Next Article in Journal / Special Issue
Effect of S-triazine Ring Substitution on the Synthesis of Organic Resorcinol-Formaldehyde Xerogels
Previous Article in Journal
Editorial on Special Issues “Aerogels” and “Aerogels 2018”
Previous Article in Special Issue
Characterization of Enriched Meat-Based Pâté Manufactured with Oleogels as Fat Substitutes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Thermoresponsive Nanogels Based on Different Polymeric Moieties for Biomedical Applications

by
Sobhan Ghaeini-Hesaroeiye
1,†,
Hossein Razmi Bagtash
1,†,
Soheil Boddohi
1,*,
Ebrahim Vasheghani-Farahani
1,* and
Esmaiel Jabbari
2
1
Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14115, Iran
2
Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 5 May 2020 / Revised: 21 June 2020 / Accepted: 25 June 2020 / Published: 4 July 2020
(This article belongs to the Special Issue Gels: 6th Anniversary)

Abstract

:
Nanogels, or nanostructured hydrogels, are one of the most interesting materials in biomedical engineering. Nanogels are widely used in medical applications, such as in cancer therapy, targeted delivery of proteins, genes and DNAs, and scaffolds in tissue regeneration. One salient feature of nanogels is their tunable responsiveness to external stimuli. In this review, thermosensitive nanogels are discussed, with a focus on moieties in their chemical structure which are responsible for thermosensitivity. These thermosensitive moieties can be classified into four groups, namely, polymers bearing amide groups, ether groups, vinyl ether groups and hydrophilic polymers bearing hydrophobic groups. These novel thermoresponsive nanogels provide effective drug delivery systems and tissue regeneration constructs for treating patients in many clinical applications, such as targeted, sustained and controlled release.

Graphical Abstract

1. Introduction

1.1. Hydrogels

Hydrogels are three-dimensional cross-linked structures based on natural or synthetic polymers. Hydrogels can be produced in different physical forms, such as slabs, macroparticles, nanoparticles and films [1,2]. Promising properties, such as high water content, biocompatibility and degradability, make hydrogels very useful for biomedical applications [3,4,5]. A salient example of biocompatible hydrogels is the injectable and temperature-sensitive poly(amino carbonate urethane) (PACU) hydrogel, which has been used as a delivery vehicle for sustained release of human growth hormone factor [6]. Similarly, the alternating hydrophilic/hydrophobic properties of poly(N-isopropylacrylamide)-co-methacrylate [P(NIPAM-co-MA] hydrogel are used for temperature sensing in biomedical applications [7]. Due to nanogels’ structural properties, hydrogels are abundantly used in drug delivery systems and fabrication of tissue scaffolds [6,8,9]. The structure of hydrogels can be modified by conjugation with appropriate ligands to improve properties such as drug entrapment, release profile and targeting [10,11,12]. In a recent study, Liao et al. presented a novel method for the preparation of multi-responsive DNA-acrylamide (DNA-AAM)-based hydrogel microcapsules [13]. Hydrogels are also used extensively in tissue engineering as more suitable materials to fabricate biodegradable scaffolds for tissue regeneration [14,15]. One important parameter which can affect the biodegradability of hydrogels is the lower critical solution temperature (LCST). As a definition, when two immiscible liquid phases appear as a result of temperature increase at the different compositions of polymer and solvent, the minimum of the coexistence curve of the phase diagram is the LCST [16]. Figure 1 represents the schematic degradation mechanism of a thermoresponsive hydrogel for drug delivery applications, which is controlled by the LCST. Below the LCST, the thermosensitive hydrogel is in the solution state but when temperature increases and it is higher than LCST, gelation occurs, and for in-vivo or in-vitro conditions under specific circumstances such as exposing to enzymes, hydrogel can degrade. Hydrogels also have shortcomings, which leads to uncertainty in their applications in medicine, which can be addressed by transforming their macro- and micro-structure to the nanoscale.

