Hyaluronan-Inorganic Nanohybrid Materials for Biomedical Applications

May 9, 2017 - have improved the features and properties of nanomaterials. However, ... of nanomaterials whose size, geometry, and surface functionalit...
0 downloads 0 Views 2MB Size
Subscriber access provided by University of Virginia Libraries & VIVA (Virtual Library of Virginia)

Review

Hyaluronan-inorganic nanohybrid materials for biomedical applications Zhixiang Cai, Hongbin Zhang, Yue Wei, and Fengsong Cong Biomacromolecules, Just Accepted Manuscript • Publication Date (Web): 09 May 2017 Downloaded from http://pubs.acs.org on May 10, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Biomacromolecules is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

Hyaluronan-inorganic nanohybrid materials for biomedical applications Zhixiang Cai†, Hongbin Zhang†*, Yue Wei†, Fengsong Cong†† †Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China ††Department of Biochemistry and Molecular Biology, School of life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China KEYWORDS: glycosaminoglycan; nanomaterials; functionality; biomedical applications; polysaccharide.

ABSTRACT: Nanomaterials, including gold, silver and magnetic nanoparticles, carbon and mesoporous materials, possess unique physiochemical and biological properties thus offering promising applications in biomedicine, such as in drug delivery, biosensing, molecular imaging and therapy. Recent advances in nanotechnology have improved the features and properties of nanomaterials. However, these nanomaterials are potentially cytotoxic and demonstrate lack of cell-specific function. Thus, they have been functionalized with various polymers, especially polysaccharides, to reduce toxicity, improve biocompatibility and stability under physiological conditions. Particularly, nanomaterials have been widely functionalized with hyaluronan (HA) to enhance their distribution in specific cells and tissues. This review highlights the most recent advances on HA-functionalized nanomaterials for biotechnological and biomedical applications,

ACS Paragon Plus Environment

1

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 72

as nanocarriers in drug delivery, contrast agents in molecular imaging and diagnostic agents in cancer therapy. A critical evaluation of barriers affecting the use of HA-functionalized nanomaterials is also discussed and insights into the outlook of the field are explored.

1. Introduction Nanotechnology is a multidisciplinary field involving fabrication and utilization of materials, devices or systems at the nanoscale.1 Advances in nanotechnology have led to the development of nanomaterials, whose size, geometry and surface functionality can be controlled at the nanoscale.2 The development of nanotechnology-based materials has boomed in the past few decades, because nanomaterials are unique as their sizes and physical properties are chemically tunable. In addition, they are expected to be useful in innovations and they play a critical role in various biomedical applications.3-5 However, unfunctionalized nanomaterials are potentially cytotoxic and lack cell-specific function.6 Thus, fabrication of nanomaterials functionalized with biological molecules and carbohydrates and their applications sharply increased in recent years.79

Hyaluronan or hyaluronic acid (HA) is a naturally occurring linear polysaccharide consisting of a repeating units of D-glucuronic acid and N-acetyl glycosamine alternately linked by β-(1,4) and β-(1,3) glycosidic bonds; as one of the most important and ubiquitous glycosaminoglycans, HA is distributed widely in the human body, such as in vitreous of the eye and in the extracellular matrix of cartilage tissues.10 The function of HA in vivo is to maintain moisture, adjust osmotic pressure, lubricate joints and absorb shock, all of which are close related to its physiochemical and rheological properties. In a series of work by our group, the physiochemical and rheological properties of HA have been investigated.11-13 In addition, HA bears functional

ACS Paragon Plus Environment

2

Page 3 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

groups, including hydroxyl carboxylic groups and N-acetyl group, which can be utilized for further chemical modifications.14 At the molecular level, HA interact with cell surface receptors, such as CD44, RHAMM, and LYVE-1 receptors, collectively known as hyaladherins, which are overexpressed in many cancer cells. Thus, HA is used as diagnostic indicator of cancerous angiogenesis and progression of various tumour types.15, physiochemical

properties,

such

as

16

biodegradability,

Moreover, HA exhibits excellent cytocompatibility,

nontoxicity,

nonimmunogenicity and high water-binding capacity.17 The physiochemical properties and multifaceted biological functions of HA have attracted a considerable attention for the development of HA-based biomaterials for various biomedical applications,18 such as drug delivery,19, 20 targeted diagnosis,21 tissue engineering,22 and molecular imaging23. Successful advances in nanotechnology have contributed to the progress in the development of HA-functionalized nanomaterials (HA-nanomaterials) that are used in biological applications. Moreover, the intriguing biological properties of HA render HA a potential targeting ligands in nanoparticle modification. Various nanomaterials, such as gold nanoparticles (AuNPs) and silver nanoparticles (AgNPs),24, 25 carbon materials (graphene, carbon nanotubes (CNTs) and quantum dots (QDs)),26, 27 magnetic iron oxides21 and mesoporous materials,28 have been functionalized with HA. HA-based nanomaterials are multifunctional nanomaterials that combine the physical and chemical properties of nanomaterials with the biological characteristics of HA. This review focuses on the recent progress in the development of HA-nanomaterials and their applications in biomedicine. HA-nanomaterials are likely to open new opportunities for rapid expansion of the biomedical applications of nanomaterials. 2. HA-functionalized nanomaterials (HA-nanomaterials)

ACS Paragon Plus Environment

3

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 72

2.1 HA-functionalized Au nanocomposites Au nanocomposites displayed biological inertness and possess distinct physical and chemical attributes, including controlled geometrical, optical, and surface chemical properties. Au nanocomposites are useful in a variety of biomedical applications because the surfaces of Au nanocomposites can be modified, allowing their attachment to a ligand, drug, or other target biomolecules. These features of Au nanocomposites have recently led to new and exciting developments that present enormous potential applications in biology and medicine.29 This development was first fully realized in a range of medical diagnostic and therapeutic applications.30 In this section, we will briefly describe the synthesis of HA-functionalized Au nanocomposites and highlight their applications in biomedical fields. Considerable efforts have been devoted to developing AuNPs with monodispersity and controlled size. Nanoparticles are defined as particulate dispersions or solid particles with a size generally in the range of 1 to 100 nm.31,

32

The development of non-toxic and eco-friendly

processes of AuNPs synthesis has become a challenging issue for many researchers. Nowadays, researchers were inspired to integrate “green chemistry” approaches to fabricate AuNPs using biopolymers.33 In particular, AuNPs were prepared by reducing AuCl4 using HA as both the reducing and stabilizing agent.34 AuNPs are considerably more stable than naked AuNPs under physiological conditions because HA bound to nanoparticles causes electrostatic repulsion. Apart from their unique physical attributes, AuNPs can be potentially applied in biomedical field given the excellent physical properties (optical and electrical) of AuNPs and the biological activities of HA. Hien and co-workers have developed a method to synthesise HA-capped AuNPs by using γirradiation method.35 Their results showed that the AuNPs exhibited narrow size distribution under high dose rate and HA concentrations, whereas they displayed wide size distribution under

ACS Paragon Plus Environment

4

Page 5 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

high Au3+ concentrations. The HA-capped AuNPs smaller than 10 nm can be potentially applied in biomedicines and cosmetics. Li and co-workers19 have recently developed a HA-Au supramolecular conjugate (HACDAuNPs) by using AuNPs bearing adamantane moieties and cyclodextrin-grafted HA (Figure 1). This supramolecular conjugate can be stably constructed because of the high affinity between βcyclodextrin (β-CD) cavity and adamantane moieties. The supramolecular conjugate was subsequently explored as an efficient targeted delivery system for various anticancer drugs, such as doxorubicin hydrochloride (DOX), paclitaxel, camptothecin (CPT), irinotecan hydrochloride (CPT-11) and topotecan hydrochloride (TPT). Taking the anticancer drug DOX as example, in vitro studies have shown that the DOX-loaded HACD-AuNPs delivery system exhibited high cellular uptake and anticancer activities that are comparable to those of free DOX but with low side effects owing to CD44 receptor-mediated endocytosis. Furthermore, the drug-loaded delivery system exhibited pH-responsive release under mildly acidic environment, such as the internal environment of a cancer cell. In a word, this smart HACD-AuNP supramolecular conjugates provide a versatile platform for targeted drug delivery characterized by high activity and low toxicity and present promising application for clinical treatment of cancer. In this study, the average diameter of HACD-AuNP was approximately 258nm with a narrow size distribution. However, the most efficient cellular uptake was observed with particles ranging from 20 to 50nm via the enhanced permeation and retention (EPR) effect.36 Therefore, the HACD-AuNP arrived at the diseased tissues through HA specific recognition by cell surface receptors (CD44).

ACS Paragon Plus Environment

5

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 72

Figure 1. Schematic of the chemical structures and the construction of HACD-AuNPs and drugloaded HACD-AuNPs: the drug delivery system containing CD-modified HA (HACD), AuNPs bearing adamantane moieties, and an anticancer drug. Reprinted by permission from Macmillan Publishers Ltd: Scientific Reports, ref 19, Copyright 2014. http://www.nature.com/srep/. In another study, AuNPs have been actively used as a delivery carrier for various biopharmaceuticals.37 In addition to its application in the delivery of anticancer drugs, AuNPs have been proposed as carriers in target-specific systemic treatment of hepatitis C virus (HCV) infection. Lee and co-workers developed a target-specific long-acting delivery system (HAAuNPs/IFNα complex) based on interferon α (IFNα) loaded on thiolated HA-modified AuNPs (HA-AuNPs). The HA-AuNPs/IFNα complex has some advantages such as enhanced serum stability in human serum and target-specific delivery capacity in liver tissue. Overall, HAAuNPs/IFNα complex is a potential new nanomedicine that demonstrates an enhanced and prolonged efficacy for the treatment of chronic HCV infection, and this finding provides new insights into the development of drug release systems to treat HCV infection.

ACS Paragon Plus Environment

6

Page 7 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

AuNPs have been widely used in various visualization and bioimaging techniques to identify chemical and biological agents.38 Reactive oxygen species (ROS) are oxygen-containing molecules bearing an unpaired electron that are highly reactive in redox reactions. Studies have shown that ROS serve as signalling molecules regulating numerous cellular process, including proliferation. It is known that ROS generated beyond the limit of the natural antioxidant defence systems are considered toxic and can damage cellular macromolecules, resulting in cell death.39 To evaluate ROS toxicity, Lee and co-workers have demonstrated the use of novel and biocompatible ROS-sensitive AuNPs to detect the level of intracellular ROS induced by various polystyrene (PS) nanoparticles with different sizes and functional groups.40 The ROS-sensitive AuNPs (HF-AuNPs) were successfully prepared by using fluorescent dye-labelled HA grafted onto the surface of AuNPs. The HF-AuNPs possessed enhanced detection sensitivity for intracellular ROS than other commercialized ROS fluorescent probes. The results revealed that smaller and more positively charged PS nanoparticles highly induced intracellular ROS generation, confirming the high cellular toxicity of these particles. Furthermore, Hyun et al.41 have designed ROS-sensitive Au nanoprobes (HHAuNPs) by using near-infrared (NIR) fluorescence dye-labelled HA and AuNPs for ischemic brain imaging. The HHAuNPs nanoparticles exhibited high stability in a wide range of pH, salt concentrations and media, and possessed a strong fluorescence signal. This study demonstrated that the HHAuNPs is a powerful tool to monitor ROS level and identify the infarct areas in ischemic brain for stroke treatment. In addition to the use of colloidal Au containing spherical particles, the use of nonspherical cylindrical particles (nanorods), nanoshells, nanocages and nanostars has been studied, and these particles are widely applied in current medical and biological research. For example, HA-capped Au nanocages (AuNCs-HA) were used to design a multi-stimuli responsive platform for

ACS Paragon Plus Environment

7

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 72

targeted, noninvasive and pinpointed intracellular DOX release (Figure 2).42 Drug-loaded nanohybrids were taken up efficiently by cancer cells via HA-CD44 interactions and subsequently degraded intracellularly into small fragments, facilitating DOX release. In vivo experiments have further revealed that NIR irradiation enhances the release of encapsulated drug and improves its therapeutic efficacy because of the excellent photothermal properties of the drug. In particular, one of the major feature of AuNCs-HA is the combination of chemotherapy and photothermal therapy that can result in an excellent synergistic effect. Thus, AuNCs-HA nanohybrids demonstrated biocompatibility, CD44-targetability, multi-stimuli responsiveness, pinpointed drug release and chemo-photothermal synergistic effects present a great potential application in cancer therapy.

Figure 2. Schematic of a multi-stimuli responsive platform based on DOX-loaded AuNCs-HA nanoparticles for pinpointed intracellular drug release and synergistic therapy: pH and NIRstimuli responsive targeted drug delivery system was activated to trigger the release of encapsulated drug after nanoparticles were internalized. Reprinted from ref 42, Copyright 2014, with permission from Elsevier.

ACS Paragon Plus Environment

8

Page 9 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

Au nanostructures have been intensively investigated because of their fascinating surface plasmon resonance (SPR) properties. The SPR properties are strongly dependent on the size, shape, and surface functionality of the Au nanostructures. NIR-absorbing Au nanorods functionalized with a biopolymeric shell and embedded in HA were recently prepared and used in carotid artery closure in vivo.43 Given that the hybrid material was rendered stable by HA, it can be easily stored because minimal optical and structural modifications occur over time. Overall, this work can open a new field in nanotechnology-based therapies employed in minimally invasive procedures in clinical practice. In addition, Liu and co-workers24 have reported on HA grafted onto Au surface to fabricate HA-Au sensor chip, which demonstrates excellent antifouling performance against protein adsorption, good stability, and compatibility. SPR spectroscopy, which is a powerful tool used to monitor molecular interaction, was used in this study to investigate nonspecific adsorption onto materials ranging from single protein solutions and complex media to HA-Au biosensor chips. The results showed that the developed HA-Au could be used to fabricate antifouling surfaces and SPR biosensor chips for sensitive detection of bovine serum albumin (BSA) and may be used in minimally invasive clinical practices. The HA-AuNPs nanomaterials for biomedical applications were summarized in Table 1. As mentioned-above, HA-functionalized AuNPs have been successfully applied in biomedical field as promising candidates for drug delivery systems and platforms for the detection of biological molecules. The motivations for AuNPs functionalized by HA are the enhanced targeting and delivery of drug to target cells and the reduced toxicity of AuNPs. However, despite the reports that HA-AuNPs used in many medical and health-related conditions are inherently nontoxic, the efficacy, potential toxicity and health impact of HA-AuNPs are still

ACS Paragon Plus Environment

9

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 72

under some scrutiny. The challenge will be the preparation of a range of HA-AuNPs nanomaterials using a common synthetic strategy with exact surface functionality for accurate comparison. Further work is still needed to clarify their efficacy and safety when taking advantage of the biological properties of HA and the unique physiochemical properties of AuNPs to fabricate HA-AuNPs especially for biological imaging, drug delivery, and cancer treatment. 2.2 HA-functionalized Ag nanocomposites Ag nanostructures have been attracting interest because of their numerous potential applications in surface-enhanced Raman scattering (SERS),44 catalysis45 and biosensing46. AgNPs exhibit exceptional antibacterial properties and thus present potential biological and medical applications.47 In recent years, Ag-based biomedical products are increasingly utilized in bioactive materials, demonstrating their capability to effectively retard and prevent bacterial infections. Various approaches for AgNP fabrication were developed in the past decades. However, with increasing awareness on environmental protection, carbohydrate polymers have been employed as reducing and stabilizing agents in AgNP synthesis to avoid the use of the conventional noxious reducing and stabilizing agents. HA has been employed as template to direct AgNP synthesis. Some typical methods for AgNP synthesis and recent applications of AgNPs are introduced. For instance, Cui and co-workers, have utilized HA to prepare different Ag nanostructures for SERS application.48 These studies have shown that the shape of Ag nanostructrues could be easily controlled by the storage time of HA and AgNO3 solution before being mixed for photoreduction. Application of Ag nanoplate has improved SERS efficiency

