ArticlePDF AvailableLiterature Review

Hyaluronic Acid‐Based Bioconjugate Systems, Scaffolds, and Their Therapeutic Potential

Wiley
Advanced Healthcare Materials
Authors:
  • Novartis Institutes for BioMedical Research (NIBR)_Switzerland
  • Freyr Global Regulatory Solutions & Services

Abstract and Figures

In recent years, the development of hyaluronic acid or hyaluronan (HA) based scaffolds, medical devices, bioconjugate systems have expanded into a broad range of research and clinical applications. Research findings over the last two decades suggest that the abundance of HA in most mammalian tissues with distinctive biological roles and chemical simplicity for modifications have made it an attractive material with a rapidly growing global market. Besides its use as native forms, HA has received much interest on so‐called “HA‐bioconjugates” and “modified‐HA systems”. In this review, the importance of chemical modifications of HA, underlying rationale approaches, and various advancements of bioconjugate derivatives with their potential physicochemical, and pharmacological advantages are summarized. This review also highlights the current and emerging HA‐based conjugates of small molecules, macromolecules, crosslinked systems, and surface coating strategies with their biological implications, including their potentials and key challenges discussed in detail.
This content is subject to copyright. Terms and conditions apply.
REVIEW
www.advhealthmat.de
Hyaluronic Acid-Based Bioconjugate Systems, Scaffolds,
and Their Therapeutic Potential
Niranjan G. Kotla,* Isma Liza Mohd Isa, Aitor Larrañaga, Balaji Maddiboyina,
Samantha K. Swamy, Gandhi Sivaraman,* and Praveen K. Vemula*
In recent years, the development of hyaluronic acid or hyaluronan (HA) based
scaffolds, medical devices, bioconjugate systems have expanded into a broad
range of research and clinical applications. Research findings over the last two
decades suggest that the abundance of HA in most mammalian tissues with
distinctive biological roles and chemical simplicity for modifications have
made it an attractive material with a rapidly growing global market. Besides
its use as native forms, HA has received much interest on so-called
“HA-bioconjugates” and “modified-HA systems”. In this review, the
importance of chemical modifications of HA, underlying rationale approaches,
and various advancements of bioconjugate derivatives with their potential
physicochemical, and pharmacological advantages are summarized. This
review also highlights the current and emerging HA-based conjugates of
small molecules, macromolecules, crosslinked systems, and surface coating
strategies with their biological implications, including their potentials and key
challenges discussed in detail.
1. Introduction
Hyaluronic acid (HA) is a naturally occurring nonprotein, lin-
ear, nonsulfated glycosaminoglycan, a major component of the
extracellular matrix (ECM) of several biological tissues. Over
the last few decades, native HA and modified-HA systems have
witnessed an increased progression due to their promising
N. G. Kotla, P. K. Vemula
Institute for Stem Cell Science and Regenerative Medicine (inStem)
Bangalore, Karnataka 560065, India
E-mail: niranjan.kotla5@gmail.com; praveenv@instem.res.in
I. L. Mohd Isa
Department of Anatomy
Faculty of Medicine
Universiti Kebangsaan Malaysia
Kuala Lumpur 56000, Malaysia
A. Larrañaga
Department of Mining-Metallurgy Engineering and Materials Science
POLYMAT
Faculty of Engineering
University of the Basque Country (UPV/EHU)
Bilbao 48013, Spain
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adhm.202203104
DOI: 10.1002/adhm.202203104
multifaceted biophysical, biochemi-
cal, viscoelastic, and tissue remodeling
properties.[– ] A fairly accurate literature
search (in between  and Present)
on the PubMed website using the key-
words “hyaluronan,” “hyaluronic acid
conjugates,” gave total   and 
hits, respectively (as shown in (Figure
1a,b). Similarly, there are more than 
clinical trial reports available (Figure c)
(https://clinicaltrials.gov/, accessed on
February, ). Noticeably, this search
can be considered as a rough and not a
bibliography of a specific research topic.
Nevertheless, the number of published
research reports suggests that after an
initial pioneering period, HA has intrigued
biomedical researchers ever since it was
discovered because of the breadth of its
unique biophysical, mechanical and bio-
logical functionalities. According to the
current projections, the worldwide HA
market will be worth approximately USD . billion by ,
as HA-based products are rapidly evolving, especially in der-
mal, cosmetic, ocular, mucosal, bone, and other biomedical
applications.[]
Recently, the U.S. Food and Drug Administration (FDA) has
proposed to reclassify HA-based compounds, devices, products,
B. Maddiboyina
Department of Medical Writing
Freyr Solu ti on s
Hyderabad, Telangana 500081, India
S. K. Swamy
Thrombosis Research Center (TREC)
Department of Clinical Medicine
UiT-The Arctic University of Norway
Tromsø 9037, Norway
G. Sivaraman
Department of Chemistry
Gandhigram Rural Institute (Deemed to be University)
Gandhigram, Tamil Nadu 624302, India
E-mail: raman474@gmail.com
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (1 of 26)
www.advancedsciencenews.com www.advhealthmat.de
1398 2451
5297
10390
21022
9342
Number of publications
Hyaluro
anan
310 51 116
919
465
Number of publications
Hyaluronic acid conjugates
Early Phase I (12)
Phase I (37)
Phase II (65)
Phase III (66)
Phase IV (90)
Not applicable (338)
Total=608
b
c
Figure 1. a,b). Graphical data showing the number of articles published from 1970 to present on “hyaluronan” and “hyaluronic acid conjugates”. Source:
PubMed, last accessed February 2023. c) Number reported of clinical trials on hyaluronan (where, Early Phase I: exploratory trials before Phase I with
very limited human exposure; Phase IV: postmarketing surveillance; not applicable: trials without FDA-defined phases, such as devices or behavioral
intervention trials. Source: https://clinicaltrials.gov/, accessed on February 2023).
and delivery systems as therapeutic drugs by citing scientific tes-
timony, especially as HA provides pain relief eects (for, e.g., in
joint inflammation) through its biochemical activities in the liv-
ing systems;[, ] however, recent clinical studies data did not show
eective therapeutic benefit of HA injections in osteoarthritis
therapy as that of placebo.[] Henceforth, future HA-based prod-
ucts will have more regulatory scrutiny while entering into clini-
cal practice.[, ]
The recent literature reports have shown that HA molecular
mass, modified-HA systems influence various cellular, molec-
ular signaling mechanisms, matrix remodeling, and tissue
homeostasis.[] Besides, thanks to the unique potential thera-
peutic properties of HA with dierent biological role(s) in ECM
organization,[, ] cell proliferation,[, ] dierentiation,[– ]
wound healing processes,[, ] immunomodulation,[]
inflammation,[, ] modulating epithelial barrier,[]
viscosupplementation,[, ] and various cellular signaling pro-
cesses associated with tissue homeostasis, matrix remodeling,
and others.[,,, ]
The present review primarily describes the functional, bio-
chemical properties, and limitations/challenges of native HA (bi-
ologically present HA). After that, from taking the existing liter-
ature into account, we put forward the criterion for developing
modified-HA-based scaolds, HA-bioconjugate derivatives, and
their potential benefits for various biomedical applications. Be-
sides, the purpose of the review is to give a clear picture of ad-
vanced findings of modified-HA scaolds, with recently reported
applications of HA-based conjugates including small and macro-
molecule conjugates, crosslinked systems, cell-based HA scaf-
folds, HA-functionalized inorganic systems, and HA biocoating
approaches are also discussed in detail.
2. Structural, Functional Aspects of HA and the
Rationale for Bioconjugation of Therapeutics
The native form of linear, long-chain HA biopolymer has repeat-
ing disaccharide units of 𝛽-,--glucuronic acid and 𝛽-,-N-
acetyl--glucosamine bound with 𝛽-glycosidic linkages (Figure
2a). HA exists in ionized or salt forms at physiological pH, such
as sodium hyaluronate. Due to the presence of carboxylic groups
on each monomer, the molecule has an anionic property, making
it very hydrophilic; besides, the abundance of negatively charged
hydroxyl groups of HA can bind and hold a high number of wa-
ter molecules.[] In biological systems, the aforementioned re-
peating disaccharides can be >  units to form high molec-
ular weight (more than MDa) polymeric networks.[, ] Litera-
ture evidences suggest that HA biopolymer single disaccharides’
average length is nm (also reported as Kuhn length); as an,
e.g., HA polymer with   repetitions may be stretched out to
 μm.[] These HA chains in the cellular matrix build a dense,
brush like structure called pericellular matrix (PCM), in which
various proteoglycans attach, and modify the structure of the HA
chains. The composition and size of the PCM layer dier from
tissue to tissue based on their biological functionality.[]
In particular, these network structures further form noncova-
lent dense assemblies with HA binding proteoglycans (aggrecan,
chondroitin sulfate, versican, etc.) based on the type of tissue
microenvironment and ECM composition.[, ] HA synthesis
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (2 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 2. a) HA disaccharide unit chemical structure and its structural, physical, and biological properties. b) The regulation of HA synthesis by the HA
synthase (HAS 1, 2, and 3 enzymes) and degradation mainly by hyaluronidase enzyme and nonenzymatic (ROS reactive oxygen species, hydrolysis,
and thermal degradation) approaches. c) HA biopolymeric networks forming noncovalent assemblies with HA binding proteoglycans in the tissue
microenvironment(s), the possible associated surface receptor interactions, and downstream signaling mechanisms.
(regulated by the HA synthase (HAS) , , and enzymes) and
degradation (enzymatic, nonenzymatic) solely depend on the tis-
sue microenvironment; the physiological concentrations of HA
in tissues are essential to perform its regulatory functions. Each
enzyme has specific functionality in synthesizing appropriate
size range of HA polymer chains. HAS mainly produces the
small chains (.–. MDa), and HAS, HAS produce longer
chains (. to MDa or more), achieving polymer chains up to
 μminlength
[] (Figure b,c).
The hydrodynamic radius or size of the HA polymer depends
on the molecular weight; as the HA molecular weight increases
( to  kDa) the hydrodynamic radius increases from to
 nm.[] HA’s biophysical features (such as molecular weight,
chain length, crosslinking nature, viscoelasticity, adhesivity, cell-
tissue specific mechanics, hydrodynamic and degradation prop-
erties) can have a significant impact on cell to tissue mechan-
ics, ECM organization, tissue homeostasis, and alter healthy ver-
sus disease condition. As a result, along with other proteogly-
cans, HA is reported to be an element for a number of healthy
and disease-related processes.[] The biophysical and mechani-
cal characteristics of HA can be tunable and dependant on dier-
ent modified-HA forms including bioconjugate systems.
The growing evidence reveals that HA plays a key role in sev-
eral biophysical and biomechanical events in the ECM by binding
with matrix components and cell receptor interactions (specific
and nonspecific), especially with CD, RHAMM/CD,
LYVE-, TLRs, HARE, layilin, and others.[] CD involved in
HA-internalization and alter the ECM components degradation,
angiogenesis, cell adhesion, proliferation, and migration; notably
the immune cells activation, tracking, apoptosis and, impor-
tantly immunotolerance.[] Upon interaction of HA, these recep-
tors induce several intracellular signaling processes including
cell growth, tissue homeostasis, mechanotransduction, tumor
metastasis, inflammation, immunomodulation, and others (see
Figure ). Regardless of the successful application of unmodified-
HA, HA-based systems, its short half-life, poor mechanical
stability, uncontrolled mechanical, swift degradation behavior,
and poor adhesivity properties should be overcome for long-term
biomedical and clinical applications. Biopolymers, including HA
with biochemical stimuli mechanisms, provide essential clues
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (3 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Cross linking
Figure 3. Schematic illustration of various chemical modifications of HA including esterification, amidation, and crosslinking on carboxylic group of
each monomer; several OH functionalization on hydroxyl groups (here hyaluronic acid shown as HA or AH).
toward developing more eective modified-HA-based conjugates
or scaolds, which require consideration of various synthesis
approaches to develop standard modified-HA-based systems
with enhanced pharmacological, and therapeutic benefits.
3. Modifications of HA and Their Derivatives
HA biopolymer is susceptible to a variety of chemical modifi-
cations through three orthogonal functional moieties (hydroxyl,
carboxyl, and amide), facilitating its use for numerous applica-
tions requiring bioconjugation, chemical crosslinking, and sur-
face modifications of the delivery system.[] Chemical modifi-
cation of HA can be accomplished in two ways via crosslink-
ing or conjugation methods. The chemical modifications of HA
have been primarily carried out at dierent sites, including car-
boxyl groups of the 𝛽--glucuronic acid (GlcUA), hydroxyl groups
of the N-acetyl-𝛽--glucosamine (GlcNAc) residue, and the re-
ducing end of the disaccharide repeating unit. It is also possi-
ble to generate primary amino groups on the HA polymer chain
by deacetylation of GlcNAc for chemical conjugation of the acid
derivatives via amide formation. The chemical reactions of HA
are carried out either in aqueous or organic solvents such as
dimethylsulfoxide or dimethylformamide.[]
More importantly, the carboxylic acid group of HA is
frequently reacted with primary amine derivatives using
carbodiimide-mediated reactions under aqueous conditions.[]
Due to its water solubility, -ethyl--[-(dimethylamino)-propyl]-
carbodiimide (EDC) is the most often used carbodiimide.
Danishefsky and Siskovic were the first to use EDC to activate
the carboxyl groups of polysaccharides (pH .), including
HA, and then interacted with an amino acid ester to form
amides.[] Several publications have described Ugi condensa-
tion for HA crosslinking, which involves employing diamine as
a crosslinker to produce diamide connections between polysac-
charide chains.[, ] The synthesis of methyl ester of HA using
trimethylsilyl diazomethane (TMSD) as the carboxylic group
activator was reported, in which TMSD reacts with HA to form
an intermediate followed by acetic acid reaction to recover
the methyl ester.[] Several publications have documented the
interaction between HA and glycidyl methacrylate to produce
methacrylated HA.[, ] Besides, the hydroxyl groups of HA have
been used for esterification (using anhydrides or acid chlorides)
or ether formation (using epoxides, ethylene sulfides, or divinyl
sulfones).[] Figure 3 illustrates the various possible chemical
modifications of HA.
A large number of HA derivatives have been documented in
the literature, each of which is intended for a specific use, in par-
ticular various delivery systems (mentioned in Figure 4 and in
the next sections). More importantly, HA is a natural ligand of
several cell surface receptors as mentioned in the previous sec-
tion including CD, which overexpressed in several types of can-
cer cells. For example, conjugation of anticancer drugs to HA
significantly improves the solubility, stability, and targetability
to cancer tissues (briefly discussed in further sections). Besides,
HA is a unique biopolymer (with unique properties of water-
solubility, biocompatibility, biodegradability, the ability to target
to cells, and presence of multiple functional groups) that com-
bines many favorable attributes as a carrier for the conjugation-
based therapeutics delivery and imaging agents for biomedical
uses. In the next sections, we have provided the recent advance-
ments of various HA-based small, macromolecule conjugates,
crosslinked, and coating systems and their potential benefits for
various biomedical applications.
4. HA-Based Small Molecule Conjugates
Since the beginning of modern-day medicine, small-molecule
drugs have dominated the pharmaceutical industry with several
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (4 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 4. Chemical structures of various reported small molecule drug–HA conjugates. a–j) HA biopolymer conjugation to model anti-inflammatory, an-
ticancer, local-anesthetic, and steroid small molecule drugs. HA-small molecule drug conjugates have been employed to improve targetability, solubility,
cellular absorption, longer half-life, increase the delivery system stability, and extended release with combinatorial biological functions.
blockbuster drug candidates in the recent years until today.[]
Some of the common challenges in developing broad range of
small molecule drug based therapeutics are lack of bioavailabil-
ity, targetability, low cellular uptake, rapid degradation/clearance,
etc. Thus, prodrugs, drug-polymer scaolds, and modified-drug
conjugates including HA-conjugate systems (along with its
added therapeutic benefits) have been used to increase the tar-
getability of the drugs, with enhanced cellular uptake, prolonged
half-life, increased stability of the delivery systems, extended re-
lease with multiple functionalities. More recent studies attempt
to utilize the aforementioned properties of HA to develop HA-
based drug conjugates and to alter the biotherapeutic activity of
HA itself based on its range of available molecular weights.[, ]
In the below (Figure ), we have shown the reported small
molecule drug–HA conjugates investigated for various therapeu-
tic applications in particular anticancer therapies.