1.2. Nanostructured Materials

Nanostructured devices are a novel class of materials with many biomedical applications [17,18,19]. The preeminent property of nanostructures is their high surface-to-volume ratio, which makes these structures injectable and improves their penetration between physiological barriers in the human body. These structures enhance disease treatment with minimum side effects and toxicity [20]. Nanostructures can be prepared in different forms, like nano-films [21], nanofibers [22,23], nanoparticles [24,25], and nanogels [26], which have the potential to be used in both drug delivery systems and tissue engineering. Recent studies indicate that nanoscale structures impact biological response; thus, they can be used to modify the surface of medical implants to decrease undesired biological responses [27,28]. Bamberger et al. synthesized a polysaccharide-based nanostructure within the 100–200 nm size range, which was modified with dextran (Dex) and polyethylene glycol (PEG) and assessed for its ability to bind to immune cells. They reported that the surface modification of nanoparticles with dextran (DEXylation) enhanced targeting with a desirable immune response [29].
In drug delivery systems, nanostructures can be injected subcutaneously or intravenously to deliver the loaded drugs to the site of injury or disease with minimum cell toxicity and immune response [30]. The surface of the drug delivery system can be modified with ligands that can be detected by receptors on the surface of malignant tumours in cancer therapy [31,32,33,34]. Stimuli-responsive nanostructures have the potential to be used for targeted delivery and controlled drug release. The pH [35], temperature [36], magnetic field [37] and redox reaction [38,39] are the most commonly used environmental factors in stimuli-responsive systems. Stimuli-responsive nanocarriers have the potential to induce enhanced permeability [40,41]. In addition, targeted delivery enables selective delivery of the drug to the diseased tissue while leaving the healthy tissue unharmed [42]. Despite the numerous advantages of nanocarriers for drug delivery, there are some challenges to be tackled, including difficulty of synthesis, low stability, and the circulation time of nanocarriers in blood circulation. In some cases, toxicity to normal cells and non-biodegradability are the main deficiencies of these structures [43]. Figure 2 shows two general types of modified nanocarriers, which can be used for the targeted delivery of both hydrophilic and hydrophobic drugs.

2. Nanogels

Nanogels are three-dimensional (3D) structures that are able to swell several times their non-swollen form [44,45]. Nanogels are nanostructured hydrogels with the advantages of both nanostructured materials and hydrogels. The two main characteristics of nanogels are their small size (up to 1000 nm) and high swelling ratio or water content [46]. Due to these properties, nanogels have become an excellent platform in many medical applications, including photo-imaging [47], tissue regeneration [48], cancer therapy [49] and gene delivery [50]. This is based on their remarkable characteristics, such as their high capacity for drug entrapment and release [51], tailorable size [52], tuneable toxicity [53], high stability, controlled and sustained drug release [54], precise targeted delivery [55], and high biodegradability [56]. Nanogels can be used for drug delivery through oral [57], pulmonary [58], nasal [59], intra-ocular [60] and topical [60] pathways. There are many methods that can be used to prepare stimuli-responsive nanogels for targeted delivery. Thermosensitive [61], pH-sensitive [62,63], glucose-sensitive [64], redox-sensitive [65], and magnetic-field-sensitive [66] nanogels are applicable to the treatment of many diseases (Figure 3). Furthermore, nanogels can be tailored as dual or multi-responsive structures [42,67]. Deng et al. explored the synthesis and properties of poly (N,N-dimethyl aminoethyl methacrylate -g- Ethylene glycol) P(DMAEMA-g-EG) nanogel carriers with 190–600 nm diameters, which showed pH, ionic strength and temperature sensitivity with LCSTs of about 35 °C [68]. The objective of this review is to describe the properties of thermoresponsive nanogels, with a focus on polymeric moieties that influence the thermoresponsive behavior of nanogels in biomedical applications. Given the wide range of applications, thermoresponsive nanogels are promising for many medical uses, such as for sensors, imaging, diagnosis, treatment and gene delivery. Different types of thermosensitive generator side groups, followed by various applications of the thermoresponsive nanogels, will be presented.