ACS Paragon Plus Environment

10

Page 11 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

compared with the application of Ag spherical NPs because SERS efficiency greatly depends on particle shape and irradiation time. By contrast, Xia and co-workers49 have utilized HA to prepare Ag nanostructures that present potential applications in SERS and biosensing. In addition, Kemp et al.34, 50 have studied the antibacterial, anticoagulant and anti-inflammatory efficacies of AgNPs stabilized by HA. The results demonstrated that these nanoparticles exhibit unique antibacterial property and high anticoagulant and anti-inflammatory efficiencies, which are useful in various biological and biomedical applications. The number of HA-based nanocomposites has increased in the past decade. In addition to prepare AgNPs by using HA, incorporation of AgNPs into HA has recently become a research hotspot because of the outstanding physical, chemical and biological properties of HA and AgNPs. For example, Chen and co-workers51 took advantage of these properties to synthesize core-sheath electrospun HA/polycaprolactone nanofibrous membranes embedded with AgNPs, this complex is used to prevent peritendinous adhesion. The HA and AgNPs in this nanofibrous membrane were used for effective lubrication and antibacterial activity, respectively. The in vitro and in vivo experiments further confirmed that this nanofibrous membrane reduces peritendinous adhesion and proliferation without exerting significant cytotoxicity. In addition, Cui and coworkers52

developed

AgNPs

embedded

in

a

layer-by-layer

assembled

HA/poly(dimethyldiallylammonium chloride) (PDDA) structure. This nanomaterial has good stability for localized SPR biosensor and excellent antibacterial capability, indicating that the film offers practical potential application as a biosensing and antimicrobial material. Encouraged by this result, Zhang et al.53 constructed an HA-AgNP-hemoglobin multilayer composite film with good biocompatibility, antibacterial, and stability properties and subsequently investigated its electrocatalytic properties. The results showed that this film presents a great potential use in

ACS Paragon Plus Environment

11

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 72

biosensing. Zhang and co-workers25 subsequently described a complex hydrogel formed from HA and PVA and embedded with AgNPs, this hydrogel displayed a high antibacterial property and can be potentially used as wound dressing material. Moreover, studies have reported on nanocomposites based on HA and AgNPs, and these nanocomposites demonstrate good biomedical activity.47,

54-56

The majority of these studies have focused on the antibacterial

property of HA-Ag nanocomposites. Earlier studies of biological applications for AgNPs focused mainly on antibacterial issue. Recently, AgNPs have been reported for their potent antitumor activities owing to a possible mechanism of intracellular induction of ROS and resulting in DNA damage.57 However, the nonspecific delivery and poor cellular uptake have significantly limited AgNPs used in nanomedicine field. Moreover, they also have poor stability and relatively high cytotoxicity. In order to overcome these limitations, coated or functionalized AgNPs with biopolymers is required.58 In a work done by Liang and co-workers,59 AgNPs were synthesized by using HA, which acts as a reducing agent and stabilizer, as well as a targeting ligand. The antitumor efficacy of HA-AgNPs was significantly enhanced, in comparison with the non-modified AgNPs, which can be attributed to the CD44-mediated endocytosis. The work was possibly the first to report the targeted antitumor efficacy based on AgNPs. This idea was further developed by Zhang et al.60 Due to the novel physiochemical and optical properties of AgNPs, HA-AgNPs was first developed as a nanoplatform for X-ray computed tomography (CT) and single-photon emission computed tomography (SPECT) imaging. Furthermore, the obtained HA-AgNPs were spherical, ultrasmall and monodisperse, and demonstrated excellent long-term stability and low cytotoxicity. Clearly, this study exhibited greater potential for in vivo applications (Figure 3). As

ACS Paragon Plus Environment

12

Page 13 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

a matter of course, further evidence is needed to adequately assess the long-term toxicity of HAAgNPs exposure on human physiology.

Figure 3. Schematic of HA-Coated AgNPs as a nanoplatform for in vivo imaging applications: the HA-AgNPs were used as a nanoplatform for X-ray CT and SPECT imaging after being radiolabeled with 99mTc. Reprinted from ref 60. Copyright 2016 American Chemical Society. The HA-AgNPs nanomaterials for biomedical applications were also summarized in Table 1. In conclusion, AgNPs are increasingly utilized in HA-based biomatials, proven to be very effective in retarding and preventing bacterial infections. However, there is still a large amount of development required in drug delivery system and in vivo bioimaing applications. In most cases, as we have already stated, although these studies have demonstrated that the HA/AgNPs have low-toxicity, numerous in vitro studies of AgNPs have showed long-term toxic effects to cells exposure on human physiology. Thus, the health impact of AgNPs will promote researchers to

carefully

design

AgNPs-based

ACS Paragon Plus Environment

nanomaterials.

13

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Page 14 of 72

Table 1 The major nanomaterials of HA-AuNPs and HA-AgNPs for biomedical applications. Systems HA-capped AuNPs HA-AuNP/interferon α complex

Mw of HA

Nanomaterials

Nanomaterials

(Da)

dimensions (nm)

shapes

Not given

4-10

12k

29.25

Properties

Applications

Nanoshell

None

Monodispersity

Biomedicines and cosmetics

None

35

Nanoshell

Interferon α

Treatment of HCV infection

None

37

None

41, 61

Fluorescein-labeled

High stability and target-specific High stability and

Nanoprobes for reactive oxygen

strong fluorescence

species detection and treatment

AuNPs (HHAuNPs)

signal

of stoke

Drug loaded gold

Combination of

HA immobilized on

nanocages @ HA

17k

100k

~20

~50

Nanoshell

None

Nanocages

DOX

(AuNCs-HA)

HA onto Au substrate

350k

10.5

Au substrate

None

HA stabilized iodinecontaining nanoparticles with Au

35k

105

Photothermal

Nanoshell

ablation

nanoshell

dendrimer-entrapped

238-248

Nanoshell

None

the surface of AuNPs

therapy Resist nonspecific adsorption from complex media in SPR

performance

biosensors

Excellent dispersibility and stability; multifunctional

X-ray CT imaging and photothermal therapy of tumors

Reference

MDA-MB-231 and NIH-3T3

42

cells

None

24

MCF-7

62

ray attenuation

CT/magnetic resonance (MR) dual-mode imaging

Hepatocellular carcinoma

63

(HCCLM3 cells)

r1 relaxivity

46k

258

Supramolecular conjugates

AuNPs) HA immobilized on

systems for synergistic cancer

antifouling

intensity and favorable

CD modified HA-

conjugates (HACD-

intracellular drug release

High stability, and

dispersibility; high X6k

AuNPs

AuNP supramolecular

Multi-stimuli responsive

models

Good stability and

HA-modifed manganese-chelated

chemotherapy and photothermal therapy

1000k,

Tumor or cell

Drugs

1k-4k

55.9±3.1

Nanoshell

DOX, PTX, CPT, CPT-11, and TPT

None

High cellular uptake and anticancer activities; low side effects

Versatile platform for the targeted delivery of anticancer

19

drugs

Cellular probe;

A mediator of laser-induced

photodamage media

photothermal cell damage

ACS Paragon Plus Environment

MCF-7 cells

MDA-MB-435 S, MDA-MB-453

64

and NIH/3T3

14

Page 15 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Biomacromolecules

AuNP-HA nanoassembly

12k

150.2±3.1

Nanoparticle

Photodynamic therapy

Biostable, photo- and enzyme-activatable nanomaterial

Photothermally boosted

MDA-MB-231

photodynamic tumor ablation

cells

65

HA-gold nanorod/death receptor 5 antibody

10k

91.28

Nanorod

(HA-AuNR/DR5 Ab)

Death

Photoacoustic imaging

A novel theranostic platform for

receptor 5

and antibody cancer

noninvasive transdermal

antibody

therapy

treatment of skin cancers

HCT116 cancer

66

cells

complex

FITC–HA functionalized AuNPs

Sensitive, rapid and Not given

6.0±0.7

Nanoshell

None

HAase

HA fibers with incorporated AgNPs

High antibacterial 1750k

None

Fiber

None

(HA-Ag NPs) HA-coated AgNPs

accurate analysis of

305.5±15.1

Nanoshell

None

clinical diagnosis of HAaserelated diseases, such as bladder

None

67

cancer Antibacterial activity and cell

Mouse fibroblast

viability

cell line 3T3

High cell uptake and

AgNP-mediated cancer

MCF-7 and

anticancer activities

treatment

SW480 cells

A nanoplatform for X-ray CT

Lewis lung

and SPECT imaging

carcinoma

activity; low cytotoxicity

100k

Accurate detection of HAase for

54

59

Ultrasmall and monodisperse; HA-coated AgNPs

22k

13.5

Nanoshell

None

excellent long-term stability and low

60

cytotoxicity

ACS Paragon Plus Environment

15

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 72

2.3 HA-functionalized graphene (HA-graphene) and its derivatives Studies have recently focused on the development of graphene-based nanomaterials as new materials to be used in biomedical fields (mainly explored for their application in drug delivery, biosensing, and molecular imaging) because graphene possesses unique and extraordinary mechanical, thermal, chemical and optical properties.68-71 However, the instability of graphenebased nanomaterials under physiological conditions has hindered their wide application. Therefore, a number of strategies have been developed to improve the physiological stability of graphene-based nanomaterials, among which HA-graphene and its derivatives have greatly attracted attention. To address this problem of instability, recently we have prepared pyreneconjugated-HA (HA-Py) to facilitate the exfoliation and stabilization of laminar materials including graphene, hexagonal boron nitride, molybdenum disulfide and CNTs in water under sonication.72 Moreover, the HA-Py conjugate that stabilizes the reduced graphene oxide (rGO) to be used in fabricating composite nanomaterials involving noble metals (Au, Ag, Pd and Pt) was further investigated.73 HA-Py is not only a stabilizing agent in this system but also facilitates and controls the decoration of metals on HA-Py-rGO. The Au-HA-Py-rGO hybrid nanomaterials also exhibited a high electrochemical/catalytic activity. Therefore, the hybrid nanocomposites can be employed as sensing material for a wide range of biomedical and pharmaceutical applications. Overall, gaining a stable and biocompatible graphene remains challenging. In recent decades, investigators have found that graphene and GO are toxic to biological systems, greatly hindering their wide applications in the biomedical field.74 Therefore, surface functionalization of graphene and its derivatives is a crucial step. Given the versatile surface functionalization and ultra-high surface area of graphene and its derivatives, these materials can be easily modified and functionalized with biomolecules to obtain graphene-based nanomaterials

ACS Paragon Plus Environment

16

Page 17 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

to be used in biomedical applications.75 In general, the cellular environment in tumor tissue is more acidic than in normal tissues.76 The application of graphene and its derivatives applied in drug delivery systems has been reported in the past decades; π-π interaction weakens in an acidic environment, resulting in pH responsive drug release.77 This section presents the combination of HA with graphene-based nanomaterials for biomedical applications. Several reports have described the combination of HA with graphene-based nanomaterials for drug delivery. Miao and co-workers have recently prepared an HA-coated rGO used to construct a targeted anti-cancer drug delivery system.78 They first prepared cholesteryl-modified HA (CHA), which they used to coat rGO nanosheets to form CHA-rGO nanohybrid as nanoplatform for DOX loading. The CHA-rGO nanohybrid showed increased colloidal stability under physiological conditions and improved in vivo safety compared to rGO. DOX-loaded CHA-rGO nanohybrid displayed increased antitumor efficacy compared with free DOX or rGO/DOX because of the high distribution and prolonged retention of CHA-rGO/DOX in tumour sites. Wu and co-workers subsequently prepared HA-conjugated GO for the development of a drug delivery system (Figure 4).79 The GO-HA showed a negligible hemolytic activity and low cytotoxicity. The resulting DOX-loaded GO-HA exhibited high efficiency in targeted drug delivery in HeLa cells and can potentially inhibit tumour growth.

ACS Paragon Plus Environment

17

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 72

Figure 4. Schematic of the preparation of DOX-loaded GO-HA: (A) adipic acid dihydrazide (ADH) functionalized GO (GO-ADH); (B) HA conjugated GO-ADH (GO-HA); (C) DOX loaded onto GO-HA; (D) intravenous administration of GO-HA/DOX. Reprinted from ref 79, Copyright 2013, with permission from Elsevier. Considerable efforts have been devoted to exploring graphene-based stimuli-responsive controlled drug delivery systems (CDDSs) by taking advantage of the acidic tumour microenvironment and high intracellular GSH levels in tumours. In a work related to GO-based CDDSs, HA-decorated GO nanohybrids were used in loading anticancer drug (DOX) (HA-GODOX) to fabricate a pH-dependent drug release system.80 Owing to the aromatic structure of DOX and GO, DOX can be loaded into a GO nanostructure via π-π stacking (Figure 5). HA-GODOX exhibited superior physiological stability, and possessed high drug loading capacity and drug delivery efficiency. As a result, HA-GO-DOX demonstrated higher tumour inhibition toward H22 hepatic cancer cell than free DOX and GO-DOX under the pH of the tumor micoenvironment. Therefore, pH-responsive HA-GO-DOX is a promising agent to enhance the

ACS Paragon Plus Environment

18

Page 19 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

antitumor efficacy of conventional chemotherapy against cancer. In a follow up study, Jung et al. synthesized a targeted anti-cancer drug delivery system based on HA and GO.81 They found that the nanocarrier demonstrated not only enhanced serum stability and targeted specific anticancer drug delivery but also the ability for pH-responsive drug release. As mentioned above, the asobtained nanocarrier can serve as an effective synergistic anticancer drug delivery system.

Figure 5. Schematic of the preparation of HA-GO-DOX nanohybrid: ADH-HA as both targeting and hydrophilic moieties functionalized GO and DOX nanocomposite formed by π-π stacking. Reprinted from ref 80. Copyright 2014 American Chemical Society. Graphene-based nanomaterials have also been for photothermal treatment (PTT) of cancer under low-power NIR irradiation because of their excellent light-to-heat conversion. A considerable number of studies has developed HA-graphene-based nanomaterials as targetable and photoactivity switchable nanoplatform for PTT of cancer. Khatun and co-workers recently reported on DOX-conjugated graphene in sulphide bond-linked HA nanogel, which was used as drug carrier for targeted drug delivery that demonstrates light- and pH-responsive release.69 This nanogel showed good mono-dispersibility and stability in buffer and serum, and possessed an excellent photo-luminescence property. Their results showed that the responsiveness of the nanohybrid to pH changes in cancer microenvironment triggered DOX release and effectively

ACS Paragon Plus Environment

19

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 72

inhibited growth of the A549 cell line; thus, this nanohybrid is potentially useful in constructing stimuli-responsive drug release matrices. Jung and co-workers investigated graphene oxide-HA conjugate (GO-HA) for photothermal ablation therapy of melanoma skin cancer by using NIR laser (Figure 6).82 This kind of nanohyrid material exhibited high light-to-heat conversion efficiency and low cytotoxicity. After NIR irradiation, tumour tissues were completely ablated without recurrence of tumorigenesis. This intriguing result revealed that this system is apparently useful as a therapeutic agent for transdermal chemotherapy and PTT of melanoma skin cancers.