HA-conjugated paclitaxel has shown selective toxicity toward
various human cancer cell lines (breast, colon, and ovarian) that
are known to overexpress HA receptors. Conjugating hydropho-
bic anticancer drugs such as paclitaxel and -ethyl--hydroxy-
camptothecin (SN-) to HA significantly helps to overcome the
limited aqueous solubility and stability of these chemotherapeu-
tic agents.[] By taking the advantage of receptors overexpressed
on cancer cells including CD and RHAMM, various hydropho-
bic anticancer agents (paclitaxel, docetaxel, etc.) conjugation to
HA have significantly ameliorated the solubility, stability, and tar-
geted drug delivery. In a recent study, a stimuli-responsive den-
dronized HA–docetaxel conjugate (HA–DTX–Dendron, HADD)
was synthesized to target delivery of anticancer drug (DTX)
with high biocompatibility into tumor tissues via CD recep-
tors mechanism.[] Similarly, Pan and co-workers reported HA–
doxorubicin nanoparticulate system (with a mean hydrodynamic
size of  nm), interestingly, upon intravenous administration it
showed higher levels of accumulation in the intestines for at least
 h, which has the potential to enhance the selectivity and tar-
getability of the anticancer drugs for colorectal cancer therapy.[]
Anticancer drugs including camptothecin (CPT) have low wa-
ter solubility, and low cellular uptake; hence the use of water-
soluble prodrugs have been investigated to eectively overcome
the low solubility issue of CPT. Xu et al. determined the ef-
fect of HA molecular weight (MW) on the physicochemical
properties, antitumor activity of CPT–HA conjugates in which
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (5 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
HA acted as an eective solubilization carrier and targeting
molecule.[] In recent years, anticancer drug delivery technolo-
gies have used polymer chemistry and nanotechnology to de-
velop eective dosage forms that address constraints such as
increased targetability, drug cargo internalization, and local on-
site delivery with reduced o-targeted side eects. Zhang et al.
reported a novel multifunctional mesoporous silica nanoparti-
cles (MSN)-based biotin/HAase dual-stimulus-triggered delivery
system for targeted therapeutic delivery of doxorubicin (DOX);
further this study hypothesized that once MSN-HA/Dox is pref-
erentially taken up by cancer cells via receptor-mediated endo-
cytosis, drug release is triggered by HAase-mediated degrada-
tion of HA, which is enhanced further by desthiobiotin displace-
ment by intracellular biotin.[] In a similar study, Sauraj et al.
reported xylan--fluorouracil--acetic acid (Xyl--FUAC) prodrug
conjugate for colon cancer therapy, in which free drug showed
more cytotoxic eects compared to conjugate form in in vitro
studies.[] Taken together, these findings indicate that, modified-
HA-drug conjugate systems are eective in tumor-targeting with
increased therapeutic ecacy and stability.
Recently, lot of eorts were kept in developing polymers that
carry combination of dierent therapeutics with improved out-
comes. Krishnan et al. developed clinically translatable polymer–
drug conjugates of doxorubicin and camptothecin called Dox-
orubicin and Camptothecin Tailored at Optimal Ratios (DOC-
TOR) for topical treatment of skin malignancies using HA, where
DOCTOR outperformed the clinical standard (Efudex) in terms
of cancer-cell killing selectivity while being safe for healthy skin
cells.[]
In another recent study, HA-based hydrogel conjugates were
studied for their ability to change the rheological characteris-
tics using supramolecular chemistry as a physical interaction
with cyclodextrin (CD) constructs. The supramolecular system
(between CD and PEG) and catechol polymerization (between
dopamine molecules grafted to the HA polymer) could be used
as dual physical and chemical crosslinking strategies for tunable
hydrogels for long acting donepezil delivery via subcutaneous
administration[] (Figure 5).
In addition to the anticancer drug delivery applications, in-
teractions of HA and CD have been used in investigations
of atherosclerotic plaques.[, ] In a recent study, the hydropho-
bic atorvastatin (ATV) was constructed as the core of the HA
nanoparticle (mean size  nm) system (HA-ATV-NP) capable
of delivering a substantial amount of ATV per particle. When
compared to free drug, HA-ATV NPs can target inflammatory
atherosclerotic plaques via the CD receptor and release their
payload with increased therapeutic ecacy.[] Apparently, the
well-known antimalarial medicine hydroxychloroquine (HCQ)–
HA conjugate system was recently demonstrated to improve
bioavailability and treatment eectiveness for COVID- illness
through its versatile drug conjugation technology. The HA–HCQ
conjugate and HCQ were evaluated independently against four
distinct SARS-CoV- target proteins using molecular docking
experiments.[] Despite encouraging results from several in vitro
and in vivo experiments, chemically modified or HA-conjugated
formulations required more pharmacokinetics and dynamics
understanding before being used in therapeutic settings. Even
though eorts are made in this direction, further studies need
to be established on the degree of substitution of drug moieties
on HA chain/system and its eect on stability, release, pharma-
cokinetics, etc. In Table 1 various recently reported HA-small
molecule drug conjugates are shown.
5. HA-Based Macromolecule Conjugates
Macromolecules (such as protein and peptide drugs) possess
limitations that hinder their clinical potential, due to low wa-
ter solubility, low specificity, can cause associated o-targeted
side-eects. These agents have challenges in targeting to deliv-
ery sites, and required larger doses due to their short half-lives.
Recently, several drug delivery systems have been investigated as
potential solutions for overcoming these aforementioned short-
comings. In particular, HA has been investigated for its use as a
promising macromolecule drug carrier due to many advantages
including high water-solubility, biocompatibility, and targeting
ability with selective interaction with receptors.[] Additionally,
HA displays a very high loading capacity due to the presence of a
large number of hydroxyl and carboxyl groups on HA as well as its
reducing end that serves as a masked aldehyde to conjugate ther-
apeutics to terminal chain.[] In order to overcome some of the
disadvantages associated with utilizing HA as a drug carrier, vari-
ous chemical modifications (mentioned in the previous sections)
have been developed, including esterification of HA, chemically
modifying HA with carbodiimide, and crosslinking of HA with
either divinyl sulfone, glycidyl ether, or dialdehyde.[] In this sec-
tion, we will discuss and highlight recent advancements in the
HA-based systems for peptide and protein delivery for more e-
cient therapeutic applications (Table 2).
Recently, several works have investigated conjugation of HA
to serum albumin proteins in order to promote improved anti-
cancer drug delivery with enhanced therapeutic ecacy. Albu-
min (an endogenous protein) has been regarded as a promis-
ing drug carrier due to its nontoxicity, biodegradability, and
biocompatibility.[] A study by Edelman. et al. utilized the
HA–CD complex to create HA–bovine serum albumin (BSA)
nanoparticle conjugates loaded with paclitaxel. Researchers ob-
served that Maillard reaction-based covalent conjugates of BSA–
HA were able to self-assemble into nanoparticles (tonm)in
which hydrophobic molecules such as paclitaxel could be loaded
eciently. The paclitaxel-loaded BSA–HA nanoparticles were
selectively internalized, displayed high toxicity against CD-
expressing cancer cells against cells lacking CD expression
with selective targeting mechanism.[] This work suggests that
BSA–HA conjugates may be used as eective nanodelivery plat-
forms to enhance the delivery of hydrophobic chemotherapeutic
drugs.
In addition to research being done on anticancer drugs conju-
gated with HA to utilize HA’s binding anity for CD+cells,
HA derivatives are being investigated as potential facilitators for
prolonging the therapeutic activity of certain anti-inflammatory
medications. Over the last two decades, there have been multi-
ple studies in which researchers have investigated the improved
therapeutic eects of synthesizing conjugates of crosslinked-HA
with anti-inflammatory medications such as dexamethasone to
treat postoperative peritoneal adhesions,[] oligoethyleneimine
to treat ulcerative colitis[] (Figure 6), and anti-TNF-𝛼to help
heal severe burns.[] HA has drawn particular attention in the
field of arthritic diseases because, in addition to improving clear-
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (6 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 5. a,b) Schematic synthesis illustration of polypseudorotaxane and polydopamine-based hydrogel structures, as well as the design and fabrication
of a PDM-loaded HD-PEG/-CD hydrogel construct for subcutaneous, long-acting injectable delivery of donepezil. Reproduced with permission.[46]
Copyright 2021, Elsevier.
ance time and stability of antiarthritic drugs, HA is also a major
component of the cartilage extracellular matrix and can poten-
tially provide increased therapeutic ecacy when conjugated to
anti-inflammatory agents.[]
A relatively new field of research, progressively more stud-
ies are being conducted on the eects of anti-neurodegenerative
drugs conjugated with HA. HA has been considered an ideal
candidate as both a central nervous system tissue engineering
scaold and drug delivery device with regards to biocompatibil-
ity due to the existence of HA in the extracellular matrix of the
brain.[] HA has also been suggested for use alongside mes-
enchymal stem cell therapy as a tool to combat neurodegenerative
disease. Mesenchymal stem cells are currently being researched
as a potential method of regenerating neural networks in inflam-
matory sites where neural injuries or degeneration occur and be-
cause they express CD, it is proposed that HA’s anity for
CD might lead to some novel targeted anti-neurodegenerative
therapies.[]
Similarly, engineered hydrogels based on HA and fibronectin
were examined for their mechanobiological characteristics on hu-
man mesenchymal stem cells (MSCs), which mimic the quali-
ties of ECM to govern stem cells in both D and D settings.[]
Moreover, studies on HA-based antibody conjugates confirmed
the dual targeting ability of therapeutic agents in liver metas-
tasis treatment. Lee et al. reported a novel hyaluronate (HA)–
death receptor antibody (DR Ab) conjugate system by chem-
ical conjugation between the hydrazide group of PDPH ((-(-
pyridyldithio)propionyl hydrazide) and aldehyde group of DR
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (7 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 1 . Examples of recent reports on HA-small molecule drug conjugates, chemistry, and therapeutic applications.
Bioconjugation type Therapeutic agent Description of the conjugate and
linkage
Therapeutic application and outcome Refs.
Conjugation of doxorubicin
(DOX) and camptothecin
CPT to HA
Doxorubicin and
Camptothecin
Dual drug(s) covalent conjugated HA
micelles (amide and ester linkages)
Skin cancer lesions
Enhanced cancer-cell killing specificity with
superior safety to healthy skin cells)
[45]
HA–docetaxel conjugate
(HA–DTX–Dendron, HADD)
Docetaxel Conjugation of mal-GFLG-DTX,
mal-Cy5.5, and Dendron-8-Glu to HA
Triple negative breast cancer (TNBC)
HA in HADD increased DTX delivery to
tumor cells with rich CD44 receptors
interaction.
HADD inhibited tumor growth in
MDA-MB-231 tumor-bearing mice by
99.71%, compared to free DTX
[40]
HA–PEG/𝛼-CD hydrogel system Donepezil Polypseudorotaxane structure,
polydopamine bond-based
crosslinked HA hydrogel
The HD-PEG/𝛼-CD/PDM 8.5 injectable
hydrogel system gelled immediately and
delivered donepezil in sustained manner
via subcutaneous route
[46]
HA–hydroxychloroquine
conjugate (HA–HCQ)
Hydroxy chloroquine Conjugation of HA–HCQ via ester
linkage
In silico antiviral activity of HA–HCQ
toward different SARS-CoV-2 protein
molecular targets
[49]
HA–metformin (MET)
phospholipid sonocomplexes
(HA–MPS)
Metformin Conjugation of HA–MET via amide
linkage
Pancreatic ductal adenocarcinoma
Enhanced lipophilicity and targeting
potential of MET via CD44 binding
[50]
HA–doxorubicin conjugate
(HA–DOX)
Doxorubicin HA–DOX conjugates via nucleophilic
acyl substitution
Colorectal cancer
HA–Dox shows higher accumulation in the
intestinal regions upon intravenous
injection with enhanced inhibition of
colorectal cancer
[41]
HA-conjugated hybrid
nanoparticle system
Curcumin HA surface-conjugated PLGA–chitosan
nanoparticle (amide linkage)
Colon targeted drug delivery
HA functionalized nanohybrid particles are
effective in delivering loaded drugs orally
to the lower gastrointestinal tract (GIT)
[51]
HA–carnosine conjugates
(HyCar)
Carnosine Linking of HA to Car through an amide
bond
Neurodegenerative disorders (Alzheimer’s)
HyCar inhibits the formation of
amyloidtype aggregates and able to
dissolve the amyloid fibrils and to reduce
A𝛽-induced toxicity
[52]
HA–paclitaxel conjugate
(HA–PTX)
Paclitaxel Self-assembled HA–PTX polymeric
prodrug and methoxy poly(ethylene
glycol)–poly (lactide) block
copolymer (mPEG-PLA) to produce
nanoparticle composite (mPPHP
NPs)
Antitumor activity
mPPHP NPs preferentially accumulate in
the tumor site via EPR effect and enter
cancer cells by CD44-mediated endocytosis
[53]
HA–atorvastatin (ATV)
conjugate
Atorvastatin Conjugation of ATV-amine 7 with
carboxylic acid of HA via amide
linkage
Inhibition of atherosclerotic plaque
inflammation
Potential of delivering ATV, with CD44
receptor targetability in inflammatory
atherosclerotic plaques
[48]
PMX-conjugated HA
(HA-ADH-PMX)
Pemetrexed (PMX) HA-ADH-PMX synthesized by reacting
HA-ADH with
N-hydroxysuccinimide-activated
PMX (NHS-PMX)
For malignant pleural mesothelioma
(MPM) treatment
[54]
HA–QT conjugate micelles Quercetin Conjugates of HA–quercetin (HA–QT)
via ester linkage
A tumor cell-targeted prodrug was
developed for QT, using HA conjugate
micelle carrier
Shown enhanced inhibition on tumor
growth in H22 tumor-bearing mice
[55]
(Continued)
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (8 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 1 . (Continued).
Bioconjugation type Therapeutic agent Description of the conjugate and
linkage
Therapeutic application and outcome Refs.
HA-SS-DOX prodrug
self-assembled nanoparticles
Doxorubicin Hydrophilic HA-SS-N3 with a
hydrophobic DOX conjugate
copolymer self-assembled
nanoparticle
Targeted cancer chemotherapy
NPs were effectively internalized by tumor
cells overexpressed by the CD44 receptors
[56]
Cur–HA–SPu conjugate Curcumin Curcumin grafted HA modified pullulan
polymers by esterification
Cur–HA–SPu polymer showed accelerated
wound healing ability with anti-microbial,
antioxidant activities
[57]
HA–MTX conjugate Methotrexate HA–MTX conjugate with the ester
linkage
Rheumatoid arthritis (RA)
Ester linkage cleavable in a mildly acidic
environment of RA
HA–MTX conjugate has promising
potential for RA therapy
[58]
HA–cinnamaldehyde (CA)
nanofibers
Cinnamaldehyde CA conjugation to HA through
acid–labile hydrazone bond
Antitumor effect by economized
photodynamic therapy
Novel strategy that specifically boost tumor
oxidative stress for reinforced oxidation
therapy
[59]
HA–icariin (HA–Ica) conjugate
hydrogel
Icariin Methacrylic anhydride (MA)-modified
icariin (MIca) nanoparticles
dispersed in HA–MA hydrogel
HA–Ica hydrogel exhibited controlled
release of Ica with good cytocompatibilty.
Potentially used as a scaffold for cartilage
tissue engineering
[60]
Chol–Suc–HA conjugate
self-assembled nanoparticles
(NPs)
Docetaxel (DTX) Amphiphilic cholesteryl succinoyl HA
(Chol–Suc–HA) conjugates
self-assembled into DTX-loaded NPs
(ester linkage)
Chol–Suc–HA NPs were promising drug
vehicles for systemic targeting of
CD44-overexpressed cancers
[61]
Ab. Further, pyridyldisulfide group of PDPH–DRAb conjugated
to thiol group of kDa thiol end-modified HA. Interestingly, due
to the dual-targeting ability, HA–DR Ab conjugate seemed to be
highly accumulated in the liver and have shown eective inhi-
bition of tumor growth in liver metastasis mice model (Figure
7). Despite the advancements of HA-based macromolecular con-
jugates; molecular weight dependent eects, specific conjugates
characterization, purification and conjugates kinetic, mechanis-
tic aspects should be studied for further in order to enter into
clinics.
6. Combination of HA and Other Biopolymer
Crosslinked Systems
Growing evidences demonstrate that biopolymer scaolds can
be formulated by blending or crosslinking HA with naturally
derived or synthetic macromolecules for various biomedical
applications.[, ] Recent findings suggest that intra-articular
injections of nonchemically modified hybrid complex of high
molecular weight (HMW) sodium hyaluronate and sodium chon-
droitin without crosslinking agents has eectively reduced hip
pain and osteoarthritis functionality over time in symptomatic
hip osteoarthritis patients.[] The polymer interactions such as
hydrophobic and electrostatic interactions allow the combination
of the HA and other biopolymers such as collagen to form newly
blend matrices.
Interestingly, in another study, an optimal self-assembled bio-
component hydrogel of poly(ethyleneglycol) (PEG), crosslinked
type I/II collagen combined with HA has been demonstrated
to mimic the D inflammatory microenvironment of the in-
tervertebral disc that can regulate cell shape, extracellular ma-
trix content, and inflammatory signaling with understanding the
role of glycans.[] Biofunctionalization of collagen and high-
sulfated HA on the biodegradable synthetic scaold of three-
armed methacrylate-terminated macromers/PEG increased scaf-
fold pore size that has maintained cell viability, improved osteo-
genesis, and mineralization of new bone matrix.[] Besides, HA
has been also combined with laminin–hydrogel as a luminal filler
for collagen-based and chitosan-based nerve conduits that have
shown functional recovery and tissue regeneration in the sciatic
nerve injury model.[] The combination of HA with sodium al-
ginate (as bioink system, capable of being printed without em-
ploying chemical crosslinking agents) exhibited higher viscos-
ity, by maintaining the gross structure of the total hydrogel,[]
here HA increased the bioactivity of the encapsulated cells. Simi-
larly, the D-printed chitosan scaold, when combined with HA,
has shown a higher mechanical strength while maintaining its
structure, allowing chondrocytes to infiltrate into the scaold and
colonize the pores, forming fibroblast-like cells and promoting
chondrogenesis.[]
It is well known that chemical modification of functional
groups of HA will allow covalent crosslinking of the biopoly-
mer. The carboxylic group (COOH) can be modified through
the process of amidation, Ugi condensation, and ester forma-
tion, whereas the hydroxyl group (OH) of HA can be modified
via the ether, hemiacetal formation using glutaraldehyde; ester,
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (9 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 2 . Recent findings of HA–macromolecular conjugates to facilitate enhanced targeted delivery, and/or therapeutic effects.