Thermosensitive Nanogels

Thermosensitive nanogels are soft nanostructured materials that respond to temperature changes in the surrounding medium. Two approaches are used to prepare thermosensitive nanogels. In the first approach, thermosensitive polymer units are incorporated in the backbone or the main structure of a nanogel-forming polymer to induce thermosensitivity. In the second approach, hydrophobic moieties are attached as side groups to a hydrophilic polymer backbone to impart temperature sensitivity [69,70,71]. The LCST of the polymer decreases as the fraction of polymer units with a hydrophobic side group is increased. Therefore, the gelation temperature can be tuned by changing the degree of substitution of backbone units with hydrophobic moieties.
The mechanism responsible for thermosensitivity is the extent of the molecular interactions which could be categorized as hydrophobic or hydrophilic, depending on the free energy change of the surrounding solvent. A positive change or a negative change in the free energy of a mixture indicates its hydrophobicity or hydrophilicity, respectively [72]. Association of water molecules, as the governing interaction in the system, is the main cause of free energy change. Water molecules at temperatures lower than the LCST align well around hydrophilic parts. However, at temperatures above the LCST, owing to the hydrophobicity of the surrounding groups, water molecules start detaching, with a consequent phase separation between the water and polymers. Moreover, as the temperature increases, the alignment of water molecules collapses due to hydrophobic moieties, and the entropy of the system increases, which leads to gel formation. In other words, when the temperature is lower than the LCST, hydrophilic–hydrophilic interactions are stronger than hydrophobic–hydrophobic interactions, and this increases polymer solubility. As the temperature increases, hydrophobic–hydrophobic interactions become more important than hydrophilic–hydrophilic interactions, which eventually leads to aggregation of hydrophobic moieties and nanogel formation.

3. Thermosensitive Polymers

3.1. Polymers Bearing Amide Groups

3.1.1. PNIPAM

Poly (N-isopropyl acrylamide) (PNIPAM) is a thermosensitive polymer containing hydrophilic (C = O, NH) and hydrophobic groups (i.e., CH3). PNIPAM is synthesized by free radical polymerization. This polymer is abundantly investigated in tissue engineering and drug delivery applications [73,74,75]. Although the LCST of PNIPAM is about 32 °C, which makes this polymer an appropriate temperature-sensitive biomaterial, the non-biodegradability of PNIPAM impedes its widespread use in clinical applications. PNIPAM-based drug carriers can be modified with different functional groups for targeted drug delivery [76], controlled release [77], imaging and tracking [47], as well as other functionalities [66]. Zhou et al. investigated doxorubicin (DOX) release from a temperature-sensitive and photoluminescent hydrogel using PNIPAM and cadmium telluride quantum dots (CdTe QDs) (photoluminescent inducer) with polyacrylamide (PAA) as a crosslinker. Results demonstrated that the rate of drug release could be adjusted by external temperature [78]. Molina et al. formulated a near-infrared (NIR) absorbing nanogel based on N-isopropylacrylamide– dendritic polyglycerol–polyaniline (NIPAM-dPG-PANI) for photothermal cancer therapy (Figure 4). The size of nanogels was about 150–240 nm, and in-vitro MTT [3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide] assays on A2780 cells and in-vivo (mice) investigations were performed. In this research, the results indicated that mice could tolerate a 500 mg/kg dose of nanogels in 5 days without substantial toxicity [79]. Śliwa et al. synthesized a temperature-responsive nanogel with a hydrodynamic diameter of 150–650 nm for the controlled release of orange II. This nanogel was prepared by polymerization of 1-vinylimidazole (Vim) and PNIPAM monomers, with bisacrylamide (BAM) as the crosslinker [51].

3.1.2. PNIPMAM

Poly (N-isopropyl methacrylamide) (PNIPMAM) is another amide-bearing thermosensitive polymer that contains methyl groups attached to the α-carbon, with a higher swelling ratio in the aqueous medium compared to PNIPAM [80]. Despite having similar characteristics to PNIPAM, there are remarkable differences between the two polymers for drug delivery applications. One important difference is the higher LCST of PNIPMAM (38 °C) compared to PNIPAM (32 °C), due to the higher hydrophilicity of PNIPAM compared to PNIPMAM [81]. Cors et al. synthesized a core–shell thermosensitive nanogel based on PNIPMAM as the core and PNIPAM as the shell in order to understand the swelling and shrinking behaviour of the polymer [82]. The results of this study indicated a linear increase in swelling with temperature in the range of 25 to 35 °C. Peters et al. prepared a thermosensitive PNIPMAM-based core–shell nanogel for cancer therapy for controlled and triggered release of DOX. The cytotoxicity of the synthesized nanogels was investigated with L929 fibroblasts, and low toxicity on cells was demonstrated [83]. In Figure 5, Deshpande et al. prepared core–shell nanogels using PNIPMAM as the shell and gold nanoparticles as the core for sustained, triggered release of DOX [84].