Figure 6. Schematic of targeted delivery of GO-HA and photothermal ablation: transdermal delivery of GO-HA into melanoma skin cancer cells and the subsequent photothermal ablation therapy using NIR irradiation. Reprinted from ref 82. Copyright 2014 American Chemical Society. Insufficient visualization of the delivery, distribution, metabolism and digestion of PTT has seriously restricted the wide application of PTT in the biomedical field. Therefore, development of different imaging-guided PTT nanoplatforms is necessary. To address this issue, researchers have exerted a considerable effort to explore imaging-guided PTT nanoplatforms that demonstrate powerful imaging and therapeutic capacities, and considerable achievements have been achieved over the past years. For example, Miao et al. have reported a photoresponsive NIR

ACS Paragon Plus Environment

20

Page 21 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

imaging agent (indocyanine green, ICG) loaded onto HA-conjugated rGO (HA-rGO) that can be utilized as image-guided synergistic antitumor PTT.83 Compared with the photostability of free ICG, that of ICG-containing HA-rGO or rGO was substantially enhanced. In this system, ICGloaded HA-rGO nanohyrbid material had high photostability and photothermal antitumor efficiency, which can be developed as a potential theranostic nanoplatform to track and monitor targeted drug, as well as a synergistic photothermal antitumor therapy. A similar finding was reported by Li et al., wherein photosensitizers (chlorin Ce6) were effectively loaded onto the surface of HA-GO that had high colloidal stability and enhanced photodynamic efficiency, which was developed as target and photoactivity switchable nanoplatform for photodynamic therapy (PDT).84 Graphene-based nanomaterials have also attracted significant interest in the area of bioimaging because they have been found to be photoluminescent in the visible and infrared regions.75, 85 Based on their intrinsic high NIR absorbance, functionalized GO and rGO have been used for live cell imaging.86, 87 In a recent study, for this purpose, branched polyethylenimine (BPEI) conjugated to GO has been designed to be used as a gene delivery vector and as a bioimaging tool because of its low cytotoxicity and high gene delivery efficiency.88 In another case, Khatun and co-workers combined HA with graphene to prepare a light- and pH-responsive drug release system, useful for the delivery of DOX and killing tumor cells.69 Graphene in the nanomaterials acted as an optical imaging contrast agent as well as a heat source when excited by laser irradiation. Although graphene derivatives have been increasingly investigated in bioimaging field, to our best knowledge, in most previous reports, much attention has been paid to utilize organic fluorescent dyes functionalized graphene derivatives for in vitro and in vivo fluorescence imaging.75

ACS Paragon Plus Environment

21

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 72

It is worth noting that all these HA-functionalized graphene and its derivatives nanomaterials are prepared by coupling via N-ethyl-N’-dimethylaminopropyl-carbodiimide (EDC) and Nhydroxysuccinimide (NHS) chemistry. Indeed, hybrid nanomaterials with various functionalities prepared via EDC/NHS chemistry have extensively been reported. This method offers some advantages for the simplicity of preparation and manipulation. HA-graphene and its derivatives for biomedical applications were summarized in Table 2. Overall, owing to the excellent physiochemical properties and biocompatibility, these nanocarriers of HA-graphene and its derivatives are greatly promising candidates as multifunctional nanoplatforms that combine both therapeutic components and multimodal imaging. These nanoplatforms can bypass many biological barriers to enhance the targeting efficacy. However, there are a few major obstacles in the biomedical application of graphene-based nanomaterials, i.e., the nonbiodegradable nature and long-term toxicity of graphene and its derivatives, even modified by HA. In addition, the in vivo behavior of HA-graphene nanomaterials with different structures, sizes, and surface properties remain unknown. At the same time, HA-graphene nanomaterials are difficult and expensive to manufacture at large scales with optimal quality. Thus, tremendous studies on HAgraphene and its derivatives are still warranted to be carried out before going to clinical trials. The future of HA-graphene and its derivatives utilized in biomedical fileds looks brighter than ever, yet many obstacles remain to be conquered. 2.4 HA-functionalized CNTs (HA-CNTs) CNTs have recently emerged as promising nanomaterials in nanomedicine, wherein they serve as drug delivery vehicles and bioimaging agents owing to their unique structure and properties, including high aspect ratio, unique optical property, high drug-loading capability and enhanced cellular uptake.89,

90

However, because of the very high long-range van der Waals forces of

ACS Paragon Plus Environment

22

Page 23 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

attraction, pristine CNTs are limited by technological barriers, such as high aggregation tendency and poor aqueous dispersibility, thereby limiting their applications in biomedicine.91 Furthermore, an issue worthy of consideration is that pristine CNTs are highly toxic when applied in in vitro and in vivo experiments.92 Thus, approaches must be developed to confer CNTs with improved water solubility and high biocompatibility and reduce their systemic toxicity. In particular, covalent functionalization of CNTs is one of the most powerful approaches to render CNTs these properties.93 HA has been used for targeted cancer therapy owing to its unique and excellent biological properties. Thus, HA-CNTs are promising tumourtargeting drug delivery agents for cancer treatment. Moreover, recent expansion in HA-CNTs, along with the identification of disease-specific molecular target and imaging capabilities, has promoted the development of multifunctional, drug-loaded CNTs.27, 94-96 This section presents the latest achievement in the development of HA-CNTs used for biomedical applications. A considerable number of works have prepared HA-conjugated CNTs, generating new nanomaterials for delivery of insoluble anticancer drugs. For example, Yao and co-workers97 have fabricated a drug delivery system based on chitosan (CHI)-coated single-walled CNTs (SWNTs) loaded with salinomycin (SAL) and functionalized with HA (SAL-SWNTs-CHI-HA) with enhanced cellular uptake and therapeutic efficiency to gastric cancer stem cells (CSCs) (Figure 7). The results showed that the targeted drug delivery system significantly inhibited the migration and invasion of CSCs. Unlike pristine CNTs, HA-CNTs are stable in PBS and culture media. Similarly, Mo and co-workers98 have described a strategy to introduce DOX into HA and CHI-functionalized SWNTs (DOX-SWNTs-CHI-HA), which displayed high water solubility, low toxicity, high therapeutic efficacy against cancer and minimal adverse effects. Overall, this nanocomposite is a candidate for targeted cancer chemotherapy.

ACS Paragon Plus Environment

23

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 72

Figure 7. Schematic of the preparation of SAL-SWNTs-CHI-HA: chitosan (CHI)-coated SWNTs loaded with salinomycin (SAL) functionalized with HA. Reprinted from ref 97, Copyright 2014, with permission from Elsevier. DOX is the most extensively studied drug that is loaded onto carriers. Studies have focused on the development of DOX-loaded HA-CNTs as drug carriers to deliver DOX to various human tumour cells, such as lung epithelial cancer cell line A549 cells and Hela cells. For instance, Datir and colleagues91 reported a facile method to synthesize therapeutic agent, which is DOX loaded in HA-conjugated multi-walled CNTs (HA-MWNTs) via π-π stacking interaction. The in vitro and in vivo results showed that the cytotoxicity and tumour growth inhibitory effect of DOX-loaded HA-MWCNTs were higher than those of an equivalent concentration of free DOX, while reducing drug-associated cardiotoxicity. Thus, these properties render the synthesized HAMWNTs as suitable carriers in targetable drug delivery. In addition, other versatile nanomaterials based on HA-CNTs have been highlighted for drug delivery of anticancer agents for cancer treatments.95, 99 Overall, these HA-CNTs have been used as targetable drug delivery systems. Compared with traditional chemotherapy, CNTs have recently captured tremendous interest in laser-triggered PDT and PTT because of their strong light absorbance, high photothermal

ACS Paragon Plus Environment

24

Page 25 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

conversion efficiency and low tissue toxicity. The fundamental issues in PTT are focused on selective and effective phototherapy agents. For this reason, PTT nanoplatform based on HACNTs were developed recently. For example, hematoporphyrin monomethyl ether was introduced onto HA-CNTs for synergistic PTT and PDT antitumor therapy.100 In this study, the HA-CNTs nanomaterial had high optical absorbance in NIR region that can be applied for photothermal therapy. The results confirmed that the therapeutic effects of HA-CNTs are significantly higher than those of PTT or PDT treatment. In a follow-up study, another nanoplatform a nanophototherapy agent formed by conjugating ICG to HA nanoparticles encapsulated with SWCNTs, was tested for PTT and PDT treatments.94 The obtained nanoplatform exhibits favourable structural stability, biocompatibility, targetability and photothermal conversion efficiency, indicating the potential application of this nanophototherapy in CD44-targeted and image-guided dual PDT and PTT (Figure 8). The excellent properties of the nanoplatform allows for new exploration on the applications of PDT and PTT, and this nanoplatform demonstrated potential clinical translation. These nanocomposites are candidates for targeted PTT and PDT cancer treatment through sequential irradiation-activated apoptotic therapy. In addition to tumour-targeting drug delivery agents, HA-CNTs can enhance bone repair and regeneration and serve as a biosensing platform.101-103

ACS Paragon Plus Environment

25

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 72

Figure 8. Schematic of the formation of a dual targeted phototherapy agent, that is, ICGconjugated to HA nanoparticles encapsulated with SWCNTs: CD44 targeted delivery of ICGcoupled HA nanoparticles into tumors and following phototherapy use a NIR laser to irradiate the tumor area. Reprinted from ref 94. Copyright 2016 American Chemical Society. As mentioned above, several studies in the biomedical field have reported that HAfunctionalized CNTs demonstrates enhanced biocompatibility, physiological stability and absence of severe toxicity. However, the safety profile of CNTs remain largely undefined, limiting their practical application. To address this problem, researchers have conducted pioneering works to investigate the hazardous effects of MWCNTs. For example, exposure to MWCNT can lead to inflammation, fibrosis and granuloma formation in lungs. Therefore, evaluating the health risks of MWCNTs is urgently needed. An important application of HAMWCNT nanocomposite in reducing pulmonary injury was not discovered until recently.104 The results demonstrated that HA in nanocomposite significantly eliminated lung injury and reduced MWCNT-induced epithelial injury.

ACS Paragon Plus Environment

26

Page 27 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

In view of the above-mentioned concerns on HA-CNTs, we present in this section the progress made in the biomedical applications of HA-CNTs. HA-CNTs for biomedical applications were summarized in Table 2. We highlight the important HA- and CNT-based nanocomposites reported in literature and discuss their properties and their envision applications. The problem of specificity of CNTs in cell targeting may be solved by using HA as a targeting ligand. However, a major obstacle for the biomedical applications of HA-CNTs described in this section is the polydispersity of CNTs, which limits the reproducibility of the results and often affords inconsistent data. Furthermore, there are many pharmacology and toxicology challenges of these nanomaterials in vivo. Thus, more researches on HA-CNTs should be devoted to considering these important unresolved issues and challenges. Despite the factor that there are still many unresolved issues and challenges in HA-CNTs nanomedicine, the unique physiochemical properties and biological activity of HA-CNTs are attractive for various novel applications in biomedical fields. 2.5 HA-functionalized QDs (HA-QDs) and HA-functionalized carbon QDs (HA-CQDs) Fluorescent semiconductor nanocrystals (known as QDs) consist of hundreds to many thousands of atoms, which have been widely used in biosensing and bioimaging because of their unique optical and electronic properties.105, 106 Compared with conventional organic fluorescent dyes, QDs present many advantages, such as broad absorption spectra, symmetric size-tunable emission, high quantum yield and strong resistance to photobleaching, these properties render QDs with a broad application potential in the biomedical field.107 However, the major concern for QDs is their high toxicity resulting from the use of heavy metals in their production.108 Therefore, improvement in biocompatibility of QDs is an important challenge in their biomedical applications. The applications of QDs for specific absorption under physiological conditions

ACS Paragon Plus Environment

27

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 72

have not yet been reported. To address this issue, researchers have achieved rapid progress in surface functionalization of QDs, providing fundamental insights into reduction of systemic toxicity and enhancement of cellular targetability. Surface functionalization of QDs with various biocompatible and bioactive compounds, such as lipids, antibodies, peptides and polysaccharides, promotes their biomedical applications.106, 109 Proper surface functionalization is vital in successful applications of QDs in biomedicine. Among the QDs, polysaccharide-functionalized QDs have gained increasing attention because of their unique physiochemical and biological properties. HA-QDs not only reduce the systemic side effects of QDs but also render cancer cell targetable. Studies have focused on fabricating HA-QDs and investigating their biomedical application as bioimaging agents.42,

110, 111

For

example, Kim and colleagues112 have explored the use of QDs (CdSe) as bioimaging agents to assess the possibility of using HA derivatives as target-specific drug delivery carriers for treatment of liver diseases. Expectedly, the HA-QDs demonstrated a promising ability to actively target cells that cause chronic liver diseases through endocytosis. The results showed that HA and its derivatives demonstrated biocompatibility and cellular uptake characteristics, suggesting their potential as promising drug carriers for the treatment of various chronic liver diseases. To explore the targetable and biocompatible imaging agents, Bhang and colleagues fabricated HA-QDs nanocomposite through simple electrostatic attractions between HA and QDs (CdSe/CdS/ZnS Core/Shell/Shell) for cancer imaging and real-time visualization of changes in lymphatic vessels (Figure 9).113 Their work validated that HA-QDs exert significantly low cytotoxicity. In addition, they can label and visualize the lymphatic vessels in vitro and in vivo, reflecting the feasibility of using HA-QD nanocomposites as a biocompatible and targetable bioimaging agent. Overall, these techniques have opened up the use of QDs in biomedical

ACS Paragon Plus Environment

28

Page 29 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

applications, resulting in reduced cytotoxicity and enhanced targetability for bioimaging and drug delivery.

Figure 9. Schematic of HA-QDs conjugate. HA conjugated positively modified QDs by multiple electrostatic adsorption. Reprinted from ref 113. Copyright 2009 American Chemical Society. Although a considerable amount of effort has been made to improve the biocompatibility of QDs, the inherent cytotoxicity of QDs is one of the major obstacles limiting their further clinical applications. This issue prompted the fabrication of an alternative material, CQDs, which have attracted attention because of their inherently low toxicity, excellent biocompatibility, low cost, remarkable optical properties and chemical inertness.114 They have been applied in biomedical fields, such as in bioimaging, biosensing, drug delivery and cancer therapies. However, the lack of specific cell targeting on the surface of CQDs has been a critical problem to overcome in their biomedical applications. Therefore, in achieving increased cell selectivity, facile surface functionalization is required CQDs with high fluorescence stability, superior biocompatibility and targetability. Based on these considerations, HA-CQDs have recently attracted increasing attention and were successfully designed and used for targeted specific bioimaging and targeted

ACS Paragon Plus Environment

29

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 72

tumour theranostics.115 An example was presented by Zhang and colleagues116, who synthesized and utilized HA-CQDs as real-time bioimaging agent for targeted specific delivery of HA derivatives, whereas fluorescent CQDs were used as bioimaging agent for cancer cells. Given the high targeting ability of HA with CD44, the HA-CQDs expectedly showed a high target-specific delivery of HA-CQDs into the liver. The synthesized HA-CQDs possessed uniform sizedistribution, highly hydrophilic surface, and superior fluorescence property. The results confirmed that the HA-CQDs can not only be used as a drug delivery carrier to treat liver diseases but is also a promising bioimaging agent. In other works, HA was introduced on the surface of CQDs and graphene QD (GQD), to enhance their targetability.117, 118 These findings have confirmed that CQDs and GQD can be used as in vivo fluorescent probes and in vitro drug transporters. In addition to serving as drug carriers and fluorescent tracers, HA-CQDs have been designed for tumour diagnosis and chemotherapy. HA-QDs and HA-CQDs have been increasingly attracting attention for simultaneous drug delivery and fluorescent tracking primarily because of the integration of the optical properties of QDs or CQDs and the cancer cell targetability of HA. Table 2 shows HA-QDs and HA-CQDs for molecular imaging. However, there are several limitations associated with using HA-QDs and HA-CQDs in biomedical field because QDs are highly toxic at relatively low level,119, 120 even functionalized with HA, which may limit their use in clinical studies. The extent of cytotoxicity of HA-QDs and HA-CQDs in vivo needs to be determined. Another important issue is the lack of fundamental research concerning the effect of complex biological environments (salts, pH, and temperature) on optical properties of HA-QDs and HA-CQDs. In conclusion, irrespective of work on HA-QDs and HA-CQDs, further studies concerning these specific problems is still needed thus meeting the specific requirements in clinical applications.