Macromolecule type Description of conjugation and design Therapeutic application Research outcome Refs.
Interferon-𝛼(INF𝛼) Aldehyde-modified 100 kDa HA
dissolved in 5 mg mL1sodium
acetate buffer and conjugated with
IFN𝛼
Hepatitis C virus (HPC) Rate of intracellular uptake was
significantly higher for HA–INF𝛼system
HA–INF𝛼system showed significantly
enhanced targeting of liver and longer
residence time
[65]
Bovine serum albumin
(BSA)
BSA and 6.4 kDa HA conjugate with
paclitaxel
Targeted cancer therapy Improved selective internalization and
increased toxicity on CD44+cells for
BSA–HA system compared to HA and BSA
alone
[66]
Human serum albumin
(HSA)
HSA and erlotinib (ERT) NPs modified
with 5400 Da HA
Lung cancer ERT albumin nanoparticles modified with
HA/HSA showed superior antiproliferative
effect on lung carcinoma cells compared to
free ERT and ERT–HSA nanoparticles
[67]
Glucagon Microneedle array patch of
methacrylated-HA, combined with
insulin formed via polymerization
with crosslinker/photoinitiator post
UV irradiation
Hypoglycemia Microneedles were able to release
glucagon in response to elevated insulin
levels in interstitial fluid in vascular and
lymph capillary networks, preventing the
risk of hypoglycemia
[68]
Branched
oligoethyleneimine (bOEI)
bOEI-100 kDa HA conjugate system
synthesized via condensation
reaction
Ulcerative colitis (UC) Intraperitoneal injection of bOEI–HA in
UC-induced rats showed significantly less
inflammatory activity via less secretion of
TNF-𝛼, IL-6, and PGE2 compared to bOEI
alone
[69]
Epidermal growth factor
(EGF)
HA–EGF conjugate synthesized via
coupling reaction between
aldehyde-modified HA and
N-termine amine group in EGF
Skin wound healing and
regeneration
More efficient skin regeneration and
transdermal delivery from HA–EGF patch
into normal skin and peripheral tissues
near wound site than free EGF
[70]
Bilirubin (BR) 100 kDa HA–BR systems to facilitate
the formation of self-assembled
nanoparticles
Ulcerative colitis (UC) HA–BR system showed significantly
increased expression of anti-inflammatory
cytokines and decrease in proinflammatory
cytokines.
Improved microbiota diversity and effective
targeting via CD44 on the gastrointestinal
endothelium
[71]
O-aminophenol HA and o-aminophenol conjugated via
condensing reaction with carboxyl
groups of HA
Matrix metalloproteinase 2
(MMP-2) inhibition
Significant inhibition of MMP-2 expression
with HA-o-aminophenol conjugate
compared to HA conjugates with other
biologically active amines including
Procaine and Streptocide
[72]
Interferon alpha (INF𝛼) 2a Acetyl-modified HA conjugated to
INF𝛼2a via an aldehyde-quenching
reaction
Ovarian cancer HA–INF𝛼2a conjugate showed an
improved anticancer efficacy, significantly
enhanced survival compared to free
INF𝛼2a
[73]
Death receptor 5 antibody
(DR5 Ab)
Chemical conjugation pyridyldisulfide
group of PDPH–DR5Ab conjugated
to thiol group of 5 kDa thiol
end-modified HA
Liver metastasis In vivo bioimaging showed that HA–DR5
Ab conjugate highly accumulates in the
liver and is significantly more effective in
inhibiting tumor growth compared to DR5
Ab alone
[74]
Carnosine Carboxyl group on 200 and 700 kDa
HA conjugated with N-terminal
group of carnosine
Alzheimer’s disease (AD) HA–carnosine conjugate inhibits the
formation of A𝛽aggregates, dissolves
amyloid fibers, and reduces A𝛽-induced
toxicity in vitro compared to free carnosine
and HA alone
[52]
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (10 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 6. a) Schematic illustration of the branched oligoethyleneimine (bOEI) conjugate system synthesized via condensation reaction; conjugate physic-
ochemical properties such as b,c) bOEI-HA gel volume, d) surface charge/zeta potential, and e) N-acetylglucosamine release comparison between native
HA to bOEI-HA. Further, complete biological studies revealed that the conjugated form effectively modulated the inflammatory response, enhanced the
overall therapeutic efficacy in ulcerative colitis. Reproduced with permission.[69] Copyright 2021, Wiley-VCH GmbH.
carbamate formation, and oxidization with sodium periodate.[]
For example, tyramine-modified HA synthesis through oxidation
has been used for in situ crosslinking systems, employing hydro-
gen peroxide (HO) and horseradish peroxidase (HRP) to initi-
ate crosslinking reaction at the position CCandC
O between
phenols. When combined with a porous D silk fibroin matrix,
it can form a tunable hybrid scaold, which revealed the higher
chondroinductive and biomechanical properties tailored for car-
tilage repair.[] Amidation with carbodiimides is a common strat-
egy for HA modification in the carboxylic group. The initial step
in the reaction is the activation of the carboxylic acid of HA by
EDC, which results in the formation of an O-acyl isourea inter-
mediate. The nucleophilic attack of the amine on the activated
HA in the second step of the reaction results in the formation
of the amide bond.[] For example, activation of carboxyl groups
of HA through carbodiimide mediated-reaction using EDC cou-
pling with N-hydroxysuccinimide (NHS) to stabilize the inter-
mediate, results in the formation of the amide bond in the HA
structure that can react with succinimidyl groups of -arm PEG
amine to form crosslinked PEG/HA-based hydrogels.[] Vario us
crosslinked HA-hybrid systems are compared and enumerated
in Table 3.
Besides, PEG/HA hydrogel has shown a therapeutic eect
in preventing pain, specifically alleviating thermal hyperalge-
sia and mechanical allodynia, downregulating spinal nocicep-
tion markers, and hyperinnervation, altering glycosylation, and
modulate inflammatory and protein regulatory pathways for in-
tervertebral disc repair in vivo (Figure 8).[] In various studies,
-(,-dimethoxy-,,-triazin--yl)--methylmorpholinium chlo-
ride (DMTMM) was also widely used as a crosslinking initiator
in addition to EDC. The biphasic dispersion of crosslinked HA
in non-crosslinked HA solution was formulated using DMTMM
to initiate crosslinking reaction to form crosslinked PEG/HA that
exhibited a dendritic needle-like shape morphology, biphasic-HA
system decreased inflammation and permeability in an in vitro
interstitial cystitis model.[]
It is also well known that Schi base reaction is a mechanism
of the crosslinking system of HA that reacts between aldehyde
groups of modified-HA and amino groups of other polymers
to form a hydrogel. For example, the combination of HA and
gelatin hydrogel has been used to modify surface hydrophilic-
ity of implantable polydimethylsiloxane (PDMS) via the Schi-
base reaction of the aldehyde of HA (CHO-HA) and amine of
gelatin (Gel-NH) using EDC/NHS, thus forming the implant
that has higher biocompatibility and shown to improve extra-
cellular matrix deposition, and attenuate capsule formation and
inflammation in vivo.[] The crosslinking amino group of O-
carboxymethyl chitosan and aldehyde HA via the Schi-based re-
action forming the porous structure of the chitosan/HA hydro-
gel, the reported gel has the capability of promoting abdominal
endothelial regeneration.[] Dual-crosslinking systems consist-
ing of noncovalent and covalent binding have provided an e-
cient strategy to fine-tune hydrogel injectability, degradation, and
mechanical properties. For example, electrostatic interactions be-
tween cationic methacrylate chitosan and anionic HA allowed
Schi-based reaction to enable rapid gelation that can be used as
a cell delivery system and D printing of encapsulated cell scaf-
folds. The mechanical properties of the hydrogel network can
be improved through covalent crosslinking using in situ pho-
topolymerization of methacrylates.[] Various recent key HA-
based conjugate systems, and their potential interactions, diverse
biological responses are enumerated in Table 4.
HA has been regarded as a potential therapeutic mediator
for inflammatory bowel disease (IBD) in part due to its large
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (11 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 7. a) Schematic illustration of dual-targeting mechanism of HA–DR5 Ab conjugate by targeting DR5 and HA receptors and synthesis scheme. b)
In vivo bioimaging of the liver studies revealed that enhanced fluorescence signal was found with HA–DR5 Ab conjugate, compared to free or mixture.
c) The in vivo bioimaging and antitumor efficacy of HA–DR5 Ab conjugate in liver metastasis mice model Reproduced with permission.[74] Copyright
2016, American Chemical Society.
presence in the extracellular matrix of the gastrointestinal tract
(GIT) and similarity to mucins lining the GIT, suggesting that it
plays a role in increasing epithelial barrier integrity.[] In addi-
tion, HA’s targeting ability may also be exploited in IBD as the
CD receptor is expressed on endothelial cells in the GIT.[ ]
In one of the recent study, researchers formulated HA–
bilirubin (HA–BR) conjugated nanoparticles (mean size  ±
to  ± nm) for the targeted treatment of IBD, in which
researchers were able to facilitate the aqueous formulation of
BR and exploit HA–CD interactions to target inflamed colon
epithelium and proinflammatory macrophages via oral delivery
of the HA–BR nanomedicine system. The HA–BN system has
shown a potent reactive oxygen species (ROS)-scavenging eect
in protecting colonic epithelium against ROS-mediated cytotoxic-
ity, resistance to hyaluronidase-mediated degradation, modulated
gut microbiota, recovered tight junction proteins, reduced apop-
tosis, permitted intestinal barrier functions, and exhibited anti-
inflammatory eect in an acute colitis model of mice[] (Figure
9). In a similar study, Kotla et al. reported the development of an
enema suspension system based on hyaluronan (HA) that pro-
tects the dysregulated colon epithelium by decreasing inflamma-
tion, permeability, and retaining the epithelial barrier integrity
in dextran sodium’s sulphate (DSS)-induced colitis mice model
with proteomic profiling analysis.[]
7. HA Derivatives as Cell-Based Therapeutics
The biological significance of HA, specifically its recognition ca-
pacity to alter cell behavior, tissue mechanics, and regeneration
capabilities, paves the way for the development of biomateri-
als incorporating HA for cell-based systems. These scaolds has
unique therapeutic properties due to the nature of HA molecules
capable of interacting with proteins, including aggrecan, versi-
can, lymphatic vessel endothelial receptor (LYVE-), and tu-
mor necrosis factor-𝛼stimulated gene (TSG-), and mem-
brane receptors, including a CD, hyaluronectin, hyaluronan-
mediated motility (RHAMM), stretch-activated channels,[ ] and
Ca+channels.[ ] By the aforesaid interactions, HA plays a key
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (12 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 3 . The therapeutic applications of HA with other combinatorial biopolymer systems.
Polymer scaffold HA modification and crosslinking system Findings Refs.
Hybrid complex of sodium
hyaluronate and sodium
chondroitin
Blending of hyaluronate 2.4% and sodium
chondroitin 1.6%
Intra-articular administration was tolerated in patients
Efficacy of treatment to reduce visual analogue score of
hip pain
Efficacy of treatment to reduce osteoarthritis functionality
[84]
Type I and II collagen/HA hydrogels 4-arm-PEG-SG crosslinked collagen
blended with HA
Maintained the cell morphology of nucleus pulposus and
annulus fibrosus
Decrease extracellular matrix expression of type II
collagen and aggrecan
Regulate inflammation of IL-6 and TNF-𝛼, and suppressor
of cytokine signaling proteins (SOCS) signaling
[85]
Biofunctionalization of collagen and
high-sulfated HA on three-armed
methacrylate-terminated
macromers/PEG
Collagen and HA coating on freeze-dried
scaffold
Increased efficiency of coating with collagen and
sulphated HA on high pore scaffold
Coating stability over 7 days
Elevated of ALP for osteogenic marker
Histologically evidence of mineralized matrix
[86]
HA–laminin–hydrogel filler in
collagen-based and chitosan-based
nerve conduit
Blending of HA and laminin Functional motor recovery in compound muscle action
potential (CMAP) of the tibialis anterior
Reinnervation improve muscle mass–muscle weight
ratios of hind limb muscles
Reconstruction of sciatic nerve indicated by toluidine blue
staining. Regeneration of Schwann cells cultured in
hydrogel
[87]
Bioink of HA and sodium alginate
(SA) hydrogel
Blending of HA and sodium alginate for
3D bioprinting system
Viscosity of bioink increased with HA
HA enhances the bioactivity in the encapsulated cells
Printed hydrogel maintained its gross structure and
successfully printed layer-by-layer hydrogel upon addition
of CaCl2.
[88]
3D-printed chitosan/HA scaffold Blending of HA and chitosan for 3D
bioprinting system
Maintained the structure of scaffolds for up to 6 months
in water and 21 days in culture media
Chondrocytes penetrated scaffold, colonized the pores of
the scaffold
Increased expression of chondrogenic markers (collagen
type II, aggrecan and versican, SOX9 and RUNX2)
[89]
Silk fibroin matrix
and a HA hydrogel
Tyramine-modified HA crosslinked with
H2O2and HRP concentrations blended
in silk fibroin matrix
Increase in storage modulus (G) (elasticity), high viability
on human articular chondrocytes, upregulation of
COL2A1 and SOX9 expression
Surface morphology of heterogenous and macroporous
3D scaffold and fibroblastic-shape chondrocytes
Higher sulphated glycosaminoglycan content
[90]
Crosslinked PEG/HA hydrogel Carboxyl groups of HA with primary
amines of PEG to form the crosslinked
hydrogel (carbodiimide chemistry)
Hydrogel inhibited hyperinnervation of sensory nerve
fibers (GAP43), neuropeptides (CGRP), pronociceptor
(TrkA and TRPV1)
Hydrogel suppressed spinal nociception markers (c-Fos
and substance P), altered glycosylation and modulated
inflammatory signaling of IL-6 and IL-1𝛽.
Hydrogel regulated SMAD3/TGF-𝛽3 for matrix remodeling
[92]
Biphasic dispersion of crosslinked
HA in non-crosslinked HA solution
DMTMM initiated crosslinking of HA and
4-arm PEG
Maintained cell metabolic activity, decreased IL-8 level in
HTB-2 cells study
Decreased the apparent permeability of T84 cells
[93]
HA/gelatin hydrogel on PDMS
implant
Surface coating of hydrogel on PDMS
through Schiff-base reaction employing
aldehyde of HA (CHO-HA) and amine of
gelatin (Gel-NH2) using EDC/NHS
H-NMR of CHO–HA and gel–NH2spectra to inform
surface conjugation of PDMS and gelatin
Higher viability of human adipose-derived stem cells
Loose collagen fibers distribution, downregulation of
COL1A1, COL1A3, TGF-𝛽1, 𝛼-SMA, SMAD3, IL-𝛽1, and
IL-6
[94]
(Continued)
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (13 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 3 . (Continued).
Polymer scaffold HA modification and crosslinking system Findings Refs.
Chitosan/HA hydrogel Crosslinking amino group of
O-carboxymethyl chitosan and Aldehyde
HA via the Schiff-based reaction
Maintained viability of fibroblasts encapsulated in the
hydrogels with round shaped and uniformly distribution
Increase thickness of the regenerative endothelial tissue
Positive stained of CD31 and 𝛼-SMA for mature vascular
formation, positive stained of vimentin and COL1A2 for
extracellular matrix (ECM)
[95]
Dual-crosslinked chitosan
(CHMA)/HA (OHA) hydrogel
Methacrylate chitosan and aldehyde
functionalized HA via dynamic
electrostatic and Schiff-based reactions,
and in situ covalent chemistry using
photopolymerization of methacrylates
Steady moduli for crosslinked hydrogel under the time
sweep rheology, higher moduli for photopolymerization
hydrogel
Dynamic crosslinked hydrogel with reversable behavior
gel-liquid phase under the shear stress
Rat bone marrow derived stem cells encapsulated in
hydrogels had higher viability and homogenous cell
distribution
[96]
Figure 8. a). Schematic representation of crosslinked high-molecular weight HA and four-arm PEG amine hydrogel composite. b) Implantation of HA
hydrogel in the injured intravertebral disc region. c) Mitigation of the injury-induced pain phenotype by the implanted HA hydrogel, confocal schematic
representative staining demonstrated the presence in AF and NP tissues, particularly in the untreated injury group, of the GAP43 protein (yellow label)
for nerve ingrowth, the sensory neuropeptide CGRP (purple label), and TRPV1 (green label). Reproduced with permission.[92] Copyright 2018, American
Association for the Advancement of Science (AAAS).
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (14 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 4 . Various HA-based conjugate systems, potential interactions, and diverse biological responses.
HA system HA interactions and model applications Other findings Refs.