3.1.3. PDEAAM

Poly (N,N-diethylacrylamide) (PDEAAM) is a thermosensitive polymer studied by Idziak et al. to determine the LCST of PDEAAM at different concentrations of sodium dodecyl sulphate (SDS), using ultraviolet (UV) spectroscopy and differential scanning calorimetry (DSC). They showed that PDEAAM had a sharp phase transition with an LCST of about 33 °C [85]. Different research groups have studied PDEAAM’s properties, such as enhancement of thermosensitivity by copolymerization with 2-hydroxyethyl methacrylate (HEMA) [86] and thermal responsivity of PDEAAM hydrogels prepared by γ-ray irradiation [87]. In the past decade, PDEAAM has been used for the preparation of responsive hydrogels [88,89,90], micelles [91,92] and nanogels [93,94] for biomedical applications. Lu et al. prepared non-ionic and thermosensitive nanogels with 100-nm diameters, based on N,N-diethyl acrylamide (DEA) and N,N-dimethylacrylamide (DMA), which can be used for DNA separation by microchip electrophoresis [95]. Rieger et al. synthesized thermoresponsive PEGylated micelles and nanogels to form core–shell nanostructures, with diameters of 800 and 550 nm at 15 and 70 °C, respectively [96], which could be used as drug carriers.

3.1.4. PVCL

Poly (N-vinyl caprolactam) (PVCL) is a water-soluble amphiphilic polymer, with non-ionic, thermoresponsive characteristics. Polymer molecular weight can affect LCST. Because of its biocompatibility and low cytotoxicity, PVCL is considered an ideal thermoresponsive polymer for biomedical applications [97,98,99], particularly when compared to PNIPAM [100,101,102]. PVCL conjugation with hydrophilic units such as PEG or derivatives of PEG enables the synthesis of temperature-responsive block copolymers. For temperatures above the CPT of PVCL, these block copolymers act as amphiphilic structures to form well-defined nanoscale aggregates by self-assembly pathways. Such stimuli-responsive structures, due to their ability to assemble or disassemble without using any additive, have immense potential for use in advanced drug delivery systems [103].
PVCL has also been utilized for the preparation of stimuli-responsive nanoparticles and nanofibers [37]. For instance, González et al. prepared thermoresponsive nanofibers via the electrospinning technique and investigated their use in drug delivery systems. PVCL and hydroxymethyl acrylamide were copolymerized and used to generate Rhodamine B (RhB)-loaded nanofibers with diameters in the range of 550–1200 nm. The results demonstrated that the copolymer could be used as a biosensor or as a matrix for controlled drug delivery [22]. In another study, Kehren et al. prepared polycaprolactone (PCL) microfibers modified with PVCL-based nanogel and investigated water uptake and degradability. The results demonstrated that the thermosensitivity of nanogels was preserved irrespective of whether the nanogels were in or out of the microfiber surface. Additionally, the PVCL nanogels in the structure regulated the degradability of the PVC-modified PVCL nanogels [104]. Madhusudana et al. synthesized dual-responsive nanogels by copolymerization of N-vinyl caprolactam (VCL) and acrylamidoglycolic acid (AGA) for applications in cancer therapy. The in-vitro release of the anticancer drug 5-fluorouracil (5-Fu) from VCL-AGA nanogels was influenced by both pH and temperature [105] (Figure 6).
Other previously studied thermosensitive nanogels with polymers bearing amide groups are also introduced in Table 1. Parameters such as size, interactions, thermosensitive part, therapeutic agent, and application are provided for better comparison.