ACS Paragon Plus Environment

30

Page 31 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Biomacromolecules

Table 2 Selected HA-based carbon nanomaterials for drug delivery systems, bioimaging and tumor theranostics. Systems

Mw of HA (Da)

7000k

nanogel CHA coated rGO

Stimuli responsive

DOX; thermotherapy

Light; pH

DOX

None

HA-grafted GO

Not given

DOX

None

HA-decorated GO

3.5k

DOX

pH

time noninvasive imaging, and light-

Lung cancer cells

excellent photo-luminescence

glutathione-responsive controlled drug

(A549)

Enhanced colloidal stability and

A tumor-targeting delivery system for

KB epidermal

improved in vivo safety

anticancer agents

carcinoma cells

Negligible hemolytic activity and low

A system for DOX delivery to the

HeLa and L929

cytotoxicity

tumors and suppress tumor growth.

cells

Superior physiological stability; high

A targeted and pH-responsive drug

drug loading capacity and delivery

delivery system for controlling the

efficiency

release of DOX for tumor therapy

100k

Epirubicin

pH

100k

PTT

None

214k

PTT

None

ICG loaded onto HAanchored rGO (HArGO) nanosheets Photosensitizers (PS; Ce6) loaded HA-GO

5.8k

PDT

None

230k

DOX

None

175k-350k

DOX

None

conjugate

H22 hepatic cancer

Refer ence

69

B16F1 melanoma

specific anti-cancer effect

cells

High light-to-heat conversion efficiency

Photothermal Ablation Therapy of

B16F1 melanoma

and low cytotoxicity

Skin Cancer

cells

A theranostic nano-platform for image-

Human KB

guided and synergistic photothermal

epidermal

antitumor therapy

carcinoma cells

Cancer targeted photodynamic therapy

Hela cells

Nontoxic; high fluorescence signal

In vivo imaging and target delivery

A549 cells

High anticancer efficiency and

Targeted killing of drug-resistant lung

A549 and MRC-5

fluorescence imaging

cancer cells

cells

High photostability and photothermal antitumor efficiency

High colloidal stability and photodynamic efficiency

78

79

80

cell

pH dependent drug release and target

specific anticancer drug delivery; pHresponsive release

GO-HA conjugate

Tumor models

Mono-dispersibility; good stability;

Enhanced serum stability; targeted GO-HA conjugate

Applications

release 214k

nanosheets

Properties

Thermo and chemotherapeutic, real-

Graphene-DOX conjugate in HA

Drugs

81

82

83

84

Photochromic dye spiropyran (SP) HA (HA-SP)

71

functionalized rGO HA-modified multifunctional Q-graphene

ACS Paragon Plus Environment

121

31

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

HA-decorated GO

HA-conjugated GO

HA and CHI modified SWNTs MWCNT-HA conjugate

9.6k

Not given

DOX

Nucleic acid

Redox

None

6

DOX

pH

Not given

DOX

pH

17.5k

Epirubicin (EPI)

None

Enhanced biostability; high photothermal response

Fluorescence-switchable cancer theranostic system

Page 32 of 72

Overcome multiple biological barriers results in specific and enhanced cancer treatment Target breast cancer cells, visualize the endogenous miR-21 and inhibit its

123

MBA-MB231 cells

tumorigenicity

High water solubility; low toxicity; high

Controlled release and targeted

therapeutic efficacy

delivery to cancer cells

Reduced drug-associated cardiotoxicity

122

MDA-MB-231 cells

A “smart” platform for tumor-targeted delivery of anticancer agents.

98

Hela cells 91

A549 cells

Istearoylphosphatidyl ethanolamine-HA (DSPE-HA) conjugated with

Stable dispersity; high biosafety; high

A promising carrier for drug delivery

delivery and antitumor efficiency

in multidrug resistance (MDR) cancers

95

A549 cells

SWNTs CHI-coated SWNTs loaded with SAL functionalized with

Not given

Salinomycin

None

Enhanced cellular uptake and therapeutic efficiency

HA

ICG-HA-SWCNTs

HA-SMWCNTs

HA-Cdots conjugates

234k

>1000k

2k

PTT and PDT

None

None

None

None

None

Favourable stability; biocompatibility; photothermal conversion efficiency Reduce pulmonary toxicity potential of SMWCNs Negligible cytotoxicity; strong fluorescence

Overcome the recurrence and metastasis of gastric cancer and improve gastric cancer treatment

Gastric cancer stem cells

Theranostic nanoparticle for CD44 targeted and image-guided dual PTT

97

94

SCC7 cells

and PDT cancer therapy Reduce pulmonary injury A target-specific drug delivery carrier for the treatment of liver diseases and a promising bioimaging agent.

Human bronchial

104

epithelial cells B16F1 and HEK293

118

cells 117

HA-GQD

230k

DOX

pH

234k

None

None

HA-5β-cholanic acid functionalized SWCNTs

Nontoxic; strong fluorescence High aqueous solubility; enhanced cell penetration; selective targeting

ACS Paragon Plus Environment

Target delivery and cell imaging

A549 cells

Molecular imaging

SCC7 and 3T3 cells

96

32

Page 33 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Biomacromolecules

HA-CQDs

HA-QDs conjugate (CdSe/CdS/ZnS)

Not given

Not given

None

None

None

None

Uniform size-distribution; highly

A novel cell-specific fluorescent

hydrophilic surface; superior

probes for CD44 high expression in

fluorescence property

tumor-targeted imaging and labelling

Low cytotoxicity; biocompatibility; targetability

116

Hela cells

HeLa cells, human In vivo lymphatic vessel imaging

113

dermal fibroblast (hDF) cells

Folate-terminated PEG modified HA (FA-PEG-HA)

400k

DOX

None

Real-time and noninvasive location

Dual receptor-mediated targeting

tracking to cancer cells

tumor theranostics

115

SKOV3 cells

conjugated with CDs HA-coated QDs ((CdSe)CdZnS)

HA-QDs conjugate (CdSe)

7.5k

Not given

None

None

None

None

Excellent fluorescence stability; no significant cytotoxicity

MD-MB-231 and CD44+ cancer cell-targeted imaging

cancer cells Novel drug delivery carriers for the

Hepatic stellate cells

Biocompatibility and enhanced cellular

treatment of various chronic liver

(HSC-T6) and

uptake

diseases including hepatitis, liver

hepatoma cells

cirrhosis, and liver cancer

(HepG2)

ACS Paragon Plus Environment

124

MCF-7 breast

112

33

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 34 of 72

2.6 HA-functionalized mesoporous silica nanomaterials Mesoporous silica nanoparticles (MSNs) have been greatly attracting attention in various nanotechnological applications, such as adsorption, catalysis, sensing and separation owing to their outstanding properties, such as controlled particle size, high surface area and pore volume, well-defined pore structures and chemical stability.125,

126

In addition, MSNs are excellent

nanomaterials on the basis of their potential biomedical applications.127,

128

In particular,

researchers have investigated MSNs, which are advanced drug delivery nanocarriers that demonstrate improved drug loading and controlled delivery properties owing to their abovementioned unique physiochemical properties, excellent biocompatibility, degradability under physiological conditions and their facile functionalized surface.129,

130

However, several key

factors limit the clinical application of MSNs. For example, a drug delivery system based on MSNs cannot transport drugs to specific target sites without any drug leakage into the blood circulation. Thus, the development of MSN nanocarriers that can transport and release drugs to specific sites in a selective and controlled manner has strongly attracted the attention of researchers. In overcoming this problem, MSNs functionalized with active targeted ligands, such as enzymes, antibodies and polysaccharides, were designed and applied as controlled drug delivery carriers, and they simultaneously increase colloidal stability, biocompatibility, targetability and precise drug release.128, 131 HA-functionalized MSNs (HA-MSNs) have been developed as intelligent nanocarriers to achieve targeted and controlled drug delivery into special cancer cells.132 Recently, Zhao et al.133 have developed a redox and enzyme dual-stimuli responsive delivery system (MSN-SS-HA) based on HA conjugated MSNs by cleavable disulfide (SS) bonds (Figure 10). The MSN-SS-HA nanomaterials had long-term stability and excellent dispersibility in physiological PBS as well as good biocompatibility.

ACS Paragon Plus Environment

34

Page 35 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

Figure 10. Schematic structure of MSN-SS-HA and dual-stimuli responsive targeted drug delivery system. A) drug loading process of MSN-SS-HA, (B) magnified image of pore structure after the grafting of HA, (C) cell uptake through a CD44 receptor-mediated interaction, and (D) GSH triggered drug release inside the tumor cell. Reprinted from ref 133, Copyright 2015 Acta Materialia Inc., with permission from Elsevier. The strong tendency of agglomeration and precipitation under physiological conditions have strongly limited their practical use of MSNs in biomedical fields.130 The great potential biomedical applications of MSNs have encouraged significant amount of efforts to address the problem on MSN dispersion. Recent works aiming to achieve this goal include the work of Ma and co-workers, who reported a novel synthesis method for HA-MSNs through facile amidation reaction.134 The results showed that the HA-MSNs exhibited excellent colloidal dispersibility in physiological fluids. Furthermore, it can selectively target specific cancer cells overexpressing CD44 receptors. In Hela cells, the therapeutic effect of CPT drug encapsulated into HA-MSNs was superior over the therapeutic effect of both free CPT and CPT-loaded HA-MSNs in the presence of excess free HA. Therefore, HA-MSNs is a potential new carrier for site-selective, controlled-release delivery of anticancer drugs.

ACS Paragon Plus Environment

35

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 72

Several MSN-based controlled drug delivery systems have been synthesized by using HA capping agents that can deliver drugs without any loss before reaching the targeted location. As mentioned earlier, HA-functionalized MSN-based drug delivery systems display several unique features, such as high colloidal stability, biodegradability, biocompatibility and site-specific delivery. In recent years, stimuli-responsive drug delivery systems based on HA-MSNs that release loaded drugs in response to photodynamic treatment, redox potential and pH have also received widespread attention. As discussed above, MSNs have been utilized in drug delivery systems. Beside drug carriers, it has been proved that MSNs can also be used in bioimaging.135 In most studies, the source of the fluorescence encapsulated within MSNs is dye. Instead of dye, Shi groups have designed a new type of theranostic nano-platform based on HA functionalized carbon and Si nanocrystals encapsulated in MSNs.136 Their result showed that such nanomaterials could specifically target cancer cells overexpressing CD44, exhibiting high drug delivery efficiency, and simultaneously, image the cancer cells in the NIR-to-Vis luminescence imaging fashion without using fluorescence dye. Table 3 summarizes the major drug delivery systems and bioimaging applications derived from HA-MSNs. Despite the exciting recent progresses in cellular systems, several key challenges need to be overcome to further facilitate the development of HA-MSNs for biomedicine applications. Firstly, protocols for reproducible synthesis and functionalizations of HA-MSNs are critical technique for this process. Additionally, efforts are still needed to study the long-term biocompatibility and pharmacokinetics of HA-MSNs.

ACS Paragon Plus Environment

36

Page 37 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Biomacromolecules

Table 3 Various drug delivery systems derived from HA-MSNs Carriers

Mw of HA (Da)

Drugs

Stimuli responsive

18k134; 35k137

camptothecin;

None

5-fluorouracil HA-mesoporous carbon spheres Oligosaccharide HAmesoporous silica

HA coated C60 fullerenesilica nanoparticle

100k138; 200k139

Not given

DOX 6mercaptopurine

DOX

anticancer drug for different tumors /

Hela cells/colo-

high therapeutic efficiency

drug delivery system for colon cancer

205 cancer cells

Redox

Redox

High drug loading and anticancer

Targeted co-delivery of drugs to tumors

HCT-116

efficiency

overexpressing CD44 receptors.

cells/Hela cells

High stability, biocompatibility,

A stimulus-responsive targeted drug

and cell uptake

delivery system

None

encapsulation efficiency; high antitumor efficacy

200k

DOX

pH/redox

dispersibility; good biocompatibility

HA conjugated carbonand Si nanocrystals-

18k

CPT

None

MSNs 8-hydroxyquinolineloaded HA-MSNs

370k

Docetaxel

None

1200k-1800k

None

None

HA/poly-L-lysine bilayered silica

Reference

134, 137

therapy

Long-term stability; excellent MSN-SS-HA

Tumor models

Excellent colloidal dispersibility;

Ultrahigh loading and Not given

Applications Enhanced site-specific delivery of

Carboplatin; HA-SiO2 nanoparticles

Properties

High drug delivery efficiency; NIR-to-Vis luminescence imaging

nanoparticles

MDA-MB-231

cancer stem-like cells

human breast

Targeted drug delivery to CD44overexpressing cancer cells

HCT-116 cells

133

MCF-7 and Cancer theranostics

MDA-MB-468

136

cells Eradication of breast cancer cells and stem cells

mesenchymal stem cells

140

cancer cells

systemic toxicity

differentiation of human

28

MCF-7 and Targeted drug delivery system to

High antitumor efficacy; little

Enhance the osteogenic

HCT-116 cells

138, 139

MCF-7 cells

A useful component of scaffolds for

Mesenchymal

bone tissue regeneration approaches

stem cells

MR imaging and targeted drug delivery

4T1 cells

141

142

Small dimensions; sustained drug Magnetic/HA silica nanotubes

100k

DOX

None

release; high superparamagnetism; enhanced colloidal stability and

143

cellular uptake

ACS Paragon Plus Environment

37

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 38 of 72

2.7 HA-functionalized magnetic nanoparticles (HA-MNPs) Magnetic resonance imaging (MRI) has emerged as a powerful tool for sensitive and specific detection of early-stage cancers because it is non-invasive and provides high-resolution and tomographic real-time images at the cellular and molecular levels.144 MNPs have been extensively utilized in many biomedical applications, such as MR imaging contrast agents and drug delivery vehicles for detection and treatment of cancer and other diseases; MNPs are utilized primarily because of their chemical stability and ability to function in biological interactions at the cellular and molecular levels.145 However, one of the major challenges in MNPs is that hydrophobic ligands on the surfaces led to colloidal instability in aqueous solutions. Therefore, the surface of MNPs must be modified to ensure their efficient dispersion in liquid media. Various natural and synthetic polymers, such as dextran and PEG, have been evaluated as coatings for MNPs.145 In another aspect, although MNPs can be nonspecifically taken up by cells, the development of specific amounts of targeting moieties on MNPs can facilitate the most efficient cellular uptake and imaging in vitro. Natural polymer HA, an attractive targeting ligand, has been widely used for functionalization of MNPs. Researchers have developed HA-MNPs and investigated their biological applications, and their results provide many approaches for targeted diagnosis and treatment of CD44-overexpressing cancers through receptor-mediated endocytosis.146 In recent years, HA has been one of the most widely utilized natural polymer coatings for MNP modification for in vivo applications.147, 148 To successfully integrate HA onto the surface of MNPs (Fe3O4), researchers have explored many strategies, including physical interaction or covalent bonding.149 As therapeutic tools, HA-MNPs (MnFe2O4) have been extensively applied for targeted detection and diagnosis of CD44-overexpressing breast cancer through MR

ACS Paragon Plus Environment

38

Page 39 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

imaging.150 Lim and co-workers21 have developed HA-MNPs (MnFe2O4), which exhibited superior biocompatibility and excellent capability for targeted detection and diagnosis of CD44overexpressing breast cancer via MRI, suggesting that the nanocomposites present promising potential application as MR imaging contrast agents for accurate tumour diagnosis and therapy (Figure 11).

Figure 11. Schematic illustration of HA-MNCs (MnFe2O4) for diagnosis of CD44-overexpressing breast cancers by magnetic resonance imaging (MRI). Reprinted from ref 21, Copyright 2011, with permission from Elsevier.

Another paradigm for cellular imaging using HA-MNPs was introduced by Chung and coworkers.151 In this work, HA-functionalized iron oxide nanoparticles could efficiently label the human mesenchymal stem cells with low toxicity and could greatly enhance MRI contrast, demonstrating their promising potential application as imaging probes in biomedical field. In another aspect, multifunctional magnetite nanoparticles composed of Fe3O4 nanoparticles and photosensitizer conjugated HA were prepared to achieve enhanced tumor diagnosis and therapy (Figure 12).152 The results suggested that the multifunctional magnetite nanoparticles can target tumors with enhanced tumor therapeutic effects through photodynamic/hyperthermia-combined

ACS Paragon Plus Environment

39

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 40 of 72

treatment. Overall, several studies have reported on the fabrication of HA-MNPs for molecular imaging, particularly in combination therapy for cancer diagnosis and treatment.153-157 Some typical systems based on HA-MNPs as imaging contrast agents and as drug carriers for cancer treatment have been summarized in Table 4.

Figure 12. Schematic illustration of preparation of HA coated Fe3O4 for tumor-targeted bimodal imaging and photodynamic/hyperthermia treatment. (A) acetylated HA-pheophorbide-a (AHP) coated Fe3O4 magnetic nanoparticles (AHP@MNPs) interacting with positively charged MNP through multibinding interactions. (B) AHP@MNPs irradiated with magnetic and near infrared lasers for tumor-targeted bimodal imaging and photodynamic/hyperthermia treatment. Reproduced from ref 152 with permission of The Royal Society of Chemistry. http://dx.doi.org/10.1039/C6NR02273A. While selecting HA-MNPs for drug delivery and molecular imaging systems, many important issues should be considered including but not limited to their pharmacokinetics, cytotoxicity, stability as well as their biodegradability, biocompatibility and potential side effects.

ACS Paragon Plus Environment

40

Page 41 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

Additionally, one important goal for further designing HA-MNPs is to improve the blood-brain barrier (BBB) transport of MNPs.

ACS Paragon Plus Environment

41

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

Page 42 of 72

Table 4 Typical systems based on HA-MNPs as imaging contrast agents and as drug carriers for cancer treatment Systems

Mw of HA (Da)

Imaging modes

Properties High colloidal stability; excellent cell

Applications

Reference

Cell labelling and in vivo imaging

158

HA-coated MNPs (Fe3O4)

31k

MRI

HA-modified Fe3O4

5.8k

MRI

Negative contrast agent

Detecting endometriosis in a mouse model

159

17k

MRI

Low toxicity and enhanced MRI contrast

Specifically label mesenchymal stem cells

151

20k

MRI

Accurate breast cancer cells diagnosis

146

Not given

MRI

Drug delivery vehicles and theranostic platform

153

6.8k

MRI

Tumor-targeted bimodal imaging and photodynamic

148

Hyperthermia combination therapy

149

Detect CD44-overexpressing breast cancer

21

HA-functionalized iron oxide nanoparticles (IONPs)/PEG HA-modified MnFe2O4 HA-coated superparamagnetic iron oxide nanoparticles (HA-SPIONs) HA-conjugated SPIONs

labeling efficiency

Effective CD44 binding ability; high cell viability Enhance the efficacy of chemotherapeutic drugs; non-invasive monitoring delivery Non-toxic, biocompatible, effective cancer targeting High biocompatibility; controllable

AHP-coated MNPs (Fe3O4)

5.8k

MRI

particle sizes; desirable magnetic properties, tumor growth inhibition efficacy

1000k

MRI

20k

MRI

HA-modified MnFe2O4

Excellent biocompatibility and superior targeting efficiency with MR sensitivity No cytotoxicity; colloidal stability;

Diagnosed tumor regions and detect CD44 abundant

targeted imaging efficacy

cancer cell

157

ACS Paragon Plus Environment

42

Page 43 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

As far as the preparation technology of nanoparticles is concerned, it should be noted that the layer-by-layer (LbL) technology that utilizes the process of sequentially depositing oppositely charged polymers to build highly stable films on substrates has been a new and highly promising approach for nanomedicine applications.160 In addition to the HA-functionalized nanohybrid materials described above, great progress in HA employed for the preparation of LbL nanoparticles has been achieved for biotechnological and biomedical applications. For example, recent studies have demonstrated the use of HA-LbL nanoparticles (such as PS latex nanospheres, AuNPs and QDs) for tumor-specific cancer diagnostics and therapy and systemic delivery.161,

162

Highly versatile HA-LbL nanoparticles as tailor-made delivery vehicles have

been shown capable of enhancing the efficacy and specificity of therapeutics. Taking advantage of the LbL-assembled multilayered nanoparticles with various sizes, architectures, and chemical compositions, an available powerful platform can be constructed for drug encapsulation, triggered drug release, and hierarchical assemblies. 3. Limitations of HA that may impact HA-based nanohybrid materials A multitude of reports demonstrate that HA can modulate many biological effects such as cell adhesion and migration, tumorigenesis, cell survival and apoptosis, and inflammation etc.163 In addition, many researchers have established the concept that HA plays different roles depending on its molecular weight. For example, high molecular weight HA (generally over 1000kDa) has shown to be anti-inflammatory, however, low molecular weight HA promotes the production of inflammatory mediators and induces the tumor progression.164-166 Therefore, the molecular weight variants of HA used in a variety of biomedical applications have elicited varying biological responses.167 It is not rare to see such fact in polysaccharides.

ACS Paragon Plus Environment

43

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 44 of 72

Another important issue is the degradation of HA. HA is readily degraded into smaller fragments in the human body by enzymatic degradation of hyaluronidase (HAse), which specifically hydrolyze the β-1,4 linkage of the HA molecules.168 It should be noted that the molecular weight of HA may affect the HA uptake by cell and modulate biological responses, although the exact mechanism is still far from completely understood.169 It has been shown that low molecular weight HA derived from high molecular weight HA has distinct biological functions.170 These limitations or defects of HA itself like easy degradability and negative effects rising from low molar mass HA are often neglected in fabricating nanohybrid materials in most of literatures. Furthermore, as described earlier, HA and its conjugates have been extensively utilized in biomedical applications mostly due to its high binding affinity to CD44 receptor. Therefore, maintaining the unique biological property of HA binding to CD44 receptor is very important when modifying HA. It has been reported that CD44 interacts with a minimum HA length of 6 to 8 saccharides171, i.e., only small targeting moiety (oligosaccharide) interacts with CD44 receptor, which facilitates the utilization of HA conjugates in drug delivery. In many of the studies described above (e.g. ref 94, 97, and 123), the intracellular uptake of HA derivatives by CD44mediated endocytosis has been confirmed by using fluorescent labeled-HA derivatives. These results showed that HA derivatives can be specifically and efficiently internalized into CD44overexpressed cells. However, notably, there has been clear evidence that excessive chemical modification of HA will alter its biological functions. For example, while 35 mol.% HA modification maintained the ability to bind CD44, 68 mol.% modification lost the unique biological function of HA.110 To our knowledge, there are few work done for investigating the effect of chemical modification of HA on the HA-CD44 interaction. Therefore, taking into

ACS Paragon Plus Environment

44

Page 45 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

account the importance of maintaining the unique biological functions of HA, attention must be paid to ensure the CD44 binding ability of HA-based nanomaterials. Therefore, despite many excellent properties of HA, several limitations of HA that associate with using it to functionalize nanomaterials for biomedical applications should be stressed. These include, 1) despite that chemical modification of HA can control the degradation rate, how to exactly control and reduce the degradation rate of HA into fragments in human body remains a challenge; 2) at which level the degree of chemical modification of HA is befitting to retain the binding ability of HA to CD44 receptor; 3) low molecular weight HA may accumulate in the body, leading to local deposits that may cause unwanted side effects in the long-term; and 4) low molecular weight HA that may induce expression of proinflammatory cytokines, chemokines, and growth factors, which may cause the adverse effects on body and severely limit the extensive applications of HA-functionalized nanomaterials in biomedical filed. Hence, more extensive studies are still needed to fully clarify the influence of HA-functionalized nanomaterials on biological effects, which will enable us to bring more innovative applications of HAfunctionalized nanomaterials without progression of inflammatory disease. 4. Conclusions and Future Perspectives The application of nanotechnology in biomedical field is widely expected to change the landscape of pharmaceutical and biotechnology industries for the foreseeable future. Over the past several decades, various nanomaterials, especially Au, Ag, carbon materials, mesoporous materials and MNPs were developed for biomedical applications as imaging probes, drug carriers and contrast agents owing to their unique structures and several distinctive physical and chemical attributes. Although significant progress has been made in the field of nanomaterials,

ACS Paragon Plus Environment

45

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 46 of 72

several significant challenges must be investigated and must be addressed to promote the practical applications of these nanomaterials. For example, the most important disadvantage of these nanomaterials is the potential toxicity related to long-term safety, which has greatly limited their clinical translation. In addition, these nanomaterials cannot be specifically taken up by target cells and they lack stability under physiological conditions. Therefore, to overcome these limitations, researchers have focused on the functionalization of nanomaterials while retaining their inherent unique physiochemical properties. Since its discovery, HA has been widely used and has gained success in cosmetic and biomedical fields. HA can be employed as a carrier and a targeting ligand for the selective accumulation of therapeutic and diagnostic entities in diseased areas overexpressing CD44 receptors. In this review, we have summarized the recent developments in HA-nanomaterials, from AuNPs, AgNPs, graphene derivatives, CNTs, QDs, CQDs and mesoporous materials to MNPs. Moreover, this review highlights the applications of HA-nanomaterials in the biomedical field, such as in drug delivery, bioimaging and detection and diagnosis of cancer cells. These applications have attracted the interest of many researchers globally. Numerous studies on HAnanomaterials are in progress, and many interesting applications of HA-nanomaterials were demonstrated by diverse research groups; the biomedical applications of these nanomaterials range from tissue engineering and molecular imaging to targeted drug delivery. Various applications of HA-nanomaterials are expected to be further investigated, although scale-up production of HA-nanomaterials may remain a major challenge. In order to further facilitate the development of HA-nanomaterials for biomedical applications, the following further studies including some important considerations should be emphasized: 1) even though it has been reported that HA-nanomaterials are nontoxic in vitro, it is important to

ACS Paragon Plus Environment

46

Page 47 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

investigate the long-term toxicity of these nanomaterials exposure on human physiology; 2) equally important is considering how the HA-nanomaterials size, shape, structure, conjugated ligands, surface properties and polydispersity influence the pharmacokinetics, biodistribution and eventual side effect in vivo (These deep investigations can give the reproducibility of the results and afford consistent data); 3) when we design the HA-nanomaterials utilized in drug delivery and bioimaging systems, more research should be done considering the balance between the biological activities of HA and the physiochemical properties of inorganic nanomaterials; 4) HAnanomaterials should be an effective platform for the delivery of anticancer drugs to overcome biological barriers (e.g., vascular or cellular barrier, blood--brain barrier); 5) more investigations are still needed to get a better understanding of how we can extrapolate the potential effects on human health from their biological behavior observed in animal studies; 6) further work should aim at designing and preparing HA-nanomaterials that can be used in large-scale applications in biomedical field; 7) with knowledge gained concerning how the molecular weight of HA influences the biological effects, new developments of HA-functionalized nanomaterials and relevant strategies are needed to make either HA itself or modified nanomaterials more biocompatible and approved; and 8) HA has the positive interactions with proteins, nucleic acids and other biological compounds, which will lead to research into the introduction of proteins and nucleic acids in HA-nanomaterials. These envisions point the way to much broader, enhanced prospects for HA-nanomaterials in biomedical areas. Addressing these questions will undoubtedly accelerate the development of HA-nanomaterials for biological applications. In closing, rapid advances in the development and applications of HA-nanomaterials in many biomedical fields underline the great potential and important role of HA in these nanomaterials. As research in HA-nanomaterials progresses, we

ACS Paragon Plus Environment

47

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 48 of 72

are looking forward to developing more innovative strategies to broaden their biomedical applications. AUTHOR INFORMATION Corresponding Author Hongbin Zhang; E-mail: [email protected] Acknowledgements The authors are thankful for the financial support for this work from the National Natural Science Foundation of China (Grant No. 21074071, 21274090). Abbreviations HA, hyaluronan; HAase, hyaluronidase; Mw, molecular weight; AuNPs, gold nanoparticles; AgNPs, silver nanoparticles; GO, graphene oxide; rGO, reduced graphene oxide; CNTs, carbon nanotubes; CHI, chitosan; CSCs, gastric cancer stem cells; QDs, quantum dots; Cdots, carbon dots; GQD, graphene quantum dot; MSNs, mesoporous silica nanoparticles; DOX, doxorubicin hydrochloride; CPT, camptothecin; CPT-11, irinotecan hydrochloride; TPT, topotecan hydrochloride; β-CD, β-cyclodextrin; CHA, cholesteryl-modified HA; BPEI, branched polyethylenimine; SAL, salinomycin; HCV, hepatitis C virus; IFNα, interferon α; ROS, reactive oxygen species; PS, polystyrene; NIR, near-infrared; SPR, surface plasmon resonance; EPR, enhanced permeation and retention; ADH, adipic acid dihydrazide; BSA, bovine serum albumin; SERS, surface-enhanced raman scattering; SP, spiropyran; CT, X-ray computed tomography; SPECT, single-photon emission computed tomography; MRI, magnetic resonance imaging; FTIC, Fluorescein isothiocyanate; Py, Pyrene; Vis, visible light; CDDSs, controlled drug delivery systems; PTT, photothermal treatment; ICG, indocyanine green; PDT, photodynamic

ACS Paragon Plus Environment

48

Page 49 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

therapy; EDC, N-ethyl-N’-dimethylaminopropyl-carbodiimide; NHS, N-hydroxysuccinimide; SWNTs, single-walled CNTs; MWNTs, multi-walled CNTs; MDR, multidrug resistance; MNPs, magnetic nanoparticles; SPIONs, superparamagnetic iron oxide nanoparticles; AHP, acetylated HA-pheophorbide-a; LbL, layer-by-layer; BBB, blood-brain barrier. REFERENCES 1.

Sahoo, S. K.; Labhasetwar, V., Nanotech approaches to drug delivery and imaging. Drug

Discovery Today 2003, 8, (24), 1112-1120. 2.

El-Boubbou, K.; Huang, X. F., Glyco-Nanomaterials: Translating Insights from the

"Sugar-Code" to Biomedical Applications. Curr Med Chem 2011, 18, (14), 2060-2078. 3.

Davis, M. E.; Chen, Z.; Shin, D. M., Nanoparticle therapeutics: an emerging treatment

modality for cancer. Nat Rev Drug Discov 2008, 7, (9), 771-782. 4.

Zhang, L.; Gao, S.; Zhang, F.; Yang, K.; Ma, Q.; Zhu, L., Activatable Hyaluronic Acid

Nanoparticle as a Theranostic Agent for Optical/Photoacoustic Image-Guided Photothermal Therapy. ACS Nano 2014, 8, (12), 12250-12258. 5.

Grodzinski, P.; Silver, M.; Molnar, L. K., Nanotechnology for cancer diagnostics:

promises and challenges. Expert Review of Molecular Diagnostics 2006, 6, (3), 307-318. 6.

Hong, S. Y.; Green, M. L. H.; Davis, B. G., Recent Biotechnological Applications of

Glyco-Nanomaterials. In Petite and Sweet: Glyco-Nanotechnology as a Bridge to New Medicines, American Chemical Society: 2011; Vol. 1091, pp 1-13. 7.

Sapsford, K. E.; Algar, W. R.; Berti, L.; Gemmill, K. B.; Casey, B. J.; Oh, E.; Stewart, M.

H.; Medintz, I. L., Functionalizing Nanoparticles with Biological Molecules: Developing Chemistries that Facilitate Nanotechnology. Chemical Reviews 2013, 113, (3), 1904-2074.

ACS Paragon Plus Environment

49

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

8.

Page 50 of 72

El-Dakdouki, M.; Huang, X., Biological Applications of Hyaluronic Acid Functionalized

Nanomaterials. In Petite and Sweet: Glyco-Nanotechnology as a Bridge to New Medicines, American Chemical Society: 2011; Vol. 1091, pp 181-213. 9.

Kang, D.; Cai, Z. X.; Jin, Q. W.; Zhang, H. B., Bio-inspired composite films with

integrative properties based on the self-assembly of gellan gum-graphene oxide crosslinked nanohybrid building blocks. Carbon 2015, 91, 445-457. 10.

Fraser, J. R. E.; Laurent, T. C.; Laurent, U. B. G., Hyaluronan: its nature, distribution,

functions and turnover. Journal of Internal Medicine 1997, 242, (1), 27-33. 11.

Yu, F. Y.; Zhang, F.; Luan, T.; Zhang, Z. N.; Zhang, H. B., Rheological studies of

hyaluronan solutions based on the scaling law and constitutive models. Polymer 2014, 55, (1), 295-301. 12.

Luan, T.; Fang, Y. P.; Al-Assaf, S.; Phillips, G. O.; Zhang, H. B., Compared molecular

characterization of hyaluronan using multiple-detection techniques. Polymer 2011, 52, (24), 5648-5658. 13.

Luan, T.; Wu, L. J.; Zhang, H. B.; Wang, Y., A study on the nature of intermolecular

links in the cryotropic weak gels of hyaluronan. Carbohyd Polym 2012, 87, (3), 2076-2085. 14.

Burdick, J. A.; Prestwich, G. D., Hyaluronic Acid Hydrogels for Biomedical Applications.

Adv Mater 2011, 23, (12), H41-H56. 15.

Sherman, L.; Sleeman, J.; Herrlich, P.; Ponta, H., Hyaluronate receptors: key players in

growth, differentiation, migration and tumor progression. Current Opinion in Cell Biology 1994, 6, (5), 726-733.

ACS Paragon Plus Environment

50

Page 51 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

16.

Twomey, M.; Na, Y.; Roche, Z.; Mendez, E.; Panday, N.; He, J.; Moon, J. H.,

Fabrication of Core–Shell Nanoparticles via Controlled Aggregation of Semiflexible Conjugated Polymer and Hyaluronic Acid. Macromolecules 2013, 46, (15), 6374-6378. 17.

Lapčík, L.; Lapčík, L.; De Smedt, S.; Demeester, J.; Chabreček, P., Hyaluronan: 

Preparation, Structure, Properties, and Applications. Chemical Reviews 1998, 98, (8), 2663-2684. 18.

Kim, H.; Jeong, H.; Han, S.; Beack, S.; Hwang, B. W.; Shin, M.; Oh, S. S.; Hahn, S. K.,

Hyaluronate and its derivatives for customized biomedical applications. Biomaterials 2017, 123, 155-171. 19.

Li, N.; Chen, Y.; Zhang, Y.-M.; Yang, Y.; Su, Y.; Chen, J.-T.; Liu, Y., Polysaccharide-

Gold Nanocluster Supramolecular Conjugates as a Versatile Platform for the Targeted Delivery of Anticancer Drugs. Scientific Reports 2014, 4, 4164. 20.

Wang, L.; Zhang, H.; Qin, A.; Jin, Q.; Tang, B. Z.; Ji, J., Theranostic hyaluronic acid

prodrug micelles with aggregation-induced emission characteristics for targeted drug delivery. Science China Chemistry 2016, 59, (12), 1609-1615. 21.

Lim, E. K.; Kim, H. O.; Jang, E.; Park, J.; Lee, K.; Suh, J. S.; Huh, Y. M.; Haam, S.,

Hyaluronan-modified magnetic nanoclusters for detection of CD44-overexpressing breast cancer by MR imaging. Biomaterials 2011, 32, (31), 7941-7950. 22.

Lam, J.; Truong, N. F.; Segura, T., Design of cell-matrix interactions in hyaluronic acid

hydrogel scaffolds. Acta Biomater 2014, 10, (4), 1571-1580. 23.

El-Dakdouki, M. H.; Zhu, D. C.; El-Boubbou, K.; Kamat, M.; Chen, J.; Li, W.; Huang,

X., Development of Multifunctional Hyaluronan-Coated Nanoparticles for Imaging and Drug Delivery to Cancer Cells. Biomacromolecules 2012, 13, (4), 1144-1151.

ACS Paragon Plus Environment

51

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

24.

Page 52 of 72

Liu, X.; Huang, R.; Su, R.; Qi, W.; Wang, L.; He, Z., Grafting Hyaluronic Acid onto

Gold Surface to Achieve Low Protein Fouling in Surface Plasmon Resonance Biosensors. ACS Applied Materials & Interfaces 2014, 6, (15), 13034-13042. 25.

Zhang, F.; Wu, J.; Kang, D.; Zhang, H. B., Development of a complex hydrogel of

hyaluronan and PVA embedded with silver nanoparticles and its facile studies on Escherichia coli. J Biomat Sci-Polym E 2013, 24, (12), 1410-1425. 26.

Xing, X. R.; Liu, S.; Yu, J. H.; Lian, W. J.; Huang, J. D., Electrochemical sensor based

on molecularly imprinted film at polypyrrole-sulfonated graphene/hyaluronic acid-multiwalled carbon nanotubes modified electrode for determination of tryptamine. Biosens Bioelectron 2012, 31, (1), 277-283. 27.

Dvash, R.; Khatchatouriants, A.; Solmesky, L. J.; Wibroe, P. P.; Wei, M.; Moghimi, S.

M.; Peer, D., Structural profiling and biological performance of phospholipid-hyaluronan functionalized single-walled carbon nanotubes. J Control Release 2013, 170, (2), 295-305. 28.

Zhao, Q.; Geng, H.; Wang, Y.; Gao, Y.; Huang, J.; Wang, Y.; Zhang, J.; Wang, S.,

Hyaluronic Acid Oligosaccharide Modified Redox-Responsive Mesoporous Silica Nanoparticles for Targeted Drug Delivery. ACS Applied Materials & Interfaces 2014, 6, (22), 20290-20299. 29.

Dykman, L.; Khlebtsov, N., Gold nanoparticles in biomedical applications: recent

advances and perspectives. Chem Soc Rev 2012, 41, (6), 2256-2282. 30.

Giljohann, D. A.; Seferos, D. S.; Daniel, W. L.; Massich, M. D.; Patel, P. C.; Mirkin, C.

A., Gold Nanoparticles for Biology and Medicine. Angew Chem Int Edit 2010, 49, (19), 32803294.

ACS Paragon Plus Environment

52

Page 53 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

31.

Auffan, M.; Rose, J.; Bottero, J. Y.; Lowry, G. V.; Jolivet, J. P.; Wiesner, M. R., Towards

a definition of inorganic nanoparticles from an environmental, health and safety perspective. Nat Nanotechnol 2009, 4, (10), 634-641. 32.

Rai, M.; Yadav, A.; Gade, A., Silver nanoparticles as a new generation of antimicrobials.

Biotechnol Adv 2009, 27, (1), 76-83. 33.

Shervani, Z.; Yamamoto, Y., Carbohydrate-directed synthesis of silver and gold

nanoparticles: effect of the structure of carbohydrates and reducing agents on the size and morphology of the composites. Carbohyd Res 2011, 346, (5), 651-658. 34.

Kemp, M. M.; Kumar, A.; Mousa, S.; Park, T. J.; Ajayan, P.; Kubotera, N.; Mousa, S. A.;

Linhardt, R. J., Synthesis of Gold and Silver Nanoparticles Stabilized with Glycosaminoglycans Having Distinctive Biological Activities. Biomacromolecules 2009, 10, (3), 589-595. 35.

Hien, N. Q.; Phu, D. V.; Duy, N. N.; Quoc, L. A., Radiation synthesis and

characterization of hyaluronan capped gold nanoparticles. Carbohyd Polym 2012, 89, (2), 537541. 36.

Jiang, W.; Kim, B. Y. S.; Rutka, J. T.; Chan, W. C. W., Nanoparticle-mediated cellular

response is size-dependent. Nat Nanotechnol 2008, 3, (3), 145-150. 37.

Lee, M. Y.; Yang, J. A.; Jung, H. S.; Beack, S.; Choi, J. E.; Hur, W.; Koo, H.; Kim, K.;

Yoon, S. K.; Hahn, S. K., Hyaluronic Acid-Gold Nanoparticle/Interferon alpha Complex for Targeted Treatment of Hepatitis C Virus Infection. Acs Nano 2012, 6, (11), 9522-9531. 38.

Agasti, S. S.; Rana, S.; Park, M. H.; Kim, C. K.; You, C. C.; Rotello, V. M.,

Nanoparticles for detection and diagnosis. Adv Drug Deliver Rev 2010, 62, (3), 316-328. 39.

Sarsour, E. H.; Kumar, M. G.; Chaudhuri, L.; Kalen, A. L.; Goswami, P. C., Redox

Control of the Cell Cycle in Health and Disease. Antioxid Redox Sign 2009, 11, (12), 2985-3011.

ACS Paragon Plus Environment

53

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

40.

Page 54 of 72

Lee, K.; Lee, H.; Lee, K. W.; Park, T. G., Optical imaging of intracellular reactive

oxygen species for the assessment of the cytotoxicity of nanoparticles. Biomaterials 2011, 32, (10), 2556-2565. 41.

Hyun, H.; Lee, K.; Min, K. H.; Jeon, P.; Kim, K.; Jeong, S. Y.; Kwon, I. C.; Park, T. G.;

Lee, M., Ischemic brain imaging using fluorescent gold nanoprobes sensitive to reactive oxygen species. J Control Release 2013, 170, (3), 352-357. 42.

Wang, Z. Z.; Chen, Z. W.; Liu, Z.; Shi, P.; Dong, K.; Ju, E. G.; Ren, J. S.; Qu, X. G., A

multi-stimuli responsive gold nanocage-hyaluronic platform for targeted photothermal and chemotherapy. Biomaterials 2014, 35, (36), 9678-9688. 43.

Matteini, P.; Ratto, F.; Rossi, F.; Rossi, G.; Esposito, G.; Puca, A.; Albanese, A.; Maira,

G.; Pini, R., In vivo carotid artery closure by laser activation of hyaluronan-embedded gold nanorods. J Biomed Opt 2010, 15, (4). 44.

Oh, Y. J.; Jeong, K. H., Glass Nanopillar Arrays with Nanogap-Rich Silver Nanoislands

for Highly Intense Surface Enhanced Raman Scattering. Adv Mater 2012, 24, (17), 2234-2237. 45.

Hack, D.; Chauhan, P.; Deckers, K.; Hermann, G. N.; Mertens, L.; Raabe, G.; Enders, D.,

Combining Silver Catalysis and Organocatalysis: A Sequential Michael Addition/Hydroalkoxylation One-Pot Approach to Annulated Coumarins. Org Lett 2014, 16, (19), 5188-5191. 46.

Gao, C. B.; Lu, Z. D.; Liu, Y.; Zhang, Q.; Chi, M. F.; Cheng, Q.; Yin, Y. D., Highly

Stable Silver Nanoplates for Surface Plasmon Resonance Biosensing. Angew Chem Int Edit 2012, 51, (23), 5629-5633. 47.

Chernousova, S.; Epple, M., Silver as Antibacterial Agent: Ion, Nanoparticle, and Metal.

Angew Chem Int Edit 2013, 52, (6), 1636-1653.

ACS Paragon Plus Environment

54

Page 55 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

48.

Cui, X. Q.; Li, C. M.; Bao, H. F.; Zheng, X. T.; Zang, J. F.; Ooi, C. P.; Guo, J.,

Hyaluronan-assisted photoreduction synthesis of silver nanostructures: From nanoparticle to nanoplate. J Phys Chem C 2008, 112, (29), 10730-10734. 49.

Xia, N. X.; Cai, Y. R.; Jiang, T.; Yao, J. M., Green synthesis of silver nanoparticles by

chemical reduction with hyaluronan. Carbohyd Polym 2011, 86, (2), 956-961. 50.

Kemp, M. M.; Kumar, A.; Clement, D.; Ajayan, P.; Mousa, S.; Linhardt, R. J.,

Hyaluronan- and heparin-reduced silver nanoparticles with antimicrobial properties. Nanomedicine-Uk 2009, 4, (4), 421-429. 51.

Chen, C. H.; Chen, S. H.; Shalumon, K. T.; Chen, J. P., Dual functional core-sheath

electrospun hyaluronic acid/polycaprolactone nanofibrous membranes embedded with silver nanoparticles for prevention of peritendinous adhesion. Acta Biomater 2015, 26, 225-235. 52.

Cui, X. Q.; Li, C. M.; Bao, H. F.; Zheng, X. T.; Lu, Z. S., In situ fabrication of silver

nanoarrays in hyaluronan/PDDA layer-by-layer assembled structure. J Colloid Interf Sci 2008, 327, (2), 459-465. 53.

Zhang, F.; Wu, J.; Zhang, H. B., Construction of hyaluronan-silver nanoparticle-

hemoglobin multilayer composite film and investigations on its electrocatalytic properties. J Solid State Electr 2012, 16, (4), 1683-1692. 54.

Abdel-Mohsen, A. M.; Hrdina, R.; Burgert, L.; Abdel-Rahman, R. M.; Hasova, M.;

Smejkalova, D.; Kolar, M.; Pekar, M.; Aly, A. S., Antibacterial activity and cell viability of hyaluronan fiber with silver nanoparticles. Carbohyd Polym 2013, 92, (2), 1177-1187. 55.

Abdel-Mohsen, A. M.; Hrdina, R.; Burgert, L.; Krylova, G.; Abdel-Rahman, R. M.;

Krejcova, A.; Steinhart, M.; Benes, L., Green synthesis of hyaluronan fibers with silver nanoparticles. Carbohyd Polym 2012, 89, (2), 411-422.

ACS Paragon Plus Environment

55

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

56.

Page 56 of 72

Sacco, P.; Sechi, A.; Trevisan, A.; Picotti, F.; Gianni, R.; Stucchi, L.; Fabbian, M.; Bosco,

M.; Paoletti, S.; Marsich, E., A silver complex of hyaluronan-lipoate (SHLS12): Synthesis, characterization and biological properties. Carbohyd Polym 2016, 136, 418-426. 57.

Liu, J. H.; Zhao, Y. X.; Guo, Q. Q.; Wang, Z.; Wang, H. Y.; Yang, Y. X.; Huang, Y. Z.,

TAT-modified nanosilver for combating multidrug-resistant cancer. Biomaterials 2012, 33, (26), 6155-6161. 58.

Mandal, A.; Sekar, S.; Seeni Meera, K. M.; Mukherjee, A.; Sastry, T. P.; Mandal, A. B.,

Fabrication of collagen scaffolds impregnated with sago starch capped silver nanoparticles suitable for biomedical applications and their physicochemical studies. Physical Chemistry Chemical Physics 2014, 16, (37), 20175-20183. 59.

Liang, J. M.; Zeng, F.; Zhang, M.; Pan, Z. Z.; Chen, Y. Z.; Zeng, Y. N.; Xu, Y.; Xu, Q.;

Huang, Y. Z., Green synthesis of hyaluronic acid-based silver nanoparticles and their enhanced delivery to CD44(+) cancer cells. Rsc Adv 2015, 5, (54), 43733-43740. 60.

Zhang, X.; Yao, M. N.; Chen, M. H.; Li, L. Q.; Dong, C. Y.; Hou, Y.; Zhao, H. Y.; Jia,

B.; Wang, F., Hyaluronic Acid-Coated Silver Nanoparticles As a Nanoplatform for in Vivo Imaging Applications. Acs Applied Materials & Interfaces 2016, 8, (39), 25650-25653. 61.

Lee, H.; Lee, K.; Kim, I. K.; Park, T. G., Fluorescent Gold Nanoprobe Sensitive to

Intracellular Reactive Oxygen Species. Adv Funct Mater 2009, 19, (12), 1884-1890. 62.

Liu, X.; Gao, C.; Gu, J.; Jiang, Y.; Yang, X.; Li, S.; Gao, W.; An, T.; Duan, H.; Fu, J.;

Wang, Y.; Yang, X., Hyaluronic Acid Stabilized Iodine-Containing Nanoparticles with Au Nanoshell Coating for X-ray CT Imaging and Photothermal Therapy of Tumors. ACS Applied Materials & Interfaces 2016, 8, (41), 27622-27631.

ACS Paragon Plus Environment

56

Page 57 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

63.

Wang, R.; Luo, Y.; Yang, S.; Lin, J.; Gao, D.; Zhao, Y.; Liu, J.; Shi, X.; Wang, X.,

Hyaluronic acid-modified manganese-chelated dendrimer-entrapped gold nanoparticles for the targeted CT/MR dual-mode imaging of hepatocellular carcinoma. Scientific Reports 2016, 6, 33844. 64.

Rau, L. R.; Tsao, S. W.; Liaw, J. W.; Tsai, S. W., Selective Targeting and Restrictive

Damage for Nonspecific Cells by Pulsed Laser-Activated Hyaluronan-Gold Nanoparticles. Biomacromolecules 2016, 17, (8), 2514-2521. 65.

Han, H. S.; Choi, K. Y.; Lee, H.; Lee, M.; An, J. Y.; Shin, S.; Kwon, S.; Lee, D. S.; Park,

J. H., Gold-Nanoclustered Hyaluronan Nano-Assemblies for Photothermally Maneuvered Photodynamic Tumor Ablation. ACS Nano 2016, 10, (12), 10858-10868. 66.

Lee, H. W.; Lee, J. H.; Kim, J. S.; Mun, J. H.; Chung, J. H.; Koo, H.; Kim, C. H.; Yun, S.

H.; Hahn, S. K., Hyaluronate-Gold Nanorod/DR5 Antibody Complex for Noninvasive Theranosis of Skin Cancer. Acs Applied Materials & Interfaces 2016, 8, (47), 32202-32210. 67.

Cheng, D.; Han, W. Y.; Yang, K. C.; Song, Y.; Jiang, M. D.; Song, E. Q., One-step facile

synthesis of hyaluronic acid functionalized fluorescent gold nanoprobes sensitive to hyaluronidase in urine specimen from bladder cancer patients. Talanta 2014, 130, 408-414. 68.

Geim, A. K., Graphene: Status and Prospects. Science 2009, 324, (5934), 1530-1534.

69.

Khatun, Z.; Nurunnabi, M.; Nafiujjaman, M.; Reeck, G. R.; Khan, H. A.; Cho, K. J.; Lee,

Y. K., A hyaluronic acid nanogel for photo-chemo theranostics of lung cancer with simultaneous light-responsive controlled release of doxorubicin. Nanoscale 2015, 7, (24), 10680-10689. 70.

Lu, C. H.; Yang, H. H.; Zhu, C. L.; Chen, X.; Chen, G. N., A Graphene Platform for

Sensing Biomolecules. Angew Chem Int Edit 2009, 48, (26), 4785-4787.

ACS Paragon Plus Environment

57

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

71.

Page 58 of 72

Nahain, A.-A.; Lee, J. E.; Jeong, J. H.; Park, S. Y., Photoresponsive Fluorescent Reduced

Graphene Oxide by Spiropyran Conjugated Hyaluronic Acid for in Vivo Imaging and Target Delivery. Biomacromolecules 2013, 14, (11), 4082-4090. 72.

Zhang, F.; Chen, X. J.; Boulos, R. A.; Yasin, F. M.; Lu, H. B.; Raston, C.; Zhang, H. B.,

Pyrene-conjugated hyaluronan facilitated exfoliation and stabilisation of low dimensional nanomaterials in water. Chem Commun 2013, 49, (42), 4845-4847. 73.

Zhang, F.; Yasin, F. M.; Chen, X. J.; Mo, J. X.; Raston, C. L.; Zhang, H. B., Functional

noble metal nanostructures involving pyrene-conjugated-hyaluronan stabilised reduced graphene oxide. Rsc Adv 2013, 3, (47), 25166-25174. 74.

Shim, G.; Kim, M.-G.; Park, J. Y.; Oh, Y.-K., Graphene-based nanosheets for delivery of

chemotherapeutics and biological drugs. Adv Drug Deliver Rev 2016, 105, Part B, 205-227. 75.

Lin, J.; Chen, X.; Huang, P., Graphene-based nanomaterials for bioimaging. Adv Drug

Deliver Rev 2016, 105, Part B, 242-254. 76.

Tannock, I. F.; Rotin, D., Acid pH in Tumors and Its Potential for Therapeutic

Exploitation. Cancer Research 1989, 49, (16), 4373-4384. 77.

Yang, K.; Feng, L.; Liu, Z., Stimuli responsive drug delivery systems based on nano-

graphene for cancer therapy. Adv Drug Deliver Rev 2016, 105, Part B, 228-241. 78.

Miao, W.; Shim, G.; Kang, C. M.; Lee, S.; Choe, Y. S.; Choi, H. G.; Oh, Y. K.,

Cholesteryl hyaluronic acid-coated, reduced graphene oxide nanosheets for anti-cancer drug delivery. Biomaterials 2013, 34, (37), 9638-9647. 79.

Wu, H. X.; Shi, H. L.; Wang, Y. P.; Jia, X. Q.; Tang, C. Z.; Zhang, J. M.; Yang, S. P.,

Hyaluronic acid conjugated graphene oxide for targeted drug delivery. Carbon 2014, 69, 379389.

ACS Paragon Plus Environment

58

Page 59 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

80.

Song, E. Q.; Han, W. Y.; Li, C.; Cheng, D.; Li, L. R.; Liu, L. C.; Zhu, G. Z.; Song, Y.;

Tan, W. H., Hyaluronic Acid-Decorated Graphene Oxide Nanohybrids as Nanocarriers for Targeted and pH-Responsive Anticancer Drug Delivery. Acs Applied Materials & Interfaces 2014, 6, (15), 11882-11890. 81.

Jung, H. S.; Lee, M. Y.; Kong, W. H.; Do, I. H.; Hahn, S. K., Nano graphene oxide-

hyaluronic acid conjugate for target specific cancer drug delivery. Rsc Adv 2014, 4, (27), 1419714200. 82.

Jung, H. S.; Kong, W. H.; Sung, D. K.; Lee, M. Y.; Beack, S. E.; Keum, D. H.; Kim, K.

S.; Yun, S. H.; Hahn, S. K., Nanographene Oxide-Hyaluronic Acid Conjugate for Photothermal Ablation Therapy of Skin Cancer. Acs Nano 2014, 8, (1), 260-268. 83.

Miao, W.; Shim, G.; Kim, G.; Lee, S.; Lee, H. J.; Kim, Y. B.; Byun, Y.; Oh, Y. K.,

Image-guided synergistic photothermal therapy using photoresponsive imaging agent-loaded graphene-based nanosheets. J Control Release 2015, 211, 28-36. 84.

Li, F.; Park, S.; Ling, D.; Park, W.; Han, J. Y.; Na, K.; Char, K., Hyaluronic acid-

conjugated graphene oxide/photosensitizer nanohybrids for cancer targeted photodynamic therapy. J Mater Chem B 2013, 1, (12), 1678-1686. 85.

Peng, C.; Hu, W. B.; Zhou, Y. T.; Fan, C. H.; Huang, Q., Intracellular Imaging with a

Graphene-Based Fluorescent Probe. Small 2010, 6, (15), 1686-1692. 86.

Yang, K.; Feng, L. Z.; Shi, X. Z.; Liu, Z., Nano-graphene in biomedicine: theranostic

applications. Chem Soc Rev 2013, 42, (2), 530-547. 87.

Sun, X. M.; Liu, Z.; Welsher, K.; Robinson, J. T.; Goodwin, A.; Zaric, S.; Dai, H. J.,

Nano-Graphene Oxide for Cellular Imaging and Drug Delivery. Nano Res 2008, 1, (3), 203-212.

ACS Paragon Plus Environment

59

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

88.

Page 60 of 72

Kim, H.; Namgung, R.; Singha, K.; Oh, I. K.; Kim, W. J., Graphene Oxide-

Polyethylenimine Nanoconstruct as a Gene Delivery Vector and Bioimaging Tool. Bioconjugate Chem 2011, 22, (12), 2558-2567. 89.

Fabbro, C.; Ali-Boucetta, H.; Da Ros, T.; Kostarelos, K.; Bianco, A.; Prato, M.,

Targeting carbon nanotubes against cancer. Chem Commun 2012, 48, (33), 3911-3926. 90.

Prakash, S.; Malhotra, M.; Shao, W.; Tomaro-Duchesneau, C.; Abbasi, S., Polymeric

nanohybrids and functionalized carbon nanotubes as drug delivery carriers for cancer therapy. Adv Drug Deliver Rev 2011, 63, (14-15), 1340-1351. 91.

Datir, S. R.; Das, M.; Singh, R. P.; Jain, S., Hyaluronate Tethered, "Smart" Multiwalled

Carbon Nanotubes for Tumor-Targeted Delivery of Doxorubicin. Bioconjugate Chem 2012, 23, (11), 2201-2213. 92.

Liu, Z.; Tabakman, S.; Welsher, K.; Dai, H. J., Carbon Nanotubes in Biology and

Medicine: In vitro and in vivo Detection, Imaging and Drug Delivery. Nano Res 2009, 2, (2), 85120. 93.

Bhirde, A. A.; Patel, V.; Gavard, J.; Zhang, G. F.; Sousa, A. A.; Masedunskas, A.;

Leapman, R. D.; Weigert, R.; Gutkind, J. S.; Rusling, J. F., Targeted Killing of Cancer Cells in Vivo and in Vitro with EGF-Directed Carbon Nanotube-Based Drug Delivery. Acs Nano 2009, 3, (2), 307-316. 94.

Wang, G. H.; Zhang, F.; Tian, R.; Zhang, L. W.; Fu, G. F.; Yang, L. L.; Zhu, L.,

Nanotubes-Embedded Indocyanine Green-Hyaluronic Acid Nanoparticles for PhotoacousticImaging-Guided Phototherapy. Acs Applied Materials & Interfaces 2016, 8, (8), 5608-5617. 95.

Yao, H. J.; Sun, L.; Liu, Y.; Jiang, S.; Pu, Y. Z.; Li, J. C.; Zhang, Y. G.,

Monodistearoylphosphatidylethanolamine-hyaluronic acid functionalization of single-walled

ACS Paragon Plus Environment

60

Page 61 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

carbon nanotubes for targeting intracellular drug delivery to overcome multidrug resistance of cancer cells. Carbon 2016, 96, 362-376. 96.

Swierczewska, M.; Choi, K. Y.; Mertz, E. L.; Huang, X. L.; Zhang, F.; Zhu, L.; Yoon, H.

Y.; Park, J. H.; Bhirde, A.; Lee, S.; Chen, X. Y., A Facile, One-Step Nanocarbon Functionalization for Biomedical Applications. Nano Lett 2012, 12, (7), 3613-3620. 97.

Yao, H. J.; Zhang, Y. G.; Sun, L.; Liu, Y., The effect of hyaluronic acid functionalized

carbon nanotubes loaded with salinomycin on gastric cancer stem cells. Biomaterials 2014, 35, (33), 9208-9223. 98.

Mo, Y. F.; Wang, H. W.; Liu, J. H.; Lan, Y.; Guo, R.; Zhang, Y.; Xue, W.; Zhang, Y. M.,

Controlled release and targeted delivery to cancer cells of doxorubicin from polysaccharidefunctionalised single-walled carbon nanotubes. J Mater Chem B 2015, 3, (9), 1846-1855. 99.

Cao, X. Y.; Tao, L.; Wen, S. H.; Hou, W. X.; Shi, X. Y., Hyaluronic acid-modified

multiwalled carbon nanotubes for targeted delivery of doxorubicin into cancer cells. Carbohyd Res 2015, 405, 70-77. 100.

Shi, J. J.; Ma, R. R.; Wang, L.; Zhang, J.; Liu, R. Y.; Li, L. L.; Liu, Y.; Hou, L.; Yu, X.

Y.; Gao, J.; Zhang, Z. Z., The application of hyaluronic acid-derivatized carbon nanotubes in hematoporphyrin monomethyl ether-based photodynamic therapy for in vivo and in vitro cancer treatment. Int J Nanomed 2013, 8, 2361-2373. 101.

Martins, P. A.; Sa, M. A.; Reis, A. C.; Queiroz, C. M.; Caliari, M. V.; Teixeira, M. M.;

Ladeira, L. O.; Pinho, V.; Ferreira, A. J., Evaluation of carbon nanotubes functionalized with sodium hyaluronate in the inflammatory processes for oral regenerative medicine applications. Clin Oral Invest 2016, 20, (7), 1607-1616.

ACS Paragon Plus Environment

61

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

102.

Page 62 of 72

Mendes, R. M.; Silva, G. A. B.; Caliari, M. V.; Silva, E. E.; Ladeira, L. O.; Ferreira, A. J.,

Effects of single wall carbon nanotubes and its functionalization with sodium hyaluronate on bone repair. Life Sci 2010, 87, (7-8), 215-222. 103.

Filip, J.; Sefcovicova, J.; Tomcik, P.; Gemeiner, P.; Tkac, J., A hyaluronic acid dispersed

carbon nanotube electrode used for a mediatorless NADH sensing and biosensing. Talanta 2011, 84, (2), 355-361. 104.

Hussain, S.; Ji, Z. X.; Taylor, A. J.; DeGraff, L. M.; George, M.; Tucker, C. J.; Chang, C.

H.; Li, R. B.; Bonner, J. C.; Garantziotis, S., Multiwalled Carbon Nanotube Functionalization with High Molecular Weight Hyaluronan Significantly Reduces Pulmonary Injury. Acs Nano 2016, 10, (8), 7675-7688. 105.

Chan, W. C. W.; Maxwell, D. J.; Gao, X. H.; Bailey, R. E.; Han, M. Y.; Nie, S. M.,

Luminescent quantum dots for multiplexed biological detection and imaging. Curr Opin Biotech 2002, 13, (1), 40-46. 106.

Medintz, I. L.; Uyeda, H. T.; Goldman, E. R.; Mattoussi, H., Quantum dot bioconjugates

for imaging, labelling and sensing. Nat Mater 2005, 4, (6), 435-446. 107.

Mattoussi, H.; Palui, G.; Na, H. B., Luminescent quantum dots as platforms for probing

in vitro and in vivo biological processes. Adv Drug Deliver Rev 2012, 64, (2), 138-166. 108.

Larson, D. R.; Zipfel, W. R.; Williams, R. M.; Clark, S. W.; Bruchez, M. P.; Wise, F. W.;

Webb, W. W., Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science 2003, 300, (5624), 1434-1436. 109.

Dubertret, B.; Skourides, P.; Norris, D. J.; Noireaux, V.; Brivanlou, A. H.; Libchaber, A.,

In vivo imaging of quantum dots encapsulated in phospholipid micelles. Science 2002, 298, (5599), 1759-1762.

ACS Paragon Plus Environment

62

Page 63 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

110.

Kim, J.; Kim, K. S.; Jiang, G.; Kang, H.; Kim, S.; Kim, B. S.; Park, M. H.; Hahn, S. K.,

In Vivo Real-Time Bioimaging of Hyaluronic Acid Derivatives Using Quantum Dots. Biopolymers 2008, 89, (12), 1144-1153. 111.

Guo, M. R.; Law, W. C.; Ng, C. H.; Cai, H. X.; Liu, L. W.; Zhao, L. H.; Zhang, X. H.,

Preparation of Size Tunable, Glutathione-Responsive Hyaluronic Acid-Quantum Dot Nanohybrids Using Microemulsion Method. Sci Adv Mater 2015, 7, (2), 364-370. 112.

Kim, K. S.; Hur, W.; Park, S. J.; Hong, S. W.; Choi, J. E.; Goh, E. J.; Yoon, S. K.; Hahn,

S. K., Bioimaging for Targeted Delivery of Hyaluronic Acid Derivatives to the Livers in Cirrhotic Mice Using Quantum Dots. Acs Nano 2010, 4, (6), 3005-3014. 113.

Bhang, S. H.; Won, N.; Lee, T. J.; Jin, H.; Nam, J.; Park, J.; Chung, H.; Park, H. S.; Sung,

Y. E.; Hahn, S. K.; Kim, B. S.; Kim, S., Hyaluronic Acid-Quantum Dot Conjugates for In Vivo Lymphatic Vessel Imaging. Acs Nano 2009, 3, (6), 1389-1398. 114.

Lim, S. Y.; Shen, W.; Gao, Z. Q., Carbon quantum dots and their applications. Chem Soc

Rev 2015, 44, (1), 362-381. 115.

Jia, X.; Han, Y.; Pei, M. L.; Zhao, X. B.; Tian, K.; Zhou, T. T.; Liu, P., Multi-

functionalized hyaluronic acid nanogels crosslinked with carbon dots as dual receptor-mediated targeting tumor theranostics. Carbohyd Polym 2016, 152, 391-397. 116.

Zhang, M.; Fang, Z.; Zhao, X.; Niu, Y.; Lou, J.; Zhao, L.; Wu, Y.; Zou, S.; Du, F.; Shao,

Q., Hyaluronic acid functionalized nitrogen-doped carbon quantum dots for targeted specific bioimaging. Rsc Adv 2016, 6, (107), 104979-104984. 117.

Abdullah-Al-Nahain; Lee, J. E.; In, I.; Lee, H.; Lee, K. D.; Jeong, J. H.; Park, S. Y.,

Target Delivery and Cell Imaging Using Hyaluronic Acid-Functionalized Graphene Quantum Dots. Mol Pharmaceut 2013, 10, (10), 3736-3744.

ACS Paragon Plus Environment

63

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

118.

Page 64 of 72

Goh, E. J.; Kim, K. S.; Kim, Y. R.; Jung, H. S.; Beack, S.; Kong, W. H.; Scarcelli, G.;

Yun, S. H.; Hahn, S. K., Bioimaging of Hyaluronic Acid Derivatives Using Nanosized Carbon Dots. Biomacromolecules 2012, 13, (8), 2554-2561. 119.

Lin, P.; Chen, J. W.; Chang, L. W.; Wu, J. P.; Redding, L.; Chang, H.; Yeh, T. K.; Yang,

C. S.; Tsai, M. H.; Wang, H. J.; Kuo, Y. C.; Yang, R. S. H., Computational and ultrastructural toxicology of a nanoparticle, Quantum Dot 705, in mice. Environ Sci Technol 2008, 42, (16), 6264-6270. 120.

Yang, S. T.; Cao, L.; Luo, P. G. J.; Lu, F. S.; Wang, X.; Wang, H. F.; Meziani, M. J.; Liu,

Y. F.; Qi, G.; Sun, Y. P., Carbon Dots for Optical Imaging in Vivo. J Am Chem Soc 2009, 131, (32), 11308-11309. 121.

Luo, Y. A.; Cai, X. L.; Li, H.; Lin, Y. H.; Du, D., Hyaluronic Acid-Modified

Multifunctional Q-Graphene for Targeted Killing of Drug-Resistant Lung Cancer Cells. Acs Applied Materials & Interfaces 2016, 8, (6), 4048-4055. 122.

Yin, T.; Liu, J.; Zhao, Z.; Zhao, Y.; Dong, L.; Yang, M.; Zhou, J.; Huo, M., Redox

Sensitive Hyaluronic Acid-Decorated Graphene Oxide for Photothermally Controlled TumorCytoplasm-Selective Rapid Drug Delivery. Adv Funct Mater 2017, 1604620. 123.

Hwang, D. W.; Kim, H. Y.; Li, F.; Park, J. Y.; Kim, D.; Park, J. H.; Han, H. S.; Byun, J.

W.; Lee, Y.-S.; Jeong, J. M.; Char, K.; Lee, D. S., In vivo visualization of endogenous miR-21 using hyaluronic acid-coated graphene oxide for targeted cancer therapy. Biomaterials 2017, 121, 144-154. 124.

Wang, H. N.; Sun, H. F.; Wei, H.; Xi, P.; Nie, S. M.; Ren, Q. S., Biocompatible

hyaluronic acid polymer-coated quantum dots for CD44(+) cancer cell-targeted imaging. J Nanopart Res 2014, 16, (10).

ACS Paragon Plus Environment

64

Page 65 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

125.

Medda, L.; Casula, M. F.; Monduzzi, M.; Salis, A., Adsorption of Lysozyme on

Hyaluronic Acid Functionalized SBA-15 Mesoporous Silica: A Possible Bioadhesive Depot System. Langmuir 2014, 30, (43), 12996-13004. 126.

Suzuki, K.; Ikari, K.; Imai, H., Synthesis of silica nanoparticles having a well-ordered

mesostructure using a double surfactant system. J Am Chem Soc 2004, 126, (2), 462-463. 127.

Slowing, I. I.; Vivero-Escoto, J. L.; Wu, C. W.; Lin, V. S. Y., Mesoporous silica

nanoparticles as controlled release drug delivery and gene transfection carriers. Adv Drug Deliver Rev 2008, 60, (11), 1278-1288. 128.

Yang, P. P.; Gai, S. L.; Lin, J., Functionalized mesoporous silica materials for controlled

drug delivery. Chem Soc Rev 2012, 41, (9), 3679-3698. 129.

Tang, F. Q.; Li, L. L.; Chen, D., Mesoporous Silica Nanoparticles: Synthesis,

Biocompatibility and Drug Delivery. Adv Mater 2012, 24, (12), 1504-1534. 130.

Rosenholm, J.; Sahlgren, C.; Linden, M., Cancer-cell targeting and cell-specific delivery

by mesoporous silica nanoparticles. J Mater Chem 2010, 20, (14), 2707-2713. 131.

Gan, Q.; Zhu, J. Y.; Yuan, Y.; Liu, H. L.; Qian, J. C.; Lib, Y. S.; Liu, C. S., A dual-

delivery system of pH-responsive chitosan-functionalized mesoporous silica nanoparticles bearing BMP-2 and dexamethasone for enhanced bone regeneration. J Mater Chem B 2015, 3, (10), 2056-2066. 132.

Chen, Z. W.; Li, Z. H.; Lin, Y. H.; Yin, M. L.; Ren, J. S.; Qu, X. G., Bioresponsive

Hyaluronic Acid-Capped Mesoporous Silica Nanoparticles for Targeted Drug Delivery. ChemEur J 2013, 19, (5), 1778-1783.

ACS Paragon Plus Environment

65

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 66 of 72

133. Zhao, Q. F.; Liu, J.; Zhu, W. Q.; Sun, C. S.; Di, D. H.; Zhang, Y.; Wang, P.; Wang, Z. Y.; Wang, S. L., Dual-stimuli responsive hyaluronic acid-conjugated mesoporous silica for targeted delivery to CD44-overexpressing cancer cells. Acta Biomater 2015, 23, 147-156. 134.

Ma, M.; Chen, H. R.; Chen, Y.; Zhang, K.; Wang, X.; Cui, X. Z.; Shi, J. L., Hyaluronic

acid-conjugated mesoporous silica nanoparticles: excellent colloidal dispersity in physiological fluids and targeting efficacy. J Mater Chem 2012, 22, (12), 5615-5621. 135.

Tallury, P.; Payton, K.; Santra, S., Silica-based multimodal/multifunctional nanoparticles

for bioimaging and biosensing applications. Nanomedicine-Uk 2008, 3, (4), 579-592. 136.

He, Q. J.; Ma, M.; Wei, C. Y.; Shi, J. L., Mesoporous carbon@silicon-silica

nanotheranostics for synchronous delivery of insoluble drugs and luminescence imaging. Biomaterials 2012, 33, (17), 4392-4402. 137.

Liu, K.; Wang, Z. Q.; Wang, S. J.; Liu, P.; Qin, Y. H.; Ma, Y.; Li, X. C.; Huo, Z. J.,

Hyaluronic acid-tagged silica nanoparticles in colon cancer therapy: therapeutic efficacy evaluation. Int J Nanomed 2015, 10, 6445-6454. 138.

Wan, L.; Jiao, J.; Cui, Y.; Guo, J. W.; Han, N.; Di, D. H.; Chang, D.; Wang, P.; Jiang, T.

Y.; Wang, S. L., Hyaluronic acid modified mesoporous carbon nanoparticles for targeted drug delivery to CD44-overexpressing cancer cells. Nanotechnology 2016, 27, (13). 139.

Zhang, J.; Sun, Y. J.; Tian, B. C.; Li, K. K.; Wang, L. L.; Liang, Y.; Han, J. T.,

Multifunctional mesoporous silica nanoparticles modified with tumor-shedable hyaluronic acid as carriers for doxorubicin. Colloid Surface B 2016, 144, 293-302. 140.

Wang, H.; Agarwal, P.; Zhao, S. T.; Yu, J. H.; Lu, X. B.; He, X. M., Combined cancer

therapy with hyaluronan-decorated fullerene-silica multifunctional nanoparticles to target cancer stem-like cells. Biomaterials 2016, 97, 62-73.

ACS Paragon Plus Environment

66

Page 67 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

141.

Wang, D.; Huang, J. B.; Wang, X. X.; Yu, Y.; Zhang, H.; Chen, Y.; Liu, J. J.; Sun, Z. G.;

Zou, H.; Sun, D. X.; Zhou, G. C.; Zhang, G. Q.; Lu, Y.; Zhong, Y. Q., The eradication of breast cancer cells and stem cells by 8-hydroxyquinoline-loaded hyaluronan modified mesoporous silica nanoparticle-supported lipid bilayers containing docetaxel. Biomaterials 2013, 34, (31), 7662-7673. 142.

Amorim, S.; Martins, A.; Neves, N. M.; Reis, R. L.; Pires, R. A., Hyaluronic acid/poly-L-

lysine bilayered silica nanoparticles enhance the osteogenic differentiation of human mesenchymal stem cells. J Mater Chem B 2014, 2, (40), 6939-6946. 143.

Huang, L.; Ao, L. J.; Wang, W.; Hu, D. H.; Sheng, Z. H.; Su, W., Multifunctional

magnetic silica nanotubes for MR imaging and targeted drug delivery. Chem Commun 2015, 51, (18), 3923-3926. 144.

Veiseh, O.; Gunn, J. W.; Zhang, M. Q., Design and fabrication of magnetic nanoparticles

for targeted drug delivery and imaging. Adv Drug Deliver Rev 2010, 62, (3), 284-304. 145.

Sun, C.; Lee, J. S. H.; Zhang, M. Q., Magnetic nanoparticles in MR imaging and drug

delivery. Adv Drug Deliver Rev 2008, 60, (11), 1252-1265. 146.

Lee, T.; Lim, E. K.; Lee, J.; Kang, B.; Choi, J.; Park, H. S.; Suh, J. S.; Huh, Y. M.; Haam,

S., Efficient CD44-targeted magnetic resonance imaging (MRI) of breast cancer cells using hyaluronic acid (HA)-modified MnFe2O4 nanocrystals. Nanoscale Res Lett 2013, 8, 1-9. 147.

Kamat, M.; El-Boubbou, K.; Zhu, D. C.; Lansdell, T.; Lu, X. W.; Li, W.; Huang, X. F.,

Hyaluronic Acid Immobilized Magnetic Nanoparticles for Active Targeting and Imaging of Macrophages. Bioconjugate Chem 2010, 21, (11), 2128-2135.

ACS Paragon Plus Environment

67

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

148.

Page 68 of 72

Thomas, R. G.; Moon, M. J.; Lee, H.; Sasikala, A. R. K.; Kim, C. S.; Park, I. K.; Jeong,

Y. Y., Hyaluronic acid conjugated superparamagnetic iron oxide nanoparticle for cancer diagnosis and hyperthermia therapy. Carbohyd Polym 2015, 131, 439-446. 149.

Xiong, Z. C.; Qin, H. Q.; Wan, H.; Huang, G.; Zhang, Z.; Dong, J.; Zhang, L. Y.; Zhang,

W. B.; Zou, H. F., Layer-by-layer assembly of multilayer polysaccharide coated magnetic nanoparticles for the selective enrichment of glycopeptides. Chem Commun 2013, 49, (81), 9284-9286. 150.

Lee, Y. H.; Lee, H.; Kim, Y. B.; Kim, J. Y.; Hyeon, T.; Park, H.; Messersmith, P. B.;

Park, T. G., Bioinspired Surface Immobilization of Hyaluronic Acid on Monodisperse Magnetite Nanocrystals for Targeted Cancer Imaging. Adv Mater 2008, 20, (21), 4154-4157. 151.

Chung, H. J.; Lee, H.; Bae, K. H.; Lee, Y.; Park, J.; Cho, S. W.; Hwang, J. Y.; Park, H.;

Langer, R.; Anderson, D.; Park, T. G., Facile Synthetic Route for Surface-Functionalized Magnetic Nanoparticles: Cell Labeling and Magnetic Resonance Imaging Studies. Acs Nano 2011, 5, (6), 4329-4336. 152.

Kim, K. S.; Kim, J.; Lee, J. Y.; Matsuda, S.; Hideshima, S.; Mori, Y.; Osaka, T.; Na, K.,

Stimuli-responsive magnetic nanoparticles for tumor-targeted bimodal imaging and photodynamic/hyperthermia combination therapy. Nanoscale 2016, 8, (22), 11625-11634. 153.

El-Dakdouki, M. H.; Xia, J. G.; Zhu, D. C.; Kavunja, H.; Grieshaber, J.; O'Reilly, S.;

McCormick, J. J.; Huang, X. F., Assessing the in Vivo Efficacy of Doxorubicin Loaded Hyaluronan Nanoparticles. Acs Applied Materials & Interfaces 2014, 6, (1), 697-705. 154.

Manju, S.; Sreenivasan, K., Enhanced Drug Loading on Magnetic Nanoparticles by

Layer-by-Layer Assembly Using Drug Conjugates: Blood Compatibility Evaluation and Targeted Drug Delivery in Cancer Cells. Langmuir 2011, 27, (23), 14489-14496.

ACS Paragon Plus Environment

68

Page 69 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

155.

Li, J. C.; Hu, Y.; Yang, J.; Wei, P.; Sun, W. J.; Shen, M. W.; Zhang, G. X.; Shi, X. Y.,

Hyaluronic acid-modified Fe3O4@Au core/shell nanostars for multimodal imaging and photothermal therapy of tumors. Biomaterials 2015, 38, 10-21. 156.

Yang, R.-M.; Fu, C.-P.; Fang, J.-Z.; Xu, X.-D.; Wei, X.-H.; Tang, W.-J.; Jiang, X.-Q.;

Zhang, L.-M., Hyaluronan-modified superparamagnetic iron oxide nanoparticles for bimodal breast cancer imaging and photothermal therapy. Int J Nanomed 2017, 12, 197-206. 157.

Lee, T.; Son, H. Y.; Choi, Y.; Shin, Y.; Oh, S.; Kim, J.; Huh, Y.-M.; Haam, S., Minimum

hyaluronic acid (HA) modified magnetic nanocrystals with less facilitated cancer migration and drug resistance for targeting CD44 abundant cancer cells by MR imaging. J Mater Chem B 2017, 5, (7), 1400-1407. 158.

El-Dakdouki, M. H.; El-Boubbou, K.; Zhu, D. C.; Huang, X. F., A simple method for the

synthesis of hyaluronic acid coated magnetic nanoparticles for highly efficient cell labelling and in vivo imaging. Rsc Adv 2011, 1, (8), 1449-1452. 159.

Zhang, H.; Li, J. C.; Sun, W. J.; Hu, Y.; Zhang, G. F.; Shen, M. W.; Shi, X. Y.,

Hyaluronic Acid-Modified Magnetic Iron Oxide Nanoparticles for MR Imaging of Surgically Induced Endometriosis Model in Rats. Plos One 2014, 9, (4). 160.

Yan, Y.; Bjonmalm, M.; Caruso, F., Assembly of Layer-by-Layer Particles and Their

Interactions with Biological Systems. Chem Mater 2014, 26, (1), 452-460. 161.

Poon, Z.; Lee, J. B.; Morton, S. W.; Hammond, P. T., Controlling in Vivo Stability and

Biodistribution in Electrostatically Assembled Nanoparticles for Systemic Delivery. Nano Lett 2011, 11, (5), 2096-2103.

ACS Paragon Plus Environment

69

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

162.

Page 70 of 72

Dreaden, E. C.; Morton, S. W.; Shopsowitz, K. E.; Choi, J. H.; Deng, Z. J.; Cho, N. J.;

Hammond, P. T., Bimodal Tumor-Targeting from Microenvironment Responsive Hyaluronan Layer-by-Layer (LbL) Nanoparticles. Acs Nano 2014, 8, (8), 8374-8382. 163.

Cyphert, J. M.; Trempus, C. S.; Garantziotis, S., Size Matters: Molecular Weight

Specificity of Hyaluronan Effects in Cell Biology. International Journal of Cell Biology 2015, 2015, 8. 164.

Jiang, D. H.; Liang, J. R.; Noble, P. W., Hyaluronan as an Immune Regulator in Human

Diseases. Physiol Rev 2011, 91, (1), 221-264. 165.

Termeer, C.; Benedix, F.; Sleeman, J.; Fieber, C.; Voith, U.; Ahrens, T.; Miyake, K.;

Freudenberg, M.; Galanos, C.; Simon, J. C., Oligosaccharides of Hyaluronan activate dendritic cells via toll-like receptor 4. J Exp Med 2002, 195, (1), 99-111. 166.

Toole, B. P.; Ghatak, S.; Misra, S., Hyaluronan Oligosaccharides as a Potential

Anticancer Therapeutic. Current Pharmaceutical Biotechnology 2008, 9, (4), 249-252. 167.

Rayahin, J. E.; Buhrman, J. S.; Zhang, Y.; Koh, T. J.; Gemeinhart, R. A., High and Low

Molecular Weight Hyaluronic Acid Differentially Influence Macrophage Activation. Acs Biomater Sci Eng 2015, 1, (7), 481-493. 168.

Gushulak, L.; Hemming, R.; Martin, D.; Seyrantepe, V.; Pshezhetsky, A.; Triggs-Raine,

B., Hyaluronidase 1 and β-Hexosaminidase Have Redundant Functions in Hyaluronan and Chondroitin Sulfate Degradation. Journal of Biological Chemistry 2012, 287, (20), 16689-16697. 169.

Stern, R.; Jedrzejas, M. J., Hyaluronidases: Their genomics, structures, and mechanisms

of action. Chemical Reviews 2006, 106, (3), 818-839. 170.

West, D. C.; Hampson, I. N.; Arnold, F.; Kumar, S., Angiogenesis induced by

degradation products of hyaluronic acid. Science 1985, 228, (4705), 1324-6.

ACS Paragon Plus Environment

70

Page 71 of 72

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

171.

Platt, V. M.; Szoka, F. C., Anticancer therapeutics: Targeting macromolecules and

nanocarriers to hyaluronan or CD44, a hyaluronan receptor. Mol Pharmaceut 2008, 5, (4), 474486.

ACS Paragon Plus Environment

71

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 72 of 72

TOC

ACS Paragon Plus Environment

72