Acellular PEG/HA hydrogel Mechanism through HA–CD44 interaction
Model In vitro inflammation model of
nucleus pulposus (NP)
Hydrogel stability for up to one month in PBS
Noncytotoxic in nucleus pulposus cells
Attenuated inflammation, downregulated neurotrophic factor
[91]
DSPE–HA conjugates on
Gli1 siRNA nanoparticles
DSPE–HA binding affinity to CD44 receptor
via
Endocytosis
Model In vivo relapsed tumor models of
gastric CSCs
Successful conjugation of DSPE and HE indicated by spectrum
of methylene groups with a sharp absorbance peak of carbonyl
in the acyl chains of DSPE and peak of hydroxyl groups of HA
Increased of mean diameter of Gli1 siRNA nanoparticles
Cellular uptake of CD44+gastric cancer stem cells of siRNA
nanoparticles. Inhibited cellular proliferation of CD44+CSCs
[97]
Mesenchymal stem cell
extracellular vesicles with
HMW–HA
MSC EV bound to HMW HA results in the
interaction of CD44 receptor on
mesenchymal stem cell extracellular
vesicles (MSC EV)
Model Pseudomonas aeruginosa
(PA)-induced pneumonia in mice model
Increased MSC EV with HA uptake by human monocytes
Increased bacterial phagocytosis by LPS stimulated human
monocytes
MSC EV with CD44 siRNA treatment reduced bacterial
phagocytosis
Intravenous administration of MSC EV with HA-reduced
Pseudomonas aeruginosa CFU levels
[98]
HA–bilirubin
nanomedicine system
HA–CD44 interactions.
Model Ulcerative colitis mice model
HABN exhibited ROS-scavenging activity and protected
colonic epithelial cells from ROS-mediated cytotoxicity
HABN protected and recovered DSS-induced colitis phenotype
and improved intestinal barrier function of tight junction
associated proteins (ZO-1 and occluding-1), induced
expression of anti-microbial peptides (murine-𝛽defensin) and
regulated colonic microbiota
HABN reduced colonic epithelium apoptosis, attenuated
inflammation (IL-1𝛽,TNF-𝛼, IL-6), monocytes, and
neutrophils; increased IL-10 and TGF-𝛽
[71]
HA solution Interaction of HA and TRPV1 channels via
the hydrogel bonding on the positively
charged amino acid of TRPV1
Model Rats and TRPV1 null mice model
Reduced capsaicin evoked TRPV1 membrane ionic currents in
HEK-TRPV1- EYFP (+) cells; reduced TRPV1-single-channel
activity in HEK-TRPV1-EYFP (+) cells
Reduced latency of pain responses to heat following
subcutaneous injection of HA in the rat paw, and TRPV1 null
mice
Reduced capsaicin-evoked nerve impulse activity in saphenous
nerve of knee joint
Molecular modeling informed positively charged amino acids
(His614, Lys615, and Arg617) were the putative HA binding in
TRPV1 through H-bond with HA
ref. [99]
An injectable HA-based
microrods
Cell adhesion of fibroblasts on HA,
evidence of focal adhesion protein,
paxillin
Model Myocardial infarction of rats
Increased adhesion and spreading of neonatal rat ventricular
fibroblast on HA microrods. HA system decreased fibrosis
markers; decreased myoblast phenotype (COL1A2, alpha
smooth muscle actin (𝛼-SMA) through TGF-𝛽1/SMAD3
pathway.
HA system decreased matrix metalloproteinases (MMPs) of
MMP2 and MMP9 to prevent ECM breakdown
Increased thickness of left ventricular wall, deposition of loose
collagen fibers, stroke volume, ejection fraction in rats
Evidence of focal adhesion (actin, paxillin) at the edges of HA
microrods; positive stained of vimentin for fibroblasts and
CD68 for macrophages
[100]
HA solution Activation of TGF-𝛽/BMP signaling
Model In vitro study of human amniotic
epithelial cells (hAECs)
Increased proliferation of hAECs, maintained expression of
epithelial cell marker cytokeratin19 (CK19), CD29, CD73,
and CD166
Promoted stem cell pluripotent markers of Oct-4 and Nanog
Increased TGF-𝛽/BMP signaling
[101]
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (15 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 9. The schematic representation of hyaluronan–bilirubin nanoconjugate (HABN) role in modulating ulcerative colitis. Synthesized HABN system
showed to be nontoxic and localizes in inflamed regions of the colon of DSS-treated colitis mice and exerts therapeutic effects against acute colitis.
Reproduced with permission.[71] Copyright 2020, Springer Nature.
role in modulating biological processes such as cell survival, cell
migration, dierentiation, proliferation, and morphogenesis.[ ]
We have previously shown the therapeutic eect of crosslinked
PEG/HA hydrogel to attenuate inflammation and expression of
neurotrophic factors in an inflammation model of nucleus pul-
posus cells, possibly through binding to cell surface receptor,
CD[] and also in modulating the gut barrier integrity in colitis
condition.[]
Conjugation of di-stearoyl-phosphatidyl-ethanolamine-
hyaluronic-acid (DSPE-HA) on the small interfering RNA
(siRNA) nanoparticles has increased anity to CD, which is
capable of inhibiting glioma-associated oncogene homolog
(Gli) protein expression and gastric cancer stem cells (CSC)
tumor spheroid and colony formation, and suppress cell migra-
tion, invasion, and evidence of antitumor activity in vivo.[] The
interaction of the CD receptor on mesenchymal stem cell ex-
tracellular vesicles (MSC EV) was associated with antimicrobial
activity, demonstrating an increased binding of MSC EV bound
to HMW HA led to higher MSC EV uptake and bacterial phago-
cytosis, attenuated inflammation, and decreased the bacterial
load in Pseudomonas aeruginosa-induced pneumonia in mice.[]
The HA molecule plays a vital role in regulating cell adhe-
sion for cell–cell communications and modulation of diverse bi-
ological functions and matrix homeostasis. Injectable HA-based
microrods have been shown to increase cell adhesion and to
spread rat ventricular fibroblast on HA microrods with evidence
of focal adhesion protein (paxillin and actin) that contributed to
mechanotransduction pathways, led to higher cell proliferation,
decrease expression of myofibroblast phenotype through TGF-𝛽
and SMAD pathway in vitro. Upon injecting HA microrods in a
rat model of myocardial infarction, focal adhesion (actin and pax-
illin), and positively stained vimentin were shown, with increased
ventricular wall thickness and deposition of loose collagen, thus
improving cardiac function including stroke volume and ejection
fraction in rats.[ ] By the activation of TGF-𝛽/BMP signaling,
the HA has been reported to promote proliferation of human
amniotic epithelial cells (hAECs), maintain epithelial markers,
increase levels of anti-inflammatory factors (IL- and TGF-𝛽),
and stem cell pluripotent markers of Oct and Nanog.[]
The cross-talk between osteopontin (OPN), integrin 𝛼𝜈𝛽, and
HA was demonstrated in the osteoarthritis (OA) model, in which
the expression of HA was regulated by the interaction between
OPN and integrin 𝛼𝜈𝛽.[ ] The HA has been reported to inter-
act with the ion channel of the nociceptor to exhibit an analgesic
eect. The HA solution inhibited heat and capsaicin evoked-
intracellular calcium concentration in TRPV-EYFP channels ex-
pressed in HEK cells and dorsal root ganglia culture and pre-
vented TRPV sensitization by bradykinin, confirming the inter-
action of HA on TRPV channels of sensitized peripheral sen-
sory terminals. In in vivo, HA reduces the sensitivity of noci-
ceptor endings to noxious stimuli transduced by TRPV and de-
creased TRPV-mediated nerve impulse activity of joint nocicep-
tor fibers. The interaction of HA and TRPV was modeled via the
hydrogel bonding of HA to the positively charged amino acids of
TRPV.[] The interaction of the HA molecule and postsynap-
tic L-type Ca+channels has been implicated in modulating hip-
pocampal synaptic plasticity, in which exogenous administration
of HA has been shown to increase Cav. mediated current.[]
The HA-based systems have been widely used as a cell-delivery
system to mimic the D microenvironment of native tissue and
support cell transplantation for tissue regeneration. Treatment
of human mesenchymal stem cells (hMSCs) suspended with
batroxobin/platelet-rich plasma/HA hydrogel in nucleotomized
intervertebral disc organ culture has maintained cell viability, hy-
drogel integration with NP tissue, promoted hMSCs dierenti-
ation toward NP phenotype indicated by upregulation of aggre-
can and cytokeratin- (KRT) gene expression, representing a
promising stem cell delivery for disc regeneration.[ ]
Intra-articular injection of mesenchymal stromal cells with
HA solution has demonstrated an increase in cartilage thick-
ness and deposition of type I collagen in temporomandibular
joint osteoarthritis of the rabbit model.[ ] Jha et al. reported
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (16 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
in comparison to the other crosslinkers examined, hyaluronic
acid (HyA) hydrogels crosslinked with the MMP-degradable pep-
tide QPQGLAK promote the highest cardiac progenitor cells sur-
vival, proliferation, and endothelial cell dierentiation.[ ] In an-
other study, combination treatment of catechol-functionalized
HA patch and adipose-derived mesenchymal stem cells (ADSCs)
has demonstrated the reduced size of the diabetic wound, in-
creased angiogenesis markers of CD and von Willebrand fac-
tor, and paracrine signaling molecules of VEGF, IGF-, FGF-,
ANG-, PIK, and AKT in diabetic wound mouse models.[ ] In-
corporation of ADSCs on the composite hydrogels of methacry-
lated HA containing fragmented polycaprolactone electrospun
nanofibers (NFs) has demonstrated elongated morphology of
cells, higher cell proliferation and osteogenic dierentiation
with evidence of type I collagen (COL), alkaline phosphatase
(ALP), and runt-related transcription factor (RUNX).[ ] Chi-
tosan/HA scaold provided an excellent microenvironment for
supporting human ADSCs dierentiation toward cartilage indi-
cated by higher cell proliferation and viability with cells attached
to scaold depositing new matrix of type II collagen and ag-
grecan, and upregulation of chondrogenic markers of ACAN,
COLA, and SOX.[ ]
8. HA as a Biocoating Polymer and Its Therapeutic
Prospective
HA has been widely employed for the modification of the surface
of several delivery systems. In this sense, HA has found particu-
lar interest in the modification of nanoparticles thanks to its an-
ity for CD. This fact perfectly complements the passive target-
ing promoted by the enhanced permeability and retention (EPR)
eect associated with nanoentities and could overcome the lim-
itations of the current approaches toward clinical translation. In
a particular example,[ ] latex nanoparticles were surface modi-
fied with ten dierent natural or synthetic polyions via the layer-
by-layer (LbL) approach (Figure 10a). The carboxylated polyions
(i.e., poly(-glutamic acid), poly(-aspartic acid), poly(acrylic acid),
and HA) display an increased anity toward ovarian cancer cell
lines with respect to sulfated counterparts (Figure b). Although
the binding ability of HA-modified particles was lower than the
one observed for the other carboxylated polyions, they were ac-
cumulated within intracellular compartments, whereas poly(-
glutamic acid)- and poly(-aspartic acid)-modified nanoparticles
were preferentially observed at the cell membrane (Figure c).
Moreover, as concluded from the in vivo studies, HA allows
deeper penetration of the nanoparticles in cancerous tissues with
respect to other surface-modified nanoparticles, which can be
ascribed to its antifouling and swelling behavior (Figure d).
These properties have thus been used to improve the ecacy and
delivery of chemotherapeutic drugs (e.g., raltitrexed) and photo-
sensitizers for photodynamic therapy in a variety of in vitro and
in vivo cancer models.[, ]
Modification of nanoparticles with HA is thus regularly
adopted as a common strategy to ensure the performance of novel
therapies for cancer. This includes also cooperative approaches
where several treatments are combined to boost their synergis-
tic actions. For example, HA-decorated polymer nanoparticles
loaded with a small drug (i.e., docetaxel) in the core and coated
with a photosensitizers (i.e., anionic porphyrin) ensured ecient
uptake and colocalization of the therapeutic agents within can-
cerous cells (i.e., MDA-MB-), tremendously increasing light-
induced cell death.[ ] Besides, considering that HA is degraded
in the tumor microenvironment because of the increased levels
of hyaluronidase, HA-coated nanoparticles can preferentially de-
liver their payload at the target tissue.[, ] More complex sys-
tems have been reported in the bibliography that use HA to in-
crease the blood circulation time and tumor accumulation of na-
noentities. In a particular approach,[ ] HA was conjugated to
the surface of the nanoparticles via pH-sensitive imine bonds. In
the tumor microenvironment, HA was deshielded from the sur-
face, exposing the chitosan layer underneath and promoting cell
uptake. The ability of HA to specifically bind CD has also been
used to capture tumor cells, which could be potentially used as a
diagnosis system with increased sensitivity and selectivity.[]
HA is commonly employed together with positively charged
polyions (e.g., chitosan) to coat the surface of other biomate-
rials via the LbL approach or alternative procedures.[ ] The
modification of HA with dopamine endows adhesive proper-
ties and facilitates its incorporation on a wide variety of sub-
strates, representing an alternative or complementary method
to the widely employed LbL approach for the modification of
surfaces.[ ] In this sense, the surface of polymeric biomate-
rials was coated with polyvinylamine (PVAm) and dopamine-
modified HA via the LbL approach.[ ] Michael-type addition and
Schi base reaction between amine groups of PVAm and cate-
chol groups of dopamine facilitated the assembly process, such
coating prevented the growth of Escherichia coli and Staphylococ-
cus aureus and suppressed bacterial growth, demonstrating its
potential to create antibacterial and biocompatible coatings. Sim-
ilarly, HA/chitosan multilayer films were assembled on silicon
substrates and inhibited the adhesion and growth of Staphylococ-
cus aureus and Pseudomonas aeruginosa.[ ]
Considering the huge impact of implant-related infections,
that represent more than % of hospital acquired infections, the
development of advanced biomaterials that inhibit bacterial adhe-
sion and biofilm formation is highly desirable. Thus, antibacte-
rial drugs are usually combined within multilayer films to create
a synergistic eect between these two approaches. For example,
gentamicin sulfate was loaded into polyacrylic acid/chitosan mul-
tilayer films, which were subsequently sealed with an external
layer of HA.[ ] The release of the entrapped gentamicin sulfate
was accelerated in the presence of hyaluronidase secreted by bac-
teria, thus allowing a triggered drug release. In a similar exam-
ple, multilayer films of montmorillonite and HA were assembled
on several substrates[ ] (Figure 11a). The degradation of HA in
response to hyaluronidase promoted the release of the drug (Fig-
ure b) and resisted the pathogen adhesion through film peel-
ing (Figure c). Consequently, biofilm formation was inhibited,
thus reducing immune response and bacterial infections in an in
vivo model.
Together with bacterial infections, the integration of implants
with the surrounding tissues represents one of the main clin-
ical problems associated with biomaterials. When the integra-
tion with the surrounding bone (i.e., osseointegration) is incom-
plete, micromotions occur at the interface between the bone
and the implant, which finally results in implant failure. Here
again, the use of HA has played a pivotal role. Hybrid (i.e.,
organic/inorganic) multilayer films made from HA, chitosan,
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (17 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 10. a) Schematic representation of the layer-by-layer modification of latex nanoparticles with carboxylated and sulfated polyions. b) Fluorescence
intensity of ovarian cancer cells incubated with different configuration of nanoparticles, indicating that carboxylated nanoparticles have more affinity
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (18 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
and bioactive glass nanoparticles were grown on glass substrates
and imparted bioactivity to the otherwise bioinert support, as
demonstrated by the formation of apatite-like structures on their
surfaces.[ ] Similar coatings composed of dopamine-modified
HA and chitosan improved the adhesion and proliferation of os-
teoblasts with respect to the pristine titanium alloy used as a
control.[ ] To further promote osteoblast adhesion and prolif-
eration, HA/chitosan multilayer films were decorated with RGD
peptide (i.e., arginine–glycine–aspartic acid) via carbodiimide
chemistry or formation of disulfide bonds.[, ]
Moreover, considering the intrinsic antibacterial properties
of HA/chitosan multilayer films discussed above, this strategy
could solve two of the main problems encountered in orthopaedic
implants, namely, bacterial infections and poor osseointegration.
Despite the adhesion of osteoblasts in vitro seems to be poorer
in HA-modified implants than in pristine counterparts,[ ] in
vivo studies concluded that surface modification with HA re-
inforces the implant-tissue interface, consequently improving
osseointegration.[ ] It was hypothesized in this study that the
outermost layer of HA is dissolved with time, exposing the sub-
sequent chitosan layer and improving the integration of the ti-
tanium alloy implant. HA has also found great potential to im-
prove lubrication of biomaterials at articulating interfaces (e.g.,
implants for meniscus replacement),[ ] which is vital to reduce
irritation, pain, and inflammation of the implant. Dopamine-
modified HA and polylysine multilayer films were assembled on
than sulfated ones toward cancerous cells. c) Confocal images of various carboxylated nanoparticles incubated with ovarian cancer cells, demonstrating
that HA-modified nanoparticles are internalized by cells and not bound to their membrane. Cell membranes are stained in red, nuclei in blue, and
nanoparticles appear in green. Scale bar: 10 μm. d) Fluorescence images of tumor sections 24 h after nanoparticle administration in mice bearing
orthotopic ovarian cancer xenografts. Red and blue signals correspond to H&E staining, while nanoparticles appear in green (panel above). In the lower
panel, z-stacks from the whole-tumor are represented. Blue signal refers to collagen, while red signal is associated to the mCherry signal from tumor cells.
Nanoparticles appear in green. Scale bar: 100 μm. The quantification on the z-stacks demonstrate that hyaluronic acid-coated nanoparticles penetrate
deeper in the tissue. Reproduced with permission.[113 ] Copyright 2020, American Chemical Society.
Figure 11. a) Schematic representation of the layer-by-layer approach to coat a planar substrate with hyaluronic acid (HA)/montmorillonite (MMT) and
loaded with gentamicin (GS). Enzymatic degradation in presence of hylaruonidase triggers the release of the encapsulated drug and the peeling of the
external film. b) Gentamicin release from the multilayer films in the presence of increasing concentrations of hyaluronidase (HAS) demonstrating the
responsiveness of the system. c) Bacterial colonies before (a,b) and after (a,b) incubation with the multilayer film for 1 h. The bar graph represents the
number of bacterial colonies in each case. Reproduced with permission.[126 ] Copyright 2018, American Chemical Society.
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (19 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
polycarbonate urethane substrates. The coating promoted the re-
cruitment of a lubricious glycoprotein (i.e., PRG) from synovial
fluid and helped to dissipate only one-third of the frictional en-
ergy and reduce the coecient of friction with respect to the pris-
tine material.
Interestingly, metallic implants such as magnesium alloys,
have also benefited from HA-based coatings.[, ] In a partic-
ular example where magnesium alloys were explored as cardio-
vascular stents, polydopamine was used as an adlayer for the
subsequent immobilization of HA via covalent interactions. The
coating prolonged the degradation time of the biomaterial, while
promoting endothelialization (i.e., increased expression of CD
of vascular endothelial cells was observed in vitro). Besides, the
blood compatibility was improved (i.e., less aggregation of red
blood cells was observed in vitro) and the inflammation was at-
tenuated (i.e., lower macrophage attachment in vivo) in the HA-
coated samples with respect to the pristine magnesium allow,
which opens the possibility of this material as a coronary stent.
In many occasions, biomaterials employed as cell substrates
lack in bioactivity which enormously limit their potential use as
scaolds for tissue engineering applications. Coating the surface
of these materials with extracellular matrix components such as
HA has been also considered in this field as a strategy to im-
prove the performance of scaolds for the regeneration of sev-
eral tissues including tendon,[ ] ligaments,[ ] vocal folds,[ ]
and cartilage.[ ] For the regeneration of tendon, aligned poly(𝜖-
caprolactone) (PCL) electrospun mats were coated with HA and
chitosan via the LbL approach. In comparison to the noncoated
PCL scaold, tendon stem/progenitor cells displayed increased
expression of tenogenic markers in vitro and also in an in vivo
model of degenerative rotator cu tendon.[ ] This was ascribed
to a higher hydrophilicity and wettability provided by the multi-
layer coating, which can better resemble the conditions found in
a real cellular microenvironment.
In a dierent example,[ ] PCL electrospun microfibers were
functionalized with HA via LbL deposition based on bioorthogo-
nal reaction between s-tetrazines and trans-cyclooctenes, which
were coupled to the HA backbone. Moreover, the bioorthogonal
chemistry proposed in this study was fast, avoids long incuba-
tion times to create multilayer films. The created scaolds had
interesting features for the regeneration of vocal folds since they
were able to preserve the phenotype of vocal fold fibroblasts and
avoided their dierentiation toward myofibroblast. This is of vital
importance to suppress fibrogenesis and promoting tissue repair.
At a more basic level, the properties of HA have been exploited
to create cell substrates with controlled stiness[ ] or micropat-
terned surfaces,[ ] which enable to study important aspects of
cell biology. In the former case,[ ] poly--lysine/hyaluronic acid
multilayer films were grown on glass slides and their stiness
was controlled by dierent crosslinking degrees via the carbodi-
imide chemistry. The stiness of the substrate clearly influenced
cell fate, being stier films more appropriate for cell adhesion
and spreading. In the latter case,[ ] the cell-repulsive properties
of HA were used to create micropatterned surfaces that allow the
coculture of dierent cell lines with spatial resolution. HA has
also been involved in “substrate-mediated gene delivery.”[] In
a particular example, HA-based multilayer films were used as a
platform for the encapsulation and immobilization of gene vec-
tors, allowing more localized and ecient gene delivery. Finally,
within the field of HA as a biocoating polymer, the direct coating
of injured/damaged tissues with HA should be highlighted.[ ]
For example, a multilayer film at the nanoscale made out of chi-
tosan and HA was directly deposited on a damaged artery. The
coating prevented blood coagulation and promoted the healing
process by inhibiting restenosis and vascular cell proliferation.
9. HA-Based Inorganic Conjugate Composites
Nanomaterials including carbon, gold, silver, magnetic and
mesoporous materials have distinct physiochemical and biologi-
cal characteristics, making them attractive for use in various drug
delivery, biosensing, diagnostic, imaging, and therapy.[] These
nanomaterials, however, are potentially cytotoxic and lack cell-
specific activities, as a result, these nanocarriers have been func-
tionalized with a variety of biopolymers including HA.[ ] Var i-
ous studies have been conducted in recent years on the develop-
ment of inorganic nanocomposites functionalization with biolog-
ical molecules, polysaccharides, biopolymers, and carbohydrates
to make them less-cytotoxic, with enhanced targetability, and cell-
specificity etc.[ ] The ability of HA to target specific cells via in-
teracting with cell surface receptors such as CD, RHAMM has
been documented by a large number of studies, and it has the
potential to be used for tumor-targeted medication delivery.[ ]
In the recent years, HA-based functionalization of inorganic
nanocomposites has gained a lot of attention for their potential
to deliver target-specific payload via cell receptor interactions with
enhanced cell-specific delivery applications. Of late, from the ma-
rine mussel inspired properties (adhesive and cohesive) of cate-
chol (CA), Lee et al. developed anticancer drug DOX loaded sil-
ica core and catechol-modified HA (HA-CA) shell delivery carrier
system with pH-triggered release behavior with enhanced cyto-
toxic activity in cancer cells, in vitro[ ] (Figure 12).
In a similar line of research HA has been explored to surface
functionalize on hybrid silica nanocarriers with enriched thera-
nostic applications. The novel organic/inorganic hybrid vesicle
(HA–docetaxel/perfluoro-n-pentane@silica nanocapsule–HA–
DTX/PFP@SNC) system has shown a stronger signal compared
to DTX/PFP@SNC due to high stability, ultrasound-triggered
drug release with HA-mediated tumor targeting.[ ] An attempt
has been made to increase the therapeutic ecacy of anti-
cancer drugs (including -Fluorouracil) to target colon cancer
by HA-conjugated -FU silica nanoparticles with enhanced
drug transport into tumors than normal tissues;[ ] together
these findings suggest that HA-integration on silica carriers can
be helpful in designing advanced, cell-compatible, safer drug
delivery, and contrasting agent systems. Multimodal imaging
systems based on several modalities have gained a lot of attention
in recent years for non-invasive targeted imaging applications.
Studies were reported on developing new enzyme-specific
superparamagnetic iron oxide nanoparticles applied as a multi-
modal probe in magnetic resonance imaging and hyaluronidase
(HAdase)-sensitive optical imaging applications.[ ]
Gold-hybrid nanomaterials have exhibited great promise as
nanomedicine agents due to their optical properties, includ-
ing the surface plasmon resonance eect. Recent studies have
demonstrated the formulation of gold-nanoclustered HA (GNc-
HyNA) assemblies for photodynamic tumor ablation applica-
tion, in which tumors were completely ablated with %
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (20 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Figure 12. a) Pictorial illustration of silica core with modified-HA–catechol shell formation with different pH conditions. b) Enhanced cell-death was
observed on breast cancer cells (MDA-MB-231) with particles form compared to free-DOX. Reproduced with permission.[147 ] Copyright 2017, Elsevier.
Figure 13. a) Schematic representation of the HA–FA–DOX gold nanorod conjugate system for synergistic chemo and photothermal therapy. b) Com-
pared with non-FA-decorated nanoparticles, the FA–HA conjugated nanoparticles showed enhanced antitumor effects against MCF-7 breast tumors in
nude mice. Reproduced with permission.[152 ] Copyright 2017, American Chemical Society.
survival rate in an orthotopic breast cancer model.[ ] In a
similar approach of multimodal targetability with photother-
mal chemotherapy application, Xu et al. developed a pH/near-
infrared (NIR) dual-triggered drug release nanocarrier system
with HA-functionalized gold nanorods (GNRs) for actively tar-
geted synergetic photothermal chemotherapy (using DOX) for
breast cancer therapy. The DOX, folate-conjugated novel GNRs–
HA–FA–DOX system showed an excellent stability with sus-
tained, dual-responsive drug release behavior with targeted syn-
ergistic breast cancer treatment[ ] (Figure 13).
In recent years, graphene oxide (GO) has attracted tremen-
dous attention for its application in anticancer therapies us-
ing HA nanohybrid conjugates. Song et al reported HA–GO–
DOX nanohybrid system, which was synthesized by conjugat-
ing GO with HA (by H bond interactions) for targeted deliv-
ery of DOX; compared to free DOX the HA–GO–DOX showed
a higher tumor inhibition eect in an H hepatic cancer cell
bearing mouse model.[ ] Similarly, HA-conjugated titanium–
graphene oxide (HA-Ti@GO) with indocyanine green (ICG)
system,[ ] MGO@CD–CA–HA hybrid system[ ] (coating of
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (21 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
Tabl e 5 . Recent advancements, findings of HA–inorganic composite systems and biomedical applications.
Type of carrier Components of the delivery
system
Conjugation type In vitro/in vivo
study
Therapeutic application and findings Refs.
Silica Doxorubicin (DOX) loaded
silica core with
catechol-modified HA
(HA-CA) shell system
Amide linkage
(HA–Dopamine)
In vitro pH-triggered release behavior with
enhanced cytotoxic activity in cancer cells
[147]
Silica HA–docetaxel/perfluoro-n-
pentane@silica nanocapsule
(HA-DTX/PFP@SNC)
system
Amide linkage (HA with
surface -NH2 of SNC
In vitro +In vivo Theranostic application (imaging with
delivery)
High stability, ultrasound-triggered drug
release with HA-mediated tumor targeting
[148]
Iron oxide Paclitaxel (PTX) loaded iron
oxide nanoparticles with
modified dopamine-bovine
serum albumin (BSA)/HA
Amide linkage
(Fe3O4·DA-BSA/HA-
PTX)
In vitro Fe3O4·DA-BSA/HA was capable of
entrapping PTX with enhanced
bioavailability
Time-domain (TD)-NMR experiments
revealed the system suitability as contrast
agents in MRI
[156]
Iron oxide +
graphene oxide
Doxorubicin (Dox) and
paclitaxel (Ptx) loaded
HA–GO composite with
iron-oxide nanoparticles
Amide linkage
(GO–HA)
In vitro GO–HA–Dox/Ptx system effectively killed
CD44-expressing MDA-MB-231 cells but
not BT-474 cells (no CD44 expression)
with additional magnetothermal
functionality
[157]
Fe3O4–graphene
oxide
MGO@CD-CA-HA hybrid
system (coating of
𝛽-cyclodextrin–cholic
acid–HA polymer
(CD-CA-HA) onto the
Fe3O4–graphene oxide
(MGO))
Amide linkages (HA to
CPT, HA to CD)
In vitro +In vivo Multifunctional hybrid
photo-chemotherapy nanosystem
Synergistic multiple targeting ability with
chemo-photothermal therapy for
hepatoma treatment
[155]
Graphene oxide HA–GO–DOX nanohybrid
system
H-bonding interactions In vitro +In vivo Compared to free DOX the HA-GO-DOX
showed higher tumor inhibition effect in
an H22 hepatic cancer cell bearing mice
[153]
Gold (Gold nanorods,
GNRs)–HA–FA–DOX system
Au–catechol bonds,
hydrazone
linkages (HA–DOX)
In vitro +In vivo DOX, folate-conjugated novel
GNRs–HA–FA–DOX system showed
excellent stability with sustained,
dual-responsive drug release behavior
with targeted synergistic breast cancer
treatment
[152]
Titanium–
graphene oxide
HA-conjugated
titanium–graphene oxide
(HA-Ti@GO) with
indocyanine green (ICG)
drug
Amide bond In vitro +In vivo HA-Ti@GO/ICG multifunctional
tumor-targeted and NIR light-induced ICG
delivery system
Significantly increased ICG accumulation
in tumor tissue and prolonged in vivo
circulation of ICG
[154]
𝛽-cyclodextrin–cholic acid–HA polymer (CD–CA–HA) onto the
FeO–graphene oxide (MGO)) were reported for synergistic
chemo-photothermal combination therapies. Various recent ad-
vancements and findings of HA-conjugated inorganic based de-
livery systems are reported in Table 5.
10. Conclusion and Future Prospects
Although the continuation of native HA and modified-HA sys-
tems and their use in the biomedical applications has been doc-
umented for decades, it is only recently that research eorts on
HA are aimed at pharmacological, pathological, biological matrix
remodeling aspects in multiple human disease processes with
underlying mechanisms. This review has explored a variety of
chemical and biological aspects to be considered in developing
HA-based therapeutic conjugate systems for various biomedical
applications with latest research findings.
Despite positive results from several in vitro and in vivo ex-
perimental studies, it is surprising that very few clinical trials
with HA-based conjugates are being investigated. The number
of clinical trials success rate using conjugate systems in preci-
sion medicine has been quite limited in recent years. One of
the ongoing trials, ONCOFID-P-B (paclitaxel–HA conjugate) is
being studied in phase-III clinical trial to determine its eec-
tiveness and safety in patients with Bacillus Calmette-Guérin-
unresponsive bladder cancer.[,] Similarly, in another recently
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (22 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
completed trial HYMOVIS (partial hexadecylamide of HA conju-
gate, clear hydrogel) device was used to evaluate the beneficial
eects for knee osteoarthritis.[, ]
Besides, with the exception of Declage (developed by LG Life
Sciences in Korea) there is no commercially available drug deliv-
ery product based on HA, however, from the ongoing bioconjuga-
tion research eorts, it is expected that HA oers a great poten-
tial as a novel carrier with tunable biomimetic scaold proper-
ties for various therapeutics interventions. Nevertheless, minor
dierences in HA composition, molecular weight, type of drug
and application, and degree of conjugation alter the outcomes of
HA conjugates and must be considered when progressing those
technologies toward clinical translation.
Modified-HA systems and their biology continue to generate
interest, future studies on HA-based conjugates should focus on
including essential elements such as the utilization of HA molec-
ular weight, optimal degree of substitution, and conjugate con-
struct pharmacological and biopharmaceutical properties includ-
ing PK-PD assessments. Besides, in order to be a successful con-
jugate based product, the synthesized system should be very sim-
ple, i.e., easy preparation or scale-up, should preferably be made
of generally recognized as safe components without any toxic im-
munogenic properties. Given its unique physicochemical, phar-
macological, tissue remodeling characteristics with continuous
worldwide research eorts, HA will be successfully utilized for
the commercialization of various biopharmaceutical, medical de-
vice systems within the foreseeable future.
Acknowledgements
P.K.V. thanks Department of Biotechnology, Institute for Stem Cell Science
and Regenerative Medicine for core funds. A.L. is thankful for funds from
the Basque Government, Department of Education (IT-1766-22). I.L.M.I.
would like to thank funding agencies; Universiti Kebangsaan Malaysia un-
der the Geran Galakan Penyelidik Muda (Grant No. GGPM-2022-029),
Malaysian Ministry of Higher Education for the Fundamental Research
Grant Scheme (Grant No. FRGS/1/2022/SKK0/UKM/02/12), and Health
Research Board, Ireland under the Emerging Investigator Award for Health
2022 (Grant No. EIA-2022-010). Some of the figures were created by us-
ing BioRender.com. All authors contributed to the manuscript preparation
and approved.
Conflict of Interest
The authors declare no conflict of interest.
Author Contributions
N.G.K.: article conception; N.G.K., I.L.M.I., A.L., B.M., S.S., and G.S.: liter-
ature survey and writing; N.G.K., A.L., G.S., and P.K.V.: editing and review-
ing; All the authors: final approval.
Keywords
bioconjugation, CD44, HA-bioconjugates, hyaluronan, hyaluronic acid
Received: November 29, 2022
Revised: March 4, 2023
Published online:
[1] B.P.Toole,Nat. Rev. Cancer 2004,4, 528.
[2] A. Fallacara, E. Baldini, S. Manfredini, S. Vertuani, Polymers 2018,
10, 70.
[3] N. G. Kotla, S. Reddy, S. Rasala, J. Wankar, R. A. Bohara, J. Bayry, Y.
Rochev, A. Pandit, J. Controlled Release 2021,336, 598.
[4] R. C. Gupta, R. Lall, A. Srivastava, A. Sinha, Front. Vet. Sci 2019,6,
192.
[5] T. V. Pereira, P. Jüni, P. Saadat, D. Xing, L. Yao, P. Bobos, A. Agarwal,
C. A. Hincapié, B. R. Da Costa, BMJ 2022,378, e069772.
[6] S. Amorim, C. A. Reis, R. L. Reis, R. A. Pires, Trends Biotechnol. 2021,
39, 90.
[7] T. N. Wight, Matrix Biol. 2017,60–61, 38.
[8] T. Murakami, S. Otsuki, Y. Okamoto, K. Nakagawa, H. Wakama, N.
Okuno, M. Neo, Connect. Tissue Res. 2019,60, 117.
[9] T. E. Riehl, D. Alvarado, X. Ee, M. A. Ciorba, W. F. Stenson, Am. J.
Physiol.: Gastrointest. Liver Physiol. 2020,319, G63.
[10] A. Cañibano-Hernández, L. Saenz Del Burgo, A. Espona-Noguera,
G. Orive, R. M. Hernández, J. Ciriza, J. L. Pedraz, Mol. Pharmaceutics
2019,16, 834.
[11] J. Lam, W. E. Lowry, S. T. Carmichael, T. Segura, Adv. Funct. Mater.
2014,24, 7053.
[12] L. T. Zhang, R. M. Liu, Y. Luo, Y. J. Zhao, D. X. Chen, C. Y. Yu, J. H.
Xiao, Life Sci. 2019,232, 116669.
[13] M. Litwiniuk, A. Krejner, T. Grzela, Wounds 2016,28, 78.
[14] H. Yang, L. Song, Y. Zou, D. Sun, L. Wang, Z. Yu, J. Guo, ACS Appl.
Bio Mater. 2021,4, 311.
[15] F. Zamboni, S. Vieira, R. L. Reis, J. Miguel Oliveira, M. N. Collins,
Prog. Mater. Sci. 2018,97, 97.
[16] K. N. How, W. H. Yap, C. L. H. Lim, B. H. Goh, Z. W. Lai, Fron t.
Pharmacol. 2020,11.
[17] Y. Zheng, J. Yang, J. Liang, X. Xu, W. Cui, L. Deng, H. Zhang,
Biomacromolecules 2019,20, 4135.
[18] N. G. Kotla, I. Liza, M. Isa, S. Rasala, S. Demir, R. Singh, B. V. Baby,
S. K. Swamy, P. Dockery, V. R. Jala, Adv. Sci. 2021,2103189, 2013189.
[19] E. J. Strauss, J. A. Hart, M. D. Miller, R. D. Altman, J. E. Rosen, Am.
J. Sports Med. 2009,37, 1636.
[20] M. Gaumet, I. Badoud, P. Ammann, Osteoarthr. Cartil. 2018,26,1.
[21] G. Abatangelo, V. Vindigni, G. Avruscio, L. Pandis, P. Brun, Mol. Cells
2020,9, 1743.
[22] N. Gallo, H. Nasser, L. Salvatore, M. L. Natali, L. Campa, M. Mah-
moud, L. Capobianco, A. Sannino, M. Madaghiele, Eur. Polym. J.
2019,117, 134.
[23] J. Necas, L. Bartosikova, P. Brauner, J. Kolar, Vet. Med 2008,53, 397.
[24] M. K. Cowman, S. Matsuoka, Carbohydr. Res. 2005,340,791.
[25] P. S. Chang, L. T. McLane, R. Fogg, J. Scrimgeour, J. S. Temenoff, A.
Granqvist, J. E. Curtis, Biophys. J. 2016,110, 2739.
[26] K. J. Wolf, S. Kumar, ACS Biomater. Sci. Eng. 2019,5, 3753.
[27] Z. Zhu, Y.-M. Wang, J. Yang, X.-S. Luo, Plast. Aesthetic Res. 2017,4,
219.
[28] C. Kuehl, T. Zhang, L. M. Kaminskas, C. J. H. Porter, N. M. Davies,
L. Forrest, C. Berkland, Mol. Pharmaceutics 2016,13, 1904.
[29] C. E. Schanté, G. Zuber, C. Herlin, T. F. Vandamme, Carbohydr.
Polym. 2011,85, 469.
[30] M. O. Longas, J. D. Burden, J. Lesniak, R. M. Booth, J. A. McPencow,
J. I. Park, Biomacromolecules 2003,4, 189.
[31] T. Borke, F. M. Winnik, H. Tenhu, S. Hietala, Carbohydr. Polym. 2015,
116, 42.
[32] I. Danishefsky, E. Siskovic, Thromb. Res. 1972,1, 173.
[33] V. Crescenzi, A. Francescangeli, D. Capitani, L. Mannina, D. Renier,
D. Bellini, Carbohydr. Polym. 2003,53, 311.
[34] K. Hirano, S. Sakai, T. Ishikawa, F. Y. Avci, R. J. Linhardt, T. Toida,
Carbohydr. Res. 2005,340, 2297.
[35] S. A. Bencherif, A. Srinivasan, F. Horkay, J. O. Hollinger, K. Maty-
jaszewski, N. R. Washburn, Biomaterials 2008,29, 1739.
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (23 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
[36] J. E. Prata, T. A. Barth, S. A. Bencherif, N. R. Washburn, Biomacro-
molecules 2010,11, 769.
[37] H. X. Ngo, S. Garneau-Tsodikova, MedChemComm 2018,9, 757.
[38] D. Harrer, E. Sanchez Armengol, J. D. Friedl, A. Jalil, M. Jelkmann,
C. Leichner, F. Laffleur, Int. J. Pharm. 2021,601, 120589.
[39] S. Hu, E. Lee, C. Wang, J. Wang, Z. Zhou, Y. Li, X. Li, J. Tang, D. H.
Lee, X. Liu, Y. Shen, J. Controlled Release 2015,220, 175.
[40] W. Wang, X. Zhang, Z. Li, D. Pan, H. Zhu, Z. Gu, J. Chen, H. Zhang,
Q. Gong, K. Luo, Carbohydr. Polym. 2021,267, 118160.
[41] D. C. Pan, V. Krishnan, A. K. Salinas, J. Kim, T. Sun, S. Ravid, K. Peng,
D. Wu, M. Nurunnabi, J. A. Nelson, Z. Niziolek, J. Guo, S. Mitragotri,
Bioeng. Transl. Med. 2021,6, e10166.
[42] Z. Xu, W. Zheng, Z. Yin, Arch. Pharm. Chem. Life Sci. 2013,347, 240.
[43] M. Zhang, C. Xu, L. Wen, M. K. Han, B. Xiao, J. Zhou, Y. Zhang, Z.
Zhang, E. Viennois, D. Merlin, Cancer Res. 2016,76, 7208.
[44] Sauraj, S. U. K., P. Gopinath, Y. S. Negi, Carbohydr. Polym. 2017,157,
1442.
[45] V. Krishnan, K. Peng, A. Sarode, S. Prakash, Z. Zhao, S. K. Filippov,
K. Todorova, B. R. Sell, O. Lujano, S. Bakre, A. Pusuluri, D. Vogus,
K. Y. Tsai, A. Mandinova, S. Mitragotri, Sci. Adv. 2021,7, 24.
[46] C. R. Hwang, S. Y. Lee, H. J. Kim, K. J. Lee, J. Lee, D. D. Kim, H. J.
Cho, Carbohydr. Polym. 2021, 266, 118104.
[47] S. Hossaini Nasr, A. Tonson, M. H. El-Dakdouki, D. C. Zhu, D. Ag-
new, R. Wiseman, C. Qian, X. Huang, ACS Appl. Mater. Interfaces
2018,10, 11495.
[48] S. Hossaini Nasr, Z. Rashidijahanabad, S. Ramadan, N. Kauffman,
N. Parameswaran, K. R. Zinn, C. Qian, R. Arora, D. Agnew,X. Huang,
Nanoscale 2020,12, 9541.
[49] R. Thirumalaisamy, V. Aroulmoji, M. N. Iqbal, M. Deepa, C.
Sivasankar, R. Khan, T. Selvankumar, J. Mol. Struct. 2021,1238,
130457.
[50] M. M. Farag, N. S. Abd El Malak, S. A. Yehia, M. A. Ahmed, Int. J.
Nanomed. 2021, 1005.
[51] N. G. Kotla, O. Burke, A. Pandit, Y. Rochev, Nanomaterials 2019,9,
1246.
[52] V. Greco, I. Naletova, I. M. M. Ahmed, S. Vaccaro, L. Messina, D.
L. Mendola, F. Bellia, S. Sciuto, C. Satriano, E. Rizzarelli, Sci. Rep.
2020,10, 15998.
[53] K. Luo, S. Yin, R. Zhang, H. Yu, G. Wang, J. Li, Int. J. Pharm. 2020,
589.
[54] Y. Amano, S. Ohta, K. L. Sakura, T. Ito, Eur. J. Pharm. Sci. 2019,138,
105008.
[55] X. Pang, Z. Lu, H. Du, X. Yang, G. Zhai, Colloids Surf., B 2014,123,
778.
[56] B. Lu, F. Xiao, Z. Wang, B. Wang, Z. Pan, W. Zhao, Z. Zhu, J. Zhang,
ACS Biomater. Sci. Eng. 2020,6, 4106.
[57] Y. Duan, K. Li, H. Wang, T. Wu, Y. Zhao, H. Li, H. Tang, W. Yang,
Carbohydr. Polym. 2020,238, 116195.
[58] 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, Chem. Commun. 2014,50, 7632.
[59] X.Xu,B.Huang,Z.Zeng,J.Chen,Z.Huang,Z.Guan,M.Chen,Y.
Huang, C. Zhao, Theranostics 2020,10, 10513.
[60] L. He, J. Yang, J. Lu, Y. Xiao, Y. Fan, X. Zhang, Mater. Lett. 2014,136,
41.
[61] S. Song, H. Qi, J. Xu, P. Guo, F. Chen, F. Li, X. Yang, N. Sheng, Y. Wu,
W. Pan, Pharm. Res. 2014,31, 2988.
[62] A. Mero, M. Campisi, Polymers 2014,6, 346.
[63] B. Chen, R. J. Miller, P. K. Dhal, J. Biomed. Nanotechnol. 2014,10,4.
[64] E. J. Oh, K. Park, K. S. Kim, J. Kim, J. A. Yang, J. H. Kong, M. Y. Lee,
A. S. Hoffman, S. K. Hahn, J. Controlled Release 2010,141,2.
[65] K. S. Kim, H. Hyun, J. A. Yang, M. Y. Lee, H. Kim, S. H. Yun, H. S.
Choi, S. K. Hahn, Biomacromolecules 2015,16, 3054.
[66] R. Edelman, Y. G. Assaraf, I. Levitzky, T. Shahar, Y. D. Livney, Onco-
Targets Ther. 2017,8, 24337.
[67] Y. Shen, W. Li, Drug Des., Dev. Ther. 2018,12, 2285.
[68] J. Yu, Y. Zhang, W. Sun, A. R. Kahkoska, J. Wang, J. B. Buse, Z. Gu,
Small 2017,13, 1603028.
[69] A. Nishiguchi, T. Taguchi, Adv. Funct. Mater. 2021,31, 2100548.
[70] Y. S. Kim, D. K. Sung, W. H. Kong, H. Kim, S. K. Hahn, Biomater. Sci.
2018,6, 1020.
[71] Y. Lee, K. Sugihara, M. G. Gillilland, S. Jon, N. Kamada, J. J. Moon,
Nat. Mater. 2020,19, 118.
[72] I. Y. Ponedel’Kina, A. R. Gaskarova, E. A. Khaybrakhmanova, E. S.
Lukina, V. N. Odinokov, Carbohydr. Polym. 2016,144, 17.
[73] I. M. Montagner, A. Merlo, D. Carpanese, A. Dalla Pietà, A. Mero,
A. Grigoletto, A. Loregian, D. Renier, M. Campisi, P. Zanovello, G.
Pasut, A. Rosato, J. Controlled Release 2016,236, 79.
[74] H. Lee, B. J. Hong, J. H. Lee, S. Yeo, H. Y. Jung, J. Chung, G. O. Ahn,
S. K. Hahn, Biomacromolecules 2016,17, 3085.
[75] Z. Zhang, J. Zhang, M. Jiang, L. Zhao, S. Li, H. Sun, F. Yang, H. Liang,
Mol. Pharmaceutics 2020,17, 1405.
[76] T. Ito, I. P. Fraser, Y. Yeo, C. B. Highley, E. Bellas, D. S. Kohane, Bio-
materials 2007,28, 1778.
[77] E. E. Friedrich, L. T. Sun, S. Natesan, D. O. Zamora, R. J. Christy, N.
R. Washburn, J. Biomed. Mater. Res., Part B 2014,102, 1527.
[78] Y. Jin, R. H. Koh, S. H. Kim, K. M. Kim, G. K. Park, N. S. Hwang,
Mater. Sci. Eng., C 2020,115, 111096.
[79] A. Akhtar, A. Andleeb, T. S. Waris, M. Bazzar, A. R. Moradi, N. R.
Awan, M. Yar, J. Controlled Release 2021,330, 1152.
[80] W. C. Ballance, E. C. Qin, H. J. Chung, M. U. Gillette, H. Kong, Bio-
materials 2019,217, 119292.
[81] S. Trujillo, S. L. Vega, K. H. Song, A. San Félix, M. J. Dalby, J.
A. Burdick, M. Salmeron-Sanchez, Adv. Healthcare Mater. 2020,9,
2000989.
[82] Y. Luo, K. R. Kirker, G. D. Prestwich, J. Controlled Release 2000,69,
169.
[83] J. A. Burdick, G. D. Prestwich, Adv. Mater. 2011,23, H41.
[84] R. Papalia, V. Salini, N. Voglino, M. Fortina, S. Carta, F. Sadile, C.
Costantino, Ther. 2021,8, 151.
[85] A. Srivastava, I. L. Mohd Isa, P. Rooney, A. Pandit, Biomaterials 2017,
123, 127.
[86] J. Krieghoff, A. K. Picke, J. Salbach-Hirsch, S. Rother, C. Heinemann,
R. Bernhardt, C. Kascholke, S. Möller, M. Rauner, M. Schnabelrauch,
V. Hintze, D. Scharnweber, M. Schulz-Siegmund, M. C. Hacker, L.
C. Hofbauer, C. Hofbauer, Biomater. Res. 2019,23, 26.
[87] Z. Huang, S. Kankowski, E. Ertekin, M. Almog, Z. Nevo, S. Rochkind,
K. Haastert-Talini, Int. J. Mol. Sci. 2021,22, 6554.
[88] S. J. Lee, J. M. Seok, J. H. Lee, J. Lee, W. D. Kim, S. A. Park, Polymers
2021,13, 794.
[89] E. De Angelis, R. Saleri, P. Martelli, L. Elviri, A. Bianchera, V. Conti,
R. Bettini, B. Passeri, F. Ravanetti, Front Vet Sci 2021,8, 671776.
[90] J. T. Weitkamp, M. Wöltje, B. Nußpickel, F. N. Schmidt, D. Aibibu,
A. Bayer, D. Eglin, A. R. Armiento, P. Arnold, C. Cherif, R. Lucius, R.
Smeets, B. Kurz, P. Behrendt, Int. J. Mol. Sci. 2021,22, 3635.
[91] I. L. M. Isa, A. Srivastava, D. Tiernan, P. Owens, P. Rooney, P. Dock-
ery, A. Pandit, Biomacromolecules 2015,16, 1714.
[92] I. L. Mohd Isa, S. A. Abbah, M. Kilcoyne, D. Sakai, P. Dockery, D. P.
Finn, A. Pandit, Sci. Adv. 2018,4, eaaq0597.
[93] P. R. Rooney, V. K. Kannala, N. G. Kotla, A. Benito, D. Dupin, I.
Loinaz, L. R. Quinlan, Y. Rochev, A. Pandit, J. Biomed. Mater. Res.,
Part B 2021,109, 864.
[94] H. Joo, J. Park, C. Sutthiwanjampa, H. Kim, T. Bae, W. Kim, J. Choi,
M. Kim, S. Kang, H. Park, Pharmaceutics 2021,13, 269.
[95] Y. Deng, J. Ren, G. Chen, G. Li, X. Wu, G. Wang, G. Gu, J. Li, Sci. Rep.
2017,7, 2699.
[96] C. Han, H. Zhang, Y. Wu, X. He, X. Chen, Sci. Rep. 2020,10, 14997.
[97] H. Yao, L. Sun, J. Li, X. Zhou, R. Li, R. Shao, Y. Zhang, L. Li, Int. J.
Nanomed. 2020,15, 7013.
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (24 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
[98] L. Zhou, Q. Hao, S. Sugita, Y. Naito, H. He, C. Chung Yeh, J. W. Lee,
Stem Cell Res. Ther. 2021,12, 293.
[99] R. Caires, E. Luis, F. J. Taberner, G. Fernandez-Ballester, A. Ferrer-
Montiel, E. A. Balazs, A. Gomis, C. Belmonte, E. De La Peña, Nat.
Commun. 2015,6, 8095.
[100] L. V. Le, P. Mohindra, Q. Fang, R. E. Sievers, M. A. Mkrtschjan, C.
Solis, C. W. Safranek, B. Russell, R. J. Lee, T. A. Desai, Biomaterials
2018,169, 11.
[101] Y. B. Tian, N. X. Wang, Y. Xu, C. Y. Yu, R. M. Liu, Y. Luo, J. H. Xiao,
PeerJ 2020,8, e10104.
[102] C. A. De La Motte, S. P. Kessler, Int. J. Cell Biol. 2015,481301, 481301.
[103] E. d. L. Pea, S. Sala, J. C. Rovira, R. F. Schmidt, C. Belmonte, Pain
2002,99, 501.
[104] G. Kochlamazashvili, C. Henneberger, O. Bukalo, E. Dvoretskova,
O.Senkov,P.M.J.Lievens,R.Westenbroek,A.K.Engel,W.A.Cat-
terall, D. A. Rusakov, M. Schachner, A. Dityatev, Neuron 2010,67,
116.
[105] K. T. Dicker, L. A. Gurski, S. Pradhan-Bhatt, R. L. Witt, M. C. Farach-
Carson, X. Jia, Acta Biomater. 2014,10, 1558.
[106] Y. Yuan, Q. Liu, Z. Wu, W. Luo, Biomed Res. Int. 2020,2020, 2905634.
[107] F. Russo, L. Ambrosio, M. Peroglio, W. Guo, S. Wangler, J. Gewiess,
S. Grad, M. Alini, R. Papalia, G. Vadalà, V. Denaro, Int. J. Mol. Sci.
2021,22, 2963.
[108] R. Köhnke, M. O. Ahlers, M. A. Birkelbach, F. Ewald, M. Krueger, I.
Fiedler, B. Busse, M. Heiland, T. Vollkommer, M. Gosau, R. Smeets,
R. Rutkowski, Int. J. Mol. Sci. 2021,22, 443.
[109] A. K. Jha, K. M. Tharp, S. Browne, J. Ye, A. Stahl, Y. Yeghiazarians, K.
E. Healy, Biomaterials 2016,89, 136.
[110] C. S. Pak, C. Y. Heo, J. Shin, S. Y. Moon, S. W. Cho, H. J. Kang, Int. J.
Mol. Sci. 2021,22, 2632.
[111] M. Patel, W. G. Koh, Pharmaceutics 2020,12, 902.
[112] Y. Huang, D. Seitz, F. König, P. E. Müller, V. Jansson, R. M. Klar, Int.
J. Mol. Sci. 2019,20, 4487.
[113] S. Correa, N. Boehnke, A. E. Barberio, E. Deiss-Yehiely, A. Shi, B.
Oberlton, S. G. Smith, I. Zervantonakis, E. C. Dreaden, P. T. Ham-
mond, ACS Nano 2020,14, 2224.
[114] J. G. Rosch, M. R. Landry, C. R. Thomas, C. Sun, Nanoscale 2019,
11, 13947.
[115] Z. Hou, K. Deng, C. Li, X. Deng, H. Lian, Z. Cheng, D. Jin, J. Lin,
Biomaterials 2016,101, 32.
[116] S. Maiolino, F. Moret, C. Conte, A. Fraix, P. Tirino, F. Ungaro, S.
Sortino, E. Reddi, F. Quaglia, Nanoscale 2015,7, 5643.
[117] W. H. Chen, G. F. Luo, W. X. Qiu, Q. Lei, L. H. Liu, S. B. Wang, X. Z.
Zhang, Biomaterials 2017,117, 54.
[118] F. Zheng, C. Wang, T. Meng, Y. Zhang, P. Zhang, Q. Shen, Y. Zhang,
J. Zhang, J. Li, Q. Min, J. Chen, J. J. Zhu, ACS Nano 2019,13,
12577.
[119] H. Chen, X. Fan, Y. Zhao, D. Zhi, S. Cui, E. Zhang, H. Lan, J. Du,
Z. Zhang, S. Zhang, Y. Zhen, ACS Appl Mater Interfaces 2020,12,
22074.
[120] J. B. M. Rocha Neto, T. B. Taketa, R. A. Bataglioli, S. B. Pimentel, D.
M. Santos, A. Fiamingo, C. A. R. Costa, S. P. Campana-Filho, H. F.
Carvalho, M. M. Beppu, Appl. Surf. Sci. 2019,486, 508.
[121] O. Etienne, A. Schneider, C. Taddei, L. Richert, P. Schaaf, J. C. Voegel,
C. Egles, C. Picart, Biomacromolecules 2005,6, 726.
[122] A. I. Neto, A. C. Cibrão, C. R. Correia, R. R. Carvalho, G. M. Luz, G.
G. Ferrer, G. Botelho, C. Picart, N. M. Alves, J. F. Mano, Small 2014,
10, 2459.
[123] T. Di Zhang, X. Deng, Y. F. Wang, X. T. Wang, X. Zhang, L. L. Chen,
X. Cao, Y. Z. Zhang, C. Y. Zhang, X. Zheng, D. C. Yin, Appl. Surf. Sci.
2020,530, 147197.
[124] J. Hernandez-Montelongo, E. G. Lucchesi, I. Gonzalez, W. A. A.
Macedo, V. F. Nascimento, A. M. Moraes, M. M. Beppu, M. A. Cotta,
Colloids Surf., B 2016,141, 499.
[125] P. Ayaz, B. Xu, X. Zhang, J. Wang, D. Yu, J. Wu, Appl. Surf. Sci. 2020,
527, 146806.
[126] B. Wang, H. Liu, L. Sun, Y. Jin, X. Ding, L. Li, J. Ji, H. Chen, Biomacro-
molecules 2018,19, 85.
[127] A. C. Almeida, A. C. Vale, R. L. Reis, N. M. Alves, Int. J. Biol. Macro-
mol. 2020,157, 119.
[128] X. Zhang, Z. Li, X. Yuan, Z. Cui, X. Yang, Appl. Surf. Sci. 2013,284,
732.
[129] Y. Huang, Q. Luo, X. Li, F. Zhang, S. Zhao, Acta Biomater. 2012,8,
866.
[130] P. H. Chua, K. G. Neoh, E. T. Kang, W. Wang, Biomaterials 2008,29,
1412.
[131] S. Zankovych, M. Diefenbeck, J. Bossert, T. Mückley, C. Schrader, J.
Schmidt, H. Schubert, S. Bischoff, M. Faucon, U. Finger, K. D. Jandt,
Acta Biomater. 2013,9, 4926.
[132] H. Wan, X. Zhao, C. Lin, H. J. Kaper, P. K. Sharma, ACS Appl Mater
Interfaces 2020,12, 23726.
[133] J. an Li, L. Chen, X. qi Zhang, S. kang Guan, Mater. Sci. Eng., C 2020,
109, 110607.
[134] Y. K. Kim, S. Y. Kim, Y. S. Jang, I. S. Park, M. H. Lee, Appl. Surf. Sci.
2020,515, 146070.
[135] F. Han, P. Zhang, X. Wen, C. Lin, P. Zhao, Biomater. Sci. 2019,7,
4388.
[136] S. Deepthi, K. Jeevitha, M. Nivedhitha Sundaram, K. P. Chennazhi,
R. Jayakumar, Chem. Eng. J. 2015,260, 478.
[137] A. Ravikrishnan, H. Zhang, J. M. Fox, X. Jia, ACS Macro Lett. 2020,
9, 1369.
[138] T. J. Levingstone, E. Thompson, A. Matsiko, A. Schepens, J. P. Glee-
son, F. J. O’brien, Acta Biomater. 2016,32, 149.
[139] K. Ren, L. Fourel, C. G. Rouvière, C. Albiges-Rizo, C. Picart, Acta
Biomater. 2010,6, 4238.
[140] J. Fukuda, A. Khademhosseini, J. Yeh, G. Eng, J. Cheng, O. C.
Farokhzad, R. Langer, Biomaterials 2006,27, 1479.
[141] C. Husteden, F. Doberenz, N. Goergen, S. R. Pinnapireddy,C. Janich,
A. Langner, F. Syrowatka, A. Repanas, F. Erdmann, J. Jedelská, U.
Bakowsky, T. Groth, C. Wölk, ACS Appl. Mater. Interfaces 2020,12,
8963.
[142] B. Thierry, F. M. Winnik, Y. Merhi, M. Tabrizian, J. Am. Chem. Soc.
2003,125, 7494.
[143] X. Wang, X. Zhong, J. Li, Z. Liu, L. Cheng, Chem.Soc.Rev.2021,50,
8669.
[144] Z. Cai, H. Zhang, Y. Wei, F. Cong, Biomacromolecules 2017,18,
1677.
[145] K. E. Sapsford, W. R. Algar, L. Berti, K. B. Gemmill, B. J. Casey,
E. Oh, M. H. Stewart, I. L. Medintz, Chem. Rev. 2013,113,
1904.
[146] K. Y. Choi, G. Saravanakumar, J. H. Park, K. Park, Colloids Surf., B
2012,99, 82.
[147] J. Lee, K. C. Yoo, J. Ko, B. Yoo, J. Shin, S.-J. Lee, D. Sohn, Carbohydr.
Polym. 2017,164, 309.
[148] Q. Yang, H. Chen, Y. Bai, Y. Cao, W. Hu, L. Zhang, Adv. Healthcare
Mater. 2018,7, 1700816.
[149] K. Liu, Z. Q. Wang, S. J. Wang, P. Liu, Y. H. Qin, Y. Ma, X. C. Li, Z. J.
Huo, Int. J. Nanomed. 2015,10, 6445.
[150] D. E. Lee, A. Y. Kim, G. Saravanakumar, H. Koo, I. C. Kwon, K. Choi,
J. H. Park, K. Kim, Macromol. Res. 2011,19, 861.
[151] H. S. Han, K. Y. Choi, H. Lee, M. Lee, J. Y. An, S. Shin, S. Kwon, D.
S. Lee, J. H. Park, ACS Nano 2016,10, 10858.
[152] W. Xu, J. Qian, G. Hou, A. Suo, Y. Wang, J. Wang, T. Sun, M. Yang,
X. Wan, Y. Yao, ACS Appl. Mater. Interfaces 2017,9, 36533.
[153] E. Song, W. Han, C. Li, D. Cheng, L. Li, L. Liu, G. Zhu, Y. Song, W.
Tan, ACS Appl. Mater. Interfaces 2014,6, 11882.
[154] H. Zhang, X. Zhang, X. Zhu, J. Chen, Q. Chen, H. Zhang, L. Hou, Z.
Zhang, Nanomedicine: NBM 2018,14, 73.
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (25 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de
[155] C. Wen, R. Cheng, T. Gong, Y. Huang, D. Li, X. Zhao, B. Yu, D. Su, Z.
Song, W. Liang, Colloids Surf., B 2021,199, 111510.
[156] E. Vismara, C. Bongio, A. Coletti, R. Edelman, A. Serafini, M. Mauri,
R. Simonutti, S. Bertini, E. Urso, Y. G. Assaraf, Y. D. Livney, Molecules
2017, 1030.
[157] N. Pramanik, S. Ranganathan, S. Rao, K. Suneet, S. Jain, A. Rangara-
jan, S. Jhunjhunwala, ACS Omega 2019,4, 9284.
[158] P. F. Bassi, M. Racioppi, G. Palermo, C. Selli, M. Battaglia, P.
Gontero, C. Simeone, A. Eisendhardt, F. Vom Dorp, A. Neisius, M. F.
Arjona, D. Castro Diaz, M. Ramirez Backhaus, J. Moreno Sierra, C.
Llorente, G. Nohales Taurines, N. Giordan, A. Marinello, Eur. Urol.
2018,17, e1056.
[159] R. Hurle, G. F. Guazzoni, P. Colombo, A. Santoro, O. De Cobelli, E.
Di Trapani, G. Nohales, L. Carlos, R. Duran-Merino, M. Lazzeri, J.
Clin. Oncol. 2020,38, 486.
[160] Y. Henrotin, R. Bannuru, M. Malaise, H. K. Ea, C. Confavreux, J.
Bentin, D. Urbin-Choffray, T. Conrozier, J. P. Brasseur, P. Thomas,
A. C. Hick, A. Marinello, N. Giordan, P. Richette, BMC Musculoskele-
tal Disord. 2019,20, 293.
[161] Y. Henrotin, R. Bannuru, M. Malaise, H.-K. Ea, C. Confavreux, J.
Bentin, D. Urbin-Choffray, T. Conrozier, J.-P. Brasseur, P. Thomas,
A. Marinello, N. Giordan, P. Richette, Osteoarthritis Cartilage 2018,
26, S289.
Niranjan G. Kotla was awarded Ph.D. in biomedical engineering by University of Galway Ireland (2020)
and obtained specialist M. Pharmacy (2013), B. Pharmacy (2011) degrees. He is an innovation post-
doctoral fellow at NIBR-Novartis, Basel-Switzerland. He is an expert in drug delivery (targeted, long-
acting, controlled release), medical devices, formulation development. His research focused on
advanced drug delivery technologies development(s) using polymer chemistry,biomaterials, and
nanomedicine approaches. He is also the beneficiary of individual fellowships (FWO, Novartis inno-
vation postdoc), Julia Polak European doctorate awardee. He has published over 30 peer-reviewed
articles, books, conference abstracts; involved in various academic and industrial research collabora-
tions.
Gandhi Sivaraman received his M.Sc. in 2009 from The American College, Madurai, and Ph.D. in 2014
from Madurai Kamaraj University.Then he continued to pursue his postdoctoral research at Insti-
tute for stem cell science and regenerative medicine (inStem), Bangalore. Currently,he is working as
faculty in Gandhigram Rural Institute -Deemed to be University,Gandhigram, India. His research in-
terest includes chemical biology,biosensors, drug delivery systems, and computational chemistry.
His h-index is 41 and he has authored over 80 articles with over 4000 citations.
Praveen K. Vemula is an associate professor at inStem, Bangalore. He has completed master’s in
chemistry from Osmania University,and obtained Ph.D. from IISc , Bangalore. His expertise is de-
veloping chemical technologies for medical applications. His work spans the fields of biomateri-
als, drug discovery,drug delivery, medical devices, and chemical biology. He has published over 85
peer-reviewed papers and over 30 issued or pending national/international patents, which have been
licensed to multiple biotech companies. His technologies have led to the launch of six startup com-
panies. Thus far,over 20 products that are developed based on his technologies are in the market
worldwide.
Adv. Healthcare Mater. 2023, 2203104 © 2023 Wiley-VCH GmbH
2203104 (26 of 26)
21922659, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202203104 by NATIONAL UNIVERSITY OF IRELAND GALWAY, Wiley Online Library on [17/04/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
... In this study, we aimed to prolong the degradation time of HA scaffolds beyond the typical 24 h timeframe, observed in vivo. To achieve this, we employed divinyl sulfone (DVS) as a crosslinking agent during polymerization [6,[12][13][14]. The use of DVS implies a risk of toxicity due to the reactivity of its vinyl groups if the remnants of this molecule are not completely washed from the material [12,15]. ...
... HA is a biodegradable hydrogel approved by the US Food and Drug Administration (FDA), the most advantageous characteristics [6] of which are related to its hygroscopic properties [7] via the maintenance of the extracellular matrix of the cells upon hydration. HA is also known to preserve the viscoelastic properties of tissues, quickly adapt to varying environments, and activate cell receptors in an autocrine or paracrine manner, modulating ...
... The development of scaffolds, medical devices, and bioconjugate systems based on HA has extended to a multitude of medical and research applications [6], since the abundance of HA in mammalian tissues with different biological roles and with chemical simplicity for modifications makes it an attractive material with a rising global market. Both native and modified HA systems have experienced a great progression thanks to their promising biophysical, biochemical, viscoelastic, and tissue-remodeling properties [6]. ...
Article
Full-text available
The objective of this study was to develop and characterize a novel hyaluronic acid (HA) 3D scaffold integrated with gelatin microparticles for sustained-delivery applications. To achieve this goal, the delivery microparticles were synthesized and thoroughly characterized, focusing on their crosslinking mechanisms (vanillin and genipin), degradation profiles, and release kinetics. Additionally, the cytotoxicity of the system was assessed, and its impact on the cell adhesion and distribution using mouse fibroblasts was examined. The combination of both biomaterials offers a novel platform for the gradual release of various factors encapsulated within the microparticles while simultaneously providing cell protection, support, and controlled factor dispersion due to the HA 3D scaffold matrix. Hence, this system offers a platform for addressing injure repair by continuously releasing specific encapsulated factors for optimal tissue regeneration. Additionally, by leveraging the properties of HA conjugates with small drug molecules, we can enhance the solubility, targeting capabilities, and cellular absorption, as well as prolong the system stability and half-life. As a result, this integrated approach presents a versatile strategy for therapeutic interventions aimed at promoting tissue repair and regeneration.
... This study used amino fluorescein, a fluorescent marker, to label hyaluronic acid after functionalizing it with tyramine. As a result of the shorter subcutaneous gelling time, enzymatic processes are a good way for hydrogel creation since they may be used to establish crosslinking and are also compatible with cells [28]. ...
... The development of a hydroxyapatite/carbon nanotube/HA CNT composite with superior mechanical and biological properties is crucial for the usage of bone replacements in order to resemble true bone tissue [28]. In addition to facilitating the flow of bodily fluids, the composite's unique top has an uneven, wet surface that is ideal for cell adhesion, development, and proliferation. ...
... (*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001). challenge by conjugating MSA2 to HA, ameliorating its solubility profile and cell uptake as previously demonstrated with other drugs [59,60]. Compared to our previous approaches [53,61], the advantages reside in a simultaneous trigger of both innate and adaptive immunity with a single immunotherapeutic agent after an intracranial injection. ...
Article
Full-text available
Glioblastoma (GBM) is an aggressive brain tumor, with a highly immunosuppressive tumor immune microenvironment (TIME). In this work, we investigated the use of the STimulator of INterferon Genes (STING) pathway as an effective means to remodel the GBM TIME through the recruitment of both innate and adaptive immune cell populations. Using hyaluronic acid (HA), we developed a novel polymer-drug conjugate of a non-nucleotide STING agonist (MSA2), called HA-MSA2 for the in situ treatment of GBM. In JAWSII cells, HA-MSA2 exerted a greater increase of STING signaling and upregulation of STING-related downstream cyto-/chemokines in immune cells than the free drug. HA-MSA2 also elicited cancer cell-intrinsic immunostimulatory gene expression and promoted immunogenic cell death of GBM cells. In the SB28 GBM model, local delivery of HA-MSA2 induced a delay in tumor growth and a significant extension of survival. The analysis of the TIME showed a profound shift in the GBM immune landscape after HA-MSA2 treatment, with higher infiltration by innate and adaptive immune cells including dendritic, natural killer (NK) and CD8 T cell populations. The therapeutic potential of this novel polymer conjugate warrants further investigation, particularly with other chemo-immunotherapeutics or cancer vaccines as a promising combinatorial therapeutic approach.
... 35,36 International Journal of Bioprinting Hyaluronic acid is an acidic mucopolysaccharide initially extracted from the vitreous body of bovine eyes. 37 Owing to its distinctive molecular structure and unique physical and chemical attributes, hyaluronic acid serves various pivotal physiological roles within the body. For instance, it functions in joint lubrication, the regulation of blood vessel wall permeability, and the modulation of proteins. ...
Article
Full-text available
Three-dimensional (3D) bioprinting technologies play significant roles in various facets of the medical field, such as bioengineering, tissue repair, scaffolds, biomedical devices, and drug. As a versatile manufacturing technology, 3D bioprinting is able to overcome the constraints of other conventional methods and shows potential for future advancements in the field of biology. Nevertheless, the existing 3D bioprinting technologies still grapple with significant challenges in materials, equipment, and applications. Therefore, it is essential to select appropriate bioprinting method in alignment with the required application. In this review, we aim to cover the development, classification, and application of 3D bioprinting, with a particular emphasis on the fundamental printing principles. Additionally, we discuss the potential of 3D bioprinting in terms of materialization, structuralization, and functionalization, highlighting its prospective applications. We firmly believe that 3D printing technology will witness widespread adoption in the future, as it has the potential to address the limitations associated with multi-size, multi-material, multi-cell, and high-precision bioprinting.
Article
Full-text available
In this work, a sequential covalent immobilization of graphene oxide (GO) and hyaluronic acid (HA) is performed to obtain a biocompatible wear‐resistant nanocoating on the surface of the biomedical grade cobalt‐chrome (CoCr) alloy. Nanocoated CoCr surfaces were characterized by Raman spectroscopy and electrochemical impedance spectroscopy (EIS) in 3 g/L HA electrolyte. Tribocorrosion tests of the nanocoated CoCr surfaces were carried out in a pin on flat tribometer. The biological response of covalently HA/GO biofunctionalized CoCr surfaces with and without wear‐corrosion processes was studied through the analysis of the proteome of macrophages. Raman spectra revealed characteristic bands of GO and HA on the functionalized CoCr surfaces. The electrochemical response by EIS showed a stable and protective behavior over 23 days in the simulated biological environment. HA/GO covalently immobilized on CoCr alloy is able to protect the surface and reduce the wear volume released under tribocorrosion tests. Unsupervised classification analysis of the macrophage proteome via hierarchical clustering and principal component analysis (PCA) revealed that the covalent functionalization on CoCr enhances the macrophage biocompatibility in vitro. On the other hand, disruption of the HA/GO nanocoating by tribocorrosion processes induced a macrophage proteome which was differently clustered and was distantly located in the PCA space. In addition, tribocorrosion induced an increase in the percentage of upregulated and downregulated proteins in the macrophage proteome, revealing that disruption of the covalent nanocoating impacts the macrophage proteome. Although macrophage inflammation induced by tribocorrosion of HA/GO/CoCr surfaces is observed, it is ameliorated by the covalently grafting of HA, which provides immunomodulation by eliciting downregulations in characteristic pro‐inflammatory signaling involved in inflammation and aseptic loosening of CoCr joint arthroplasties. Covalent HA/GO nanocoating on CoCr provides potential applications for in vivo joint prostheses led a reduced metal‐induced inflammation and degradation by wear‐corrosion.
Article
Endotracheal intubation holds significant importance in trauma point care and emergency medicine. However, laryngeal injury and subsequent chronic complications can result from the friction between the mucosa and the tube during intubation. Here we present a lubricant coating based on hyaluronic acid. Inspired by mussels, hyaluronic acid was first conjugated with dopamine to obtain HD, and then a triblock copolymer Pluronic F127 (F127) was complexed with HD to form a micelle layer. Dopamine can assist coating deposition on the tube under weak alkaline conditions, while improving the coating stability. The micelle layer formed by F127 and HD provides excellent water retention and lubrication performance. The friction coefficient of the HD-F127 coating was 56% lower than that of the control, as determined by a friction performance test.
Article
Full-text available
Three-dimensional (3D) printing has gained popularity in tissue engineering and in the field of cartilage regeneration. This is due to its potential to generate scaffolds with spatial variation of cell distribution or mechanical properties, built with a variety of materials that can mimic complex tissue architecture. In the present study, horse articular chondrocytes were cultured for 2 and 4 weeks in 3D-printed chitosan (CH)-based scaffolds prepared with or without hyaluronic acid and in the presence of fetal bovine serum (FBS) or platelet lysate (PL). These 3D culture systems were analyzed in terms of their capability to maintain chondrocyte differentiation in vitro. This was achieved by evaluating cell morphology, immunohistochemistry (IHC), gene expression of relevant cartilage markers (collagen type II, aggrecan, and Sox9), and specific markers of dedifferentiated phenotype (collagen type I, Runx2). The morphological, histochemical, immunohistochemical, and molecular results demonstrated that the 3D CH scaffold is sufficiently porous to be colonized by primary chondrocytes. Thereby, it provides an optimal environment for the colonization and synthetic activity of chondrocytes during a long culture period where a higher rate of dedifferentiation can be generally observed. Enrichment with hyaluronic acid provides an optimal microenvironment for a more stable maintenance of the chondrocyte phenotype. The use of 3D CH scaffolds causes a further increase in the gene expression of most relevant ECM components when PL is added as a substitute for FBS in the medium. This indicates that the latter system enables a better maintenance of the chondrocyte phenotype, thereby highlighting a fair balance between proliferation and differentiation.
Article
Full-text available
Hyaluronan (HA) is a naturally occurring non-sulfated glycosaminoglycan (GAG), cell-surface-associated biopolymer and is the key component of tissue extracellular matrix (ECM). Along with remarkable physicochemical properties, HA also has multifaceted biological effects that include but not limited to ECM organization, immunomodulation, and various cellular processes. Environmental cues such as tissue injury, infection or cancer change downstream signaling functionalities of HA. Unlike native HA, the fragments of HA have diversified effects on inflammation, cancer, fibrosis, angiogenesis and autoimmune response. In this review, we aim to discuss HA as a therapeutic delivery system development process, source, biophysical-chemical properties, and associated biological pathways (especially via cell surface receptors) of native and fragmented HA. We also tried to address an overview of the potential role of HA (native HA vs fragments) in the modulation of inflammation, immune response and various cancer targeting delivery applications. This review will also highlight the HA based therapeutic systems, medical devices and future perspectives of various biomedical applications were discussed in detail.
Article
Full-text available
Hollow nerve guidance conduits are approved for clinical use for defect lengths of up to 3 cm. This is because also in pre-clinical evaluation they are less effective in the support of nerve re-generation over critical defect lengths. Hydrogel luminal fillers are thought to improve the re-generation outcome by providing an optimized matrix inside the bioartificial nerve grafts. We evaluated here a modified hyaluronic acid-laminin-hydrogel (M-HAL) as luminal filler for two clinically approved hollow nerve guides. Collagen-based and chitosan-based nerve guides were filled with M-HAL in two different concentrations and the regeneration outcome comprehen-sively studied in the acute repair rat sciatic nerve 15 mm critical defect size model. Autologous nerve graft (ANG) repair served as gold-standard control. At 120 days post-surgery, all ANG rats demonstrated electrodiagnostically detectable motor recovery. Both concentrations of the hy-drogel luminal filler did induce improved regeneration outcome over empty nerve guides. But neither combination with collagen- or chitosan-based nerve guides resulted in functional recov-ery comparable to the ANG repair. In contrast to our previous studies, we demonstrate here that M-HAL slightly improved the overall performance of either empty nerve guide type in the crit-ical defect size model.
Article
Full-text available
Skin cancer is one of the most common types of cancer in the United States and worldwide. Topical products are effective for treating cancerous skin lesions when surgery is not feasible. However, current topical products induce severe irritation, light-sensitivity, burning, scaling, and inflammation. Using hyaluronic acid (HA), we engineered clinically translatable polymer-drug conjugates of doxorubicin and camptothecin termed, DOxorubicin and Camptothecin Tailored at Optimal Ratios (DOCTOR) for topical treatment of skin cancers. When compared to the clinical standard, Efudex, DOCTOR exhibited high cancer-cell killing specificity with superior safety to healthy skin cells. In vivo studies confirmed its efficacy in treating cancerous lesions without irritation or systemic absorption. When tested on patient-derived primary cells and live-skin explants, DOCTOR killed the cancer with a selectivity as high as 21-fold over healthy skin tissue from the same donor. Collectively, DOCTOR provides a safe and potent option for treating skin cancer in the clinic.
Article
Full-text available
Modulation of innate immune responses is a potential strategy for treating inflammatory diseases. Although biogenic polyamines are known as immunomodulatory compounds possessing anti‐inflammatory functions, their biomedical applications have been limited owing to their cytotoxicity. Here, the discovery of an anti‐inflammatory and biocompatible compound is reported, which is a relatively low‐molecular‐weight polyamine, branched oligoethyleneimine (bOEI). Among bOEI molecules, bOEI‐300 strongly suppresses the secretion of pro‐inflammatory cytokines from stimulated primary macrophages by scavenging reactive oxygen species and inhibiting the nuclear translocation of the nuclear factor kappa‐B. Moreover, its polyampholyte‐like conjugate with hyaluronic acid improves the biocompatibility of polyamines and enhanced anti‐inflammatory functions. In a murine ulcerative colitis model, the conjugates enhance therapeutic efficacy by suppressing the production of pro‐inflammatory cytokines. This polyamine‐conjugated biopolymer has enormous potential to treat inflammatory diseases via the modulation of inflammatory responses. An anti‐inflammatory oligoethyleneimine‐conjugated biopolymer for the treatment of ulcerative colitis is demonstrated. This conjugate possesses enhanced biocompatibility and anti‐inflammatory properties through scavenging of radical oxygen species and inhibition of the nuclear translocation of nuclear factor kappa‐B in macrophages. In a murine model of ulcerative colitis, this conjugate enhances therapeutic efficacy through the suppression of pro‐inflammatory cytokine production.
Article
Full-text available
Background Although promising, clinical translation of human mesenchymal stem or stromal cell-derived extracellular vesicles (MSC EV) for acute lung injury is potentially limited by significant production costs. The current study was performed to determine whether pretreatment of MSC EV with high molecular weight hyaluronic acid (HMW HA) would increase the therapeutic potency of MSC EV in severe bacterial pneumonia. Methods In vitro experiments were performed to determine the binding affinity of HMW HA to MSC EV and its uptake by human monocytes, and whether HMW HA primed MSC EV would increase bacterial phagocytosis by the monocytes. In addition, the role of CD44 receptor on MSC EV in the therapeutic effects of HMW HA primed MSC EV were investigated. In Pseudomonas aeruginosa (PA) pneumonia in mice, MSC EV primed with or without HMW HA were instilled intravenously 4 h after injury. After 24 h, the bronchoalveolar lavage fluid, blood, and lungs were analyzed for levels of bacteria, inflammation, MSC EV trafficking, and lung pathology. Results MSC EV bound preferentially to HMW HA at a molecular weight of 1.0 MDa compared with HA with a molecular weight of 40 KDa or 1.5 MDa. HMW HA primed MSC EV further increased MSC EV uptake and bacterial phagocytosis by monocytes compared to treatment with MSC EV alone. In PA pneumonia in mice, instillation of HMW HA primed MSC EV further reduced inflammation and decreased the bacterial load by enhancing the trafficking of MSC EV to the injured alveolus. CD44 siRNA pretreatment of MSC EV prior to incubation with HMW HA eliminated its trafficking to the alveolus and therapeutic effects. Conclusions HMW HA primed MSC EV significantly increased the potency of MSC EV in PA pneumonia in part by enhancing the trafficking of MSC EV to the sites of inflammation via the CD44 receptor on MSC EV which was associated with increased antimicrobial activity.
Article
Full-text available
In-silico anti-viral activity of Hydroxychloroquine (HCQ) and its Hyaluronic Acid-derivative (HA-HCQ) towards different SARS-CoV-2 protein molecular targets were studied. Four different SARS-CoV-2 proteins molecular target i.e., three different main proteases and one helicase were chosen for In-silico anti-viral analysis. The HA-HCQ conjugates exhibited superior binding affinity and interactions with all the screened SAR-CoV-2 molecular target proteins with the exception of a few targets. The study also revealed that the HA-HCQ conjugate has multiple advantages of efficient drug delivery to its CD44 variant isoform receptors of the lower respiratory tract, highest interactive binding affinity with SARS-CoV-2 protein target. Moreover, the HA-HCQ drug conjugate possesses added advantages of good biodegradability, biocompatibility, non-toxicity and non-immunogenicity. The prominent binding ability of HA-HCQ conjugate towards Mpro (PDB ID 5R82) and Helicase (PDB ID 6ZSL) target protein as compared with HCQ alone was proven through MD simulation analysis. In conclusion, our study suggested that further in-vitro and in-vivo examination of HA-HCQ drug conjugate will be useful to establish a promising early stage antiviral drug for the novel treatment of COVID-19.
Article
Inorganic nanomaterials that have inherently exceptional physicochemical properties (e.g., catalytic, optical, thermal, electrical, or magnetic performance) that can provide desirable functionality (e.g., drug delivery, diagnostics, imaging, or therapy) have considerable potential for application in the field of biomedicine. However, toxicity can be caused by the long-term, non-specific accumulation of these inorganic nanomaterials in healthy tissues, preventing their large-scale clinical utilization. Over the past several decades, the emergence of biodegradable and clearable inorganic nanomaterials has offered the potential to prevent such long-term toxicity. In addition, a comprehensive understanding of the design of such nanomaterials and their metabolic pathways within the body is essential for enabling the expansion of theranostic applications for various diseases and advancing clinical trials. Thus, it is of critical importance to develop biodegradable and clearable inorganic nanomaterials for biomedical applications. This review systematically summarizes the recent progress of biodegradable and clearable inorganic nanomaterials, particularly for application in cancer theranostics and other disease therapies. The future prospects and opportunities in this rapidly growing biomedical field are also discussed. We believe that this timely and comprehensive review will stimulate and guide additional in-depth studies in the area of inorganic nanomedicine, as rapid in vivo clearance and degradation is likely to be a prerequisite for the future clinical translation of inorganic nanomaterials with unique properties and functionality.
Article
To achieve target delivery of anti-tumor drugs with great biocompatibility into tumor tissues, a stimuli-responsive dendronized hyaluronic acid (HA)-docetaxel conjugate (HA-DTX-Dendron, HADD) was designed and prepared. The incorporation of HA in HADD improved the delivery of DTX to tumor cells with rich CD44 receptors. Enhanced biocompatibility and therapeutic outcomes were achieved using glyodendrons-modified HA and tumor microenvironment-responsive linkers in HADD. The glycodendron was connected with HA via GSH-responsive disulfide bonds, and the drug DTX was linked to the carrier via a cathepsin B-responsive tetrapeptide GFLG. This design resulted in self-assembly nanostructures for facilitating uptake of HADD by tumor cells and rapid release of DTX to exert its therapeutic effect. Compared to free DTX, HADD showed much higher tumor growth inhibition in the MDA-MB-231 tumor-bearing mice model (up to 99.71%), and no toxicity was observed. Therefore, HADD could be employed as an efficacious nano-agent for treating triple negative breast cancer (TNBC).
Article
Hyaluronic acid has become an interesting and important polymer as an excipient for pharmaceutical products due to its beneficial properties, like solubility, biocompatibility and biodegradation. To improve the properties of hyaluronic acid, different possibilities for chemical modifications are presented, and the opportunities as novel systems for drug delivery are discussed. This review gives an overview over the production of hyaluronic acid, the possibilities of its chemical modification and the current state of in vitro and in vivo research. Furthermore, market approved and commercially available products are reviewed and derivatives undergoing clinical trials and applying for market approval are shown. In particular, hyaluronic acid has been studied for different administrations in rheumatology, ophthalmology, local anesthetics, cancer treatment and bioengineering of tissues. The present work concludes with perspectives for future administration of pharmaceuticals based on hyaluronic acid.