3.2. Polymers Bearing Polyether Groups

PEG

Polyethylene glycol (PEG) is another important water-soluble, thermoresponsive polymer. The LCSTs of PEG-based polymers can be regulated by copolymerization with hydrophobic units [142]. Hydrophobic units like methyl and ethyl groups can regulate the polarity and temperature-responsiveness of the polymer within a physiological temperature range [143]. The composition of the monomers, molecular weight, concentration, and ionic strength of the solution considerably affect the LCST of PEG-based copolymers [144]. Xia Dong et al. synthesized a thermosensitive fluorescent nanogel using the four-arm PEG–PCL for bio-imaging applications. The results of the study demonstrated the superior capability of PEG–PCL as a drug carrier for tumour cells. Further, the PEG–PCL nanogels showed fluorescent activity in vivo while satisfying biocompatibility requirements, which made this nanogel system a suitable drug carrier for tumour-targeted delivery [138] (Figure 7).

3.3. Polymers Bearing Vinyl Ether Groups

3.3.1. PMEO2MA

Poly (2-[2-methoxyethoxy] ethyl methacrylate) (PMEO2MA) is an amphiphilic and biocompatible polymer that contains PVE functional groups (O(CH=CH2)2). The phase transition behaviour of PMEO2MA is similar to PNIPAM, which enables this polymer to be used as a thermosensitive material in biomedical applications [145]. París et al. investigated the phase transition temperature of P(MEO2MA-co-DMAEMA) hydrogel. The copolymer was synthesized via free radical polymerization and the LCSTs of hydrogels with different contents of MEO2MA were investigated in PBS solution. The results demonstrated that the hydrogel was temperature and pH sensitive and LCST of the hydrogel could be tuned by changing MEO2MA content, ionic strength or the environment pH [146]. In another study, Shen et al. prepared a core–shell thermosensitive nanogel using reversible addition–fragmentation chain transfer polymerization based on PEG as the core and oligo (ethylene glycol) (OEG) as the outer layer of nanogels. MEO2MA was introduced as a thermoresponsive moiety. The synthesized nanogels with an average diameter of 40–80 nm had negligible cytotoxicity when tested on A549 cells [147].
Biglione et al. synthesized a thermosensitive nanogel based on ethylene glycol using a facile ultra-sonication technique. Nanogels with 70 to 180 nm diameter were prepared using MEO2MA and oligo (ethylene glycol) methyl ether methacrylates (OEGMA) as temperature-responsive moieties and tetra ethylene glycol di-methacrylate (TEGDMA) as the crosslinker. Cytotoxicity and cell uptake evaluations were performed on A549 cells using RhB for labelling. The results indicate that the nanogels had appropriate cytotoxicity and cell permeation profiles [148] (Figure 8).

3.3.2. OEGMA

Oligo (ethylene glycol) methyl ether methacrylate (OEGMA) has attracted attention as a new type of thermosensitive hydrogel [149]. Similar to PNIPAM, the LCST transition of OEGMA-based hydrogels is not very sensitive to external conditions. Therefore, ionic strength, concentration, and pH do not have a significant impact on the LCST transition of poly (OligoPOEGMA) [150]. Moreover, POEGMA polymers demonstrate exciting characteristics, including high anti-folding, nontoxicity, limited hysteresis, as well as adjustable temperature sensitivity [151]. Lutz et al. synthesized copolymers of MEO2MA and OEGMA via atom transfer radical polymerization and observed possible control of LCST between 26 and 90 °C by altering the monomer compositions [152]. Consequently, a large number of different types of polymers [153,154,155], micelles [156,157], vesicles [158], micro/nanogels [159], and smart POEGMA-based hydrogels [160] have been synthesized and investigated.
OEGMA-based thermoresponsive materials are widely used as drug carrier hydrogels and nanogels [161,162]. Cortes et al. prepared a thermosensitive and magnetic-responsive nanogel for intracellular remote release of DOX. The prepared nanogels had a diameter from 320 to 460 nm, and their LCST was about 47 °C, which was appropriate for a thermal, magnetic hyperthermia strategy. It was also demonstrated that DOX release from the nanogels increased by the application of an alternating magnetic field [163] (Figure 9). When a high-frequency magnetic field is applied to magnetic nanoparticles (MNPs), they can generate heat, which is useful for hyperthermia treatment and acts as driving force for drug release [164]. The thermosensitive structures were used as chemical sensors and indicators. For instance, Liu and et al. synthesized OEG-based thermoresponsive, comb-like polymers via free radical polymerization, which was used as a temperature and pH-responsive sensor [165].
Previously studied thermosensitive nanogels with polymer-bearing vinyl ether groups are also shown in Table 2. Parameters such as size, interactions, thermosensitive part, therapeutic agent and application are all summarized.

3.4. Hydrophilic Polymers Bearing Hydrophobic Groups

The second approach to developing thermosensitive polymers is using hydrophobic moieties/polymers alongside hydrophilic materials. The most widely used hydrophilic materials are polysaccharides due to their biocompatibility. Different hydrophobic materials are used to conjugate on hydrophilic units to form thermosensitive materials. Many studies have been performed based on this approach, and different hydrophobic materials, such as cholesterol [184,185], poly L-lactide (PLLA) [186,187,188,189], beta glycerophosphate (β-GP) [190,191] and pluronic F127 (F127) [192,193] have been used to synthesize thermosensitive hydrogels and nanogels for biomedical applications.

3.4.1. Cholesterol-Bearing Polymers

Cholesterol is an organic and hydrophobic molecule that exists in the mammalian body (component of the plasma membrane) and helps to make hormones, vitamin D, and compounds that aid in food digestion. In a few studies, cholesterol was used with pullulan (Plu) for nanogel formation and self-assembly of micelles [194,195,196]. Thara et al. prepared a self-assembled thermosensitive nanogel using cholesterol as a thermosensitive agent bearing hydroxypropyl cellulose (HP-Clu) with LCST of 50 °C. The diameter of nanogels was about 100 nm and 1500 nm at 37 °C and 60 °C in PBS, respectively [185]. In another study, Fujioka et al. synthesized cholesterol-bearing Plu nanogel for the delivery of bone morphogenetic protein-2 (BMP2) and Fibroblast growth factor-18 (FGF18) delivery. The results showed that the delivery of two proteins by the cholesterol-based nanogels aided in the regeneration of bone in vivo in a mouse model [197]. The protein exchange reaction, such as serum albumin with trapped BMP2 and FGF18 molecules, causes growth factor release over 8 weeks to maintain the BMP2 concentration at a certain level around the bone defects in the in-vivo condition. The schematic synthesis followed by growth factor delivery is depicted in Figure 10.

3.4.2. PLLA-Bearing Polymers

Mechanical strength and the favourable degradation rate of aliphatic polyesters, such as PLLA, poly (lactide-glycolic acid) (PLGA), and PCL, make these polymers very attractive for the preparation of thermoresponsive nanogels. These hydrophobic polymers are mostly used in tissue engineering for constructing biodegradable scaffolds. However, mass transport of oxygen, nutrients and growth factors in such scaffolds is poor, and cell adhesion to the scaffold surface due to the hydrophobicity of polyesters is poor. Na et al. synthesized poly (l-lactic acid)/poly (ethylene glycol) and alternating multi-block temperature-responsive nanoparticles for anticancer drug delivery. The cytotoxicity of the nanoparticles was investigated with Lewis lung carcinoma (LLC) cells, and the results indicated that cell toxicity was temperature dependent and increased with increasing temperature from 37 °C to 42 °C [188]. Nagahama et al. prepared lysozyme-loaded Dex-g-PLLA nanogels and investigated the effect of the hydrophobic unit on the sustained release of lysozyme by comparing the release from Dex-g-PLLA conjugate with dextran-cholesterol. The synthesized Dex-g-PLLA nanogels had low critical aggregation concentrations. The results indicated that the Dex-g-PLLA nanogel has a high potential for protein delivery with sustained release of lysozyme for one week without denaturation [187]. Kyo et al. prepared Plu-g-PLLA nanogels with 150–800 nm diameters for sustained release of DOX. The PLLA in Plu-g-PLLA serves to induce self-assembly of nanogel and improves the loading of hydrophobic drugs [71]. In a recent study, Jung et al. developed Plu-g-PLLA nanogels with succinic anhydride (SA) to deliver lysozyme as a protein drug. The average diameters of the thermosensitive nanogels were 190 nm and 540 nm at 4 °C and 37 °C, respectively. In-vivo studies in nude mice indicated the sustained release of the drug [198].

3.4.3. PLLA Bearing Polymers

Pluronic F127 is a hydrophilic non-ionic copolymer, based on non-toxic FDA-approved PEG and polypropylene glycol (PPG) segments, which is widely used in biomedical applications [192,199,200,201,202,203]. Sharma et al. developed a thermosensitive nanogel based on pluronic F127 as the carrier for the delivery of lidocaine (Lid) and prilocaine (Pl) and demonstrated by in-vitro and in-vivo experiments that the thermosensitivity of the carrier improves the delivery of Lid and PI [193]. Choi et al. prepared a nano-sponge based on heparin (Hep) and pluronic F127 and used it as a thermosensitive carrier for the controlled release of growth factors (bFGF, VEGF, BMP-2 and HGF) [204].
Previously studied thermosensitive nanogels with hydrophilic polymers bearing hydrophobic groups are shown in Table 3. Parameters such as size, interactions, thermosensitive part, therapeutic agent, and application are all summarized.

4. Conclusions

This review describes the synthesis and applications of thermoresponsive nanogels for targeted and controlled drug delivery. Thermoresponsive nanogels are discussed based on their thermally sensitive polymeric moieties. NIPAM is one of the most studied thermosensitive polymers that have been frequently used to prepare nanoparticles, hydrogels and nanogels for biomedical applications. However, non-biodegradability is limiting the use of NIPAM in clinical applications. Researchers are developing other thermoresponsive materials that are biodegradable for clinical applications while also possessing the rapid and sharp LCST of NIPAM. Materials that possess both hydrophobic and hydrophilic moieties in their molecular structure can induce nanogel formation and thermosensitivity. Based on the stated characteristics of thermosensitive nanogels, and from the authors’ perspective, thermosensitive polymers can be divided into four groups, including those bearing amide, ether and vinyl ether and hydrophilic polymers bearing hydrophobic groups. Generally, the above-mentioned thermosensitive polymers are conjugated to polysaccharides for augmenting biocompatibility as well as other desirable properties. As an alternative approach, hydrophilic polymers can be combined with hydrophobic materials such as PLLA and cholesterol to form thermosensitive nanogels. Four groups of thermosensitive materials were covered in this review, and some of the materials that are used in the synthesis of thermosensitive nanogels were presented. Multi-responsive nanogels, especially those with thermosensitive functionality, are commonly used in a vast number of biomedical applications, including cancer therapy, targeted delivery and in-situ gelation for drug release and entrapment; therefore, thermosensitive nanogels, with their invaluable functions, will become even more remarkable and important structures for drug delivery and tissue engineering applications in foreseeable future.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Abbreviations.
Table A1. Abbreviations.
ComponentAbbreviationComponentAbbreviation
(2-acetoacetoxyethyl) methacrylateAAEMA1,3-bis(carboxyphenoxy) propaneCPP
1-vinylimidazoleVim2-(2-methoxyethoxy) ethyl meth acrylate)MeO2MA
2-(5,5-dimethyl-1,3-dioxan-2-yloxy) ethyl acrylateDMDEA2-(acetylthio) ethyl methacrylateAcSEMA
2,2-bis(2-oxazoline)BOX2-aminoethyl methacrylamide hydrochlorideAEMA
2-dimethyl(aminoethyl) methacrylateDMAEM2-dimethylmaleinimido ethylacrylamideDMIAAm
2-hydroxyethyl methacrylateHEMA2-lactobionamidoethyl methacrylamideLAEMAm
2-methacryloyloxyethyl acrylateMEA2-methoxyethyl acrylateMEA
3-(trimethoxysilyl)propyl methacrylate)MPMA3-{[(2R)-2-(octadecylamino)-3-phenyl propanoyl]amino}butyrateTEAB
3-acrylamidophenylboronic acidAAPBA3-gluconamidopropyl methacrylamideGAPMA
4-(2-acryloylaminoethylamino)-7-nitro-2,1,3-benzoxadiazoleNBDAA4-acrylamidofluoresceinAFA
5-fluorouracil5-Fu6-O-vinyladipoyl-D-galactoseODGal
AcetamidophenolAPAcrylamideAAM
Acrylamidoglycolic acidAGAAcrylic acidAA
AlginateAlgAtom transfer radical polymerizationATRP
Basic fibroblast growth factorbFGFBeta glactosidaseBG
Beta glycerophosphateβ-GPBevacizumabBz
Bis (2-acryloyloxyethyl) disulfideBADSBisacrylamideBAM
Bismuth (III) oxideBi2O3Bone morphogenetic protein 2BMP-2
Bovine serum albuminBSABupivacaineBV
Butyl methacrylateBMAButyl methylacrylatePIB
Butyl methylacrylateBMACarboxymethyl cellulose sodium saltsCMC
Carboxymethyl hexanoyl chitosanCHChSCelluloseClu
ChitosanCHSCholesterolChol
Chondroitin sulfateCSCisplatinCIS
Cloud point temperatureCPTCoumarin 102C 102
Covinyl pyrrolidoneCVPCurcuminCur
Cytochrome CCyt CDendritic polyglyceroldPG
Deoxyribonucleic acid-acrylamideDNA-AAMDextranDex
Dextran methacrylatesDex-MADi(ethylene glycol) methyl ethyl methacrylateDEGMA
Diacetone acrylamideDAAMDibucaineDc
DiclofenacDfDimethyl maleinimido acrylamideDMIAAM
DoxorubicinDoxEthosuximideESM
Ethylene glycol dimethacrylateEGDMAEthylene glycol dimethacrylateEGDMA
Ethylene glycoleEGFibrinogenFib
Fibroblast growth factor 18FGF18Food and Drug AdministrationFDA
Gelatin type AGel AGlutaraldehydeGA
Graphene oxideGOHeparinHep
Hollow gold nanoparticleHGNPHuman growth hormonehGH
Human serum albuminHASHyaluronic acidHA
Hydroxymethyl acrylamideHMAAHydroxypropyl celluloseHP-Clu
IndomethacinImcIodoazomycin ArabinofuranosideIAZA
Itaconic acidIALewis lung carcinomaLLC
LidocaineLidLower critical aggregation concentrationLCAC
Lower critical solution temperatureLCSTL-ProlineL-Pro
Maleic acidMAMaleimide dithiolMDT
Malloapelta BMall BMegestrol acetateMeg
MelatoninMtMesoporous silicamSiO2
MethacrylateMAMethacrylic AcidMAA
MethotrexateMTXMethoxy-poly(ethylene glycol)Met-PEG
Monomethoxy poly(ethylene glycol)mPEGMusconeMc
N, N-di ethylacrylamideDEAAMN,N′-methylenebis(acrylamide)MBAM
N,N-diethylacrylamideDEAN,N-dimethylacrylamideDMA
N,N-dimethylaminoethyl methacrylateDMAEMAN-acryloyl-3-aminophenylboronic acidAPBA
N-acryloylglycinamideNAGANaphthalimide-based dyeNPTUA
Nile redNRNitrobenzoxadiazoleNDB
NitrobenzoxadiazoleNBDN-methylolacrylamideNMA
N-tert-butyl acrylamideNTBAN-vinylcaprolactamNVCL
N-vinylformamideVFAN-vinylpyrrolidoneVP
Oligo (ethylene glycol)OEGOligo (ethylene glycol) methacrylatesOEGMA
Oligo (ethylene glycol)methyl ether methacrylateMEO5MAOligo (ethylene oxide) monomethyl ether methacrylateOEOMA
Oligo (L-lactide)OLAPaclitaxelPTX
Phenylboronic acidPBAPhenylethynesulfonamidePES
Phosphate-buffered salinePBSPhotochromic spiropyranSP
Pluronic F127F127Poloxamer 407P407
Poly (2-(2-methoxyethoxy) ethyl meth acrylate))PMEO2MAPoly (2-(2-methoxyethoxy)ethyl methacrylate)PMEO2MA
Poly (2-(diethylamino)ethyl) methacrylatePDEAEMAPoly (2-aminoethyl methacrylamide hydrochloride)PAEMA
Poly (2-Ethoxy-2-oxo-1,3,2-dioxaphospholane)PEEPPoly (2-isopropyl-2-oxazoline)piPOz
Poly (2-methacryloyloxyethyl phosphorylcholine)PMPCpoly (2-methoxyethyl acrylate)PMEA
Poly (2-methylthioