu, K. Sundaram, J. Hilborn, O.P. Oommen, Chondroitin sulfate
derived theranostic nanoparticles for targeted drug delivery, Biomater. Sci. 4 (2016) 1310-
1313.
[183] S. Arpicco, P. Milla, B. Stella, F. Dosio, Hyaluronic acid conjugates as vectors for the
active targeting of drugs, genes and nanocomposites in cancer treatment, Molecules (MDPI).
19 (2014) 3193-3230.
[184] C.I.C. Crucho, MT. Barros, Surfactant-free polymeric nanoparticles for biomedical
applications, Microsc. Microanal. 21 (2015) 74-75.
[185] X. Shen, J.L. Shamshina, P. Berton, G. Gurau, R.D. Rogers, Hydrogels based on
cellulose and chitin: fabrication, properties, and applications, Green Chem. 18 (2016) 53-75.
[186] R. Jayakumar, A. Nair, N.S. Rejinold, S. Maya, S.J.C.P. Nair, Doxorubicin-loaded pH-
responsive chitin nanogels for drug delivery to cancer cells, Carbohyd. Polym. 87 (2012) 2352-
2356.
[187] N.S. Rejinold, A. Nair, M. Sabitha, K. Chennazhi, H. Tamura, S.V. Nair, R. Jayakumar,
Synthesis, characterization and in vitro cytocompatibility studies of chitin nanogels for
biomedical applications, Carbohyd. Polym. 87 (2012) 943-949.
[192] W-L Song, K. Song, L-Z Fan, A Versatile Strategy toward Binary Three-Dimensional
Architectures Based on Engineering Graphene Aerogels with Porous Carbon Fabrics for
Supercapacitors, ACS Appl. Mater. Interface, 7 (2015) 4257-4264.
[193] J. Muller, A.C. Jakel, D. Schwarz, L. Aufinger, F.C. Simmel, Programming Diffusion
and Localization of DNA Signals in 3D-Printed DNA-Functionalized Hydrogels, Small. 16
(2020) Article 2001815.
[194] S.G. Kelmanovich, R. Parke-Houben, C.W. Frank, Competitive swelling forces and
interpolymer complexation in pH- and temperature-sensitive interpenetrating network
hydrogels, Soft Matter. 8 (2012) 8137-8148.
[195] D.G. Lim, E. Kang, S.H. Jeong, pH-dependent nanodiamonds enhance the mechanical
properties of 3D-printed hyaluronic acid nanocomposite hydrogels, J. Nanobiotechnol. 18
(2020) Article 88.
[197] L. Li, J. Ge, P.X. Ma, B. Guo, Injectable conducting interpenetrating polymer network
hydrogels from gelatin-graft-polyaniline and oxidized dextran with enhanced mechanical
properties, RSC Adv. 5 (2015) 92490-92498.
[198] T.T. Khong, O.A. Aarstad, G. Skjåk-Bræk, K.I. Draget, K.M. Vårum, Gelling Concept
Combining Chitosan and Alginate-Proof of Principle, Biomacromol. 14 (2013) 2765-2771.
[203] S. Kaity, A. Ghosh, Comparative bio-safety and in vivo evaluation of native or modified
locust bean gum-PVA IPN microspheres, Int. J. Biol. Macromol. 72 (2015) 883-893.
[204] V.K. Thakur, M.K. Thakur, Recent trends in hydrogels based on psyllium
polysaccharide: a review, J. Clean. Product. 82 (2014) 1-15.
[205] S.K. Nitta, K. Numata, Biopolymer-based nanoparticles for drug/gene delivery and tissue
engineering, Int. J. Mol. Sci. 14 (2013) 1629-1654.
[206] D.G. Fatouros, D.A. Lamprou, A.J. Urquhart, S.N. Yannopoulos, I.S. Vizirianakis, S.
Zhang, S. Koutsopoulos, Lipid-like self-assembling peptide nanovesicles for drug delivery,
ACS Appl. Mater. Interface. 6 (2014) 8184-8189.
[207] S.V. Lale, A. Kumar, S. Prasad, A.C. Bharti, V. Koul, Folic Acid and Trastuzumab
Functionalized Redox Responsive Polymersomes for Intracellular Doxorubicin Delivery in
Breast Cancer, Biomacromol. 16 (2015) 1736-1752.
[208] Y. Lu, K. Park, Polymeric micelles and alternative nanonized delivery vehicles for poorly
soluble drugs, Int. J. Pharm. 453 (2013) 198-214.
[210] M.R. Saboktakin, N.A. Tabar, R.M. Tabatabaie, A. Maharramov, M.A. Ramazanov,
Intelligent drug delivery systems based on modified chitosan nanoparticles, Lett. Org. Chem.
9 (2012) 56-70.
[211] D. Yadav, S. Suri, A. Chaudhary, M. Beg, V. Garg, M. Asif, A. Ahmad, Stimuli
responsive polymeric nanoparticles in regulated drug delivery for cancer, Polish J. Chem.
Technol. 14 (2012) 57-64.
[213] L. Li, K.M. Huh, Y-K Lee, S.Y. Kim, Biofunctional self-assembled nanoparticles of
folate-PEG-heparin/PBLA copolymers for targeted delivery of doxorubicin, J. Mater. Chem.
21 (2011) 15288-15297.
[216] Y. Tang, Y. Li, R. Xu, S. Li, H. Hu, C. Xiao, H. Wu, L. Zhu, J. Ming, Z.J.N. Chu, H.
Xu, X. Yang, Z. Li, Self-assembly of folic acid dextran conjugates for cancer chemotherapy,
Nanoscale. 10 (2018) 17265-17274.
[217] X. Zhang, R. Zhang, J. Huang, M. Luo, X. Chen, Y. Kang, J. Wu, Albumin enhances
PTX delivery ability of dextran NPs and therapeutic efficacy of PTX for colorectal cancer, J.
Mater. Chem. B. 7 (2019) 3537-3545.
[218] Q.S. Zhao, L.I. Hu, Z.D. Wang, Z.P. Li, W.A. Wen, J. Liu, Resveratrol-loaded folic acid-
grafted dextran stearate submicron particles exhibits enhanced antitumor efficacy in non-small
cell lung cancers, Mater. Sci. Eng. C. 72 (2017) 185-191.
[219] Y. Fang, H. Wang, H-J Dou, X. Fan, X-C Fei, L. Wang, S. Cheng, A. Janin, L. Wang,
W-L Zhao, Doxorubicin-loaded dextran-based nano-carriers for highly efficient inhibition of
lymphoma cell growth and synchronous reduction of cardiac toxicity, Int. J. Nanomed. 13
(2018) 5673-5683.
[220] T. Wang, J. Hou, C. Su, L. Zhao, Y. Shi, Hyaluronic acid-coated chitosan nanoparticles
induce ROS-mediated tumor cell apoptosis and enhance antitumor efficiency by targeted drug
delivery via CD44, J. Nanobiotechnol. 15 (2017) Article 7.
[221] Z. Yang, N. Sun, R. Cheng, C. Zhao, J. Liu, Z. Tian, Hybrid nanoparticles coated with
hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically
eliminate breast cancer stem cells, J. Mater Chem. B. 5 (2017) 6762-6775.
[222] P. Parashar, M. Rathor, M. Dwivedi, S.A. Saraf, Hyaluronic acid decorated naringenin
nanoparticles: appraisal of chemopreventive and curative potential for lung cancer,
Pharmaceutics. 10 (2018) Article 33.
[223] J. Li, X. Yang, P. Yang, F.J.I. Gao, Hyaluronic acid-conjugated silica nanoparticles for
breast cancer therapy, Inorg. Nanometal Chem. 47 (2017) 777-782.
[225] X-L Guo, X-X Kang, Y-Q Wang, X-J Zhang, C-J Li, Y. Liu, L-B Du, Co-delivery of
cisplatin and doxorubicin by covalently conjugating with polyamidoamine dendrimer for
enhanced synergistic cancer therapy, Acta Biomater. 84 (2019) 367-377.
[226] S.A. Shahin, R. Wang, S.I. Simargi, A. Contreras, L.P. Echavarria, L. Qu, W. Wen, T.
Dellinger, J. Unternaehrer, F.J.N.N. Tamanoi, Hyaluronic acid conjugated nanoparticle
delivery of siRNA against TWIST reduces tumor burden and enhances sensitivity to cisplatin
in ovarian cancer, Biol. Med. 14 (2018) 1381-1394.
[227] H. Jiang, X. Shi, X. Yu, X. He, Y. An, H. Lu, Hyaluronidase enzyme-responsive targeted
nanoparticles for effective delivery of 5-fluorouracil in colon cancer, Pharm. Res. 35 (2018)
73.
[228] M.A. Raja, M. Arif, C. Feng, S. Zeenat, C-G Liu, Synthesis and evaluation of pH-
sensitive, self-assembled chitosan-based nanoparticles as efficient doxorubicin carriers, J.
Biomater. Appl. 31 (2017) 1182-1195.
[229] J. Wu, C. Tang, C. Yin, Co-delivery of doxorubicin and interleukin-2 via chitosan based
nanoparticles for enhanced antitumor efficacy, Acta Biomater. 47 (2017) 81-90.
[230] B. Cheng, F. Gao, E. Maissy, P. Xu, Repurposing suramin for the treatment of breast
cancer lung metastasis with glycol chitosan-based nanoparticles, Acta Biomater. 84 (2019)
378-390.
[233] C. Zhang, G. Shi, J. Zhang, J. Niu, P. Huang, Z. Wang, Y. Wang, W. Wang, C. Li, D.
Kong, Redox-and light-responsive alginate nanoparticles as effective drug carriers for
combinational anticancer therapy, Nanoscale. 9 (2017) 3304-3314.
[235] A.A. Markeb, N.A. El-Maali, D.M. Sayed, A. Osama, M.A.Y. Abdel-Malek, A.H. Zaki,
M.E.A. Elwanis, J.J. Driscoll, Synthesis, Structural Characterization, and Preclinical Efficacy
of a Novel Paclitaxel-Loaded Alginate Nanoparticle for Breast Cancer Treatment, Int. J. Breast
Cancer. 2016 (2016) Article 7549372.
[236] N.P. Katuwavila, A.D.L.C. Perera, S.R. Samarakoon, P. Soysa, V. Karunaratne, G.A.J.
Amaratunga, D.N. Karunaratne, Chitosan-Alginate Nanoparticle System Efficiently Delivers
Doxorubicin to MCF-7 Cells, Journal of Nanomater. 2016 (2016) Article 3178904.
[237] L. Chen, M. Qian, L. Zhang, J. Xia, Y. Bao, J. Wang, L. Guo, Y. Li, Co-delivery of
doxorubicin and shRNA of Beclin1 by folate receptor targeted pullulan-based multifunctional
nanomicelles for combinational cancer therapy, RSC Advances. 8 (2018) 17710-17722.
[238] L. Huang, B. Chaurasiya, D. Wu, H. Wang, Y. Du, J. Tu, T. J. Webster, C. Sun, Versatile
redox-sensitive pullulan nanoparticles for enhanced liver targeting and efficient cancer therapy,
Nanomed. Nanotechnol. Biol. Med. 14 (2018) 1005-1017.
[242] Y. Cao, Y.F. Tan, Y.S. Wong, M.W.J. Liew, S. Venkatraman, Recent Advances in
Chitosan-Based Carriers for Gene Delivery, Marine Drug. 17 (2019) 381.
[247] M. Xie, Y. Xu, J. Liu, T. Zhang, H. Zhang, Preparation and Characterization of Folate
Targeting Magnetic Nanomedicine Loaded with Cisplatin, Journal of Nanomater. 2012 (2012)
1-9.
[248] A.L. Urzedo, M.C. Goncalves, M.H.M. Nascimento, C.B. Lombello, G. Nakazato, A.B.
Seabra, Cytotoxicity and Antibacterial Activity of Alginate Hydrogel Containing Nitric Oxide
Donor and Silver Nanoparticles for Topical Applications, ACS Biomater. Sci. Eng. 6 (2020)
2117-2134.
[250] T.W. Wong, G. Colombo, F. Sonvico, Pectin Matrix as Oral Drug Delivery Vehicle for
Colon Cancer Treatment, AAPS PharmSciTech. 12 (2011) 201-214.
[251] J-H Lee, Y-K Lee, Y-R Choi, J. Park, S.K. Jung, Y.H. Chang, The characterization,
selenylation and anti-inflammatory activity of pectic polysaccharides extracted from Ulmus
pumila L, Int. J. Biol. Macromol. 111 (2018) 311-318.
[252] R.C. Mundargi, S.A. Patil, S.A. Agnihotri, T.M. Aminabhavi, Development of
Polysaccharide-Based Colon Targeted Drug Delivery Systems for the Treatment of
Amoebiasis, Drug Dev. Indus. Pharm. 33 (2007) 255-264.
[253] A.M.G. Eldeen, H. Amer, W.A. Helmy, Cancer chemopreventive and anti-inflammatory
activities of chemically modified guar gum, Chem. Biol. Interact. 161 (2006) 229-240.
[256] R.M. Daudt, I.C.K. Guerreiro, F.C. Olivera, R.C.S. Thys, L.D.F. Marczak,
Determination of properties of pinhão starch: Analysis of its applicability as pharmaceutical
excipient, Industrial Crop Product. 52 (2014) 420-429.
[257] A.P. Almeida, S.R. Rojo, A.T. Serra, H-V Real, A-L Simplicio, I. Delgadilho, S.V.
Costa, L.B. Costa, Microencapsulation of oregano essential oil in starch-based materials using
supercritical fluid technology, Innovative Food Sci. Innovative Technol. 20 (2013) 140-145.
[258] K. Luo, H.J. Adra, Y-R Kim, Preparation of starch-based drug delivery system through
the self-assembly of short chain glucans and control of its release property, Carbohyd. Polym.
243 (2020) Article 116385.
[259] E. Alp, F. Dmkaci, E. Guven, M. Tenniswood, Starch nanoparticles for delivery of the
histone deacetylase inhibitor CG-1521 in breast cancer treatment. Int. J. Nanomed. 14 (2019)
1335-1346.
[260] Y. Shao, L. Li, X. Gu, L. Wang, S. Mao, Evaluation of chitosan-anionic polymers based
tablets for extended-release of highly water-soluble drugs, Asian J. Pharm. Sci. 10 (2015) 24-
30.
[262] R.A. Fitaihi, F.S. Aleanizy, S. Elsamaligy, H.A. Mahmoud, M.A. Bayomi, Role of
chitosan on controlling the characteristics and antifungal activity of bioadhesive fluconazole
vaginal tablets, Saudi Pharm. J. 26 (2018) 151–161.
[263] R.V. Pai, R.R. Jain, A.S. Bannalikar, M.D. Menon, Development and Evaluation of
Chitosan Microparticles Based Dry Powder Inhalation Formulations of Rifampicin and
Rifabutin. J. Aerosol Med. Pulmon. Drug Deliv. 29 (2016) 179-195.
[264] R.K. Dutta, S. Sahu, Development of diclofenac sodium loaded magnetic nanocarriers
of pectin interacted with chitosan for targeted and sustained drug delivery, Colloid. Surf. B. 97
(2012) 19–26.
[265] W. Zhou, L. Zou, C. Liu, R. Liang, J. Chen, Storage stability and skin permeation of
vitamin C liposomes improved by pectin coating, Colloid. Surf. B. 117 (2014) 330-337.
[266] M. Sharma, R. Malik, A. Verma, P. Dwivedi, G.S. Banoth, N. Pandey, J. Sarkar, P.R.
Mishra, A.K. Dwivedi, Folic Acid Conjugated Guar Gum Nanoparticles for Targeting
Methotrexate to Colon Cancer, J. Biomed. Nanotechnol. 9 (2013) 96-106.
[267] R. Kaur, T. Garg, B. Malik, U.D. Gupta, P. Gupta, G. Rath, A.K. Goyal, Development
and characterization of spray-dried porous nanoaggregates for pulmonary delivery of anti-
tubercular drugs, Drug Deliv. 23 (2016) 872–877.
[268] X. Yuan, R. Amarnath Praphakar, M.A. Munusamy, A.A. Alarfaj, S. Kumar, M. Rajan,
Mucoadhesive guargum hydrogel inter-connected chitosan-g-polycaprolactone micelles for
Rifampicin delivery, Carbohyd. Polym. 206 (2019) 1-10.
[270] K.S. Mikkonen, Strategies for structuring diverse emulsion systems by using wood
lignocellulose-derived stabilizers, Green Chem. 22 (2020) 1019-1037.
[272] B. Wang, H. Tian, D. Xiang, Stabilizing the oil-in-water emulsion using the mixtures of
Dendrobium Officinale polysaccharides and gum arabic or propylene glycol alginate,
Molecules (MDPI) 25 (2020) Article 759.
[274] R. Koshani, M. Tavakolian, T.G.M. Ven, Natural emulgel from dialdehyde cellulose for
lipophilic drug delivery, ACS Sustain. Chem. Eng. 9 (2021) 4487-4497.
[275] S.D. Riedl, E. Hausmann, H.D. Schick, R. Bender, et al., Phase I clinical and
pharmacokinetic trial of dextran conjugated doxorubicin (AD-70, DOX-OXD), Invest. New
Drug. 11 (1993) 187-195.
[277] J.K. Kim, K-H Han, J-T Lee, Y-H Paik, S.H. Ahn, J.D. Lee, K.S. Lee, C.Y. Chon, Y.M.
Moon, Long-term clinical outcome of phase IIb clinical trial of percutaneous injection with
holmium-166/chitosan complex (Milican) for the treatment of small hepatocellular carcinoma,
Clin. Cancer Res. 12 (2006) 543-548.
[278] C. Pinnix, G.H. Perkins, E.A. Strom, W. Tereffe, W. Woodward, J.L. Oh, L. Arriaga,
M.F. Munsll, P. Kelly, K.E. Hoffmann, B.D. Smith, T.A. Buchholz, T.K. Yu, Topical
hyaluronic acid vs. standard of care for the prevention of radiation dermatitis after adjuvant
radiotherapy for breast cancer: single-blind randomized phase III clinical trial, Int. J. Radiat.
Oncol. Biol. Phys. 83 (2012) 1089-1094.
[279] M.F. Pritchard, L.C. Powell, G.E. Menzies, P.D. Lewis, K. Hawkins, C. Wright, I. Doull,
T.R. Walsh, E. Onsoyen, E. Dessen, R. Myrvold, P.D. Rye, A.H. Myrset, H.N.E. Stevens, L.A.
Hodges, G.M. Gregor, J.B. Neilly, K.E. Hill, D.W. Thomas, A New Class of Safe
Oligosaccharide Polymer Therapy To Modify the Mucus Barrier of Chronic Respiratory
Disease, Mol. Pharm. 13 (2016) 863-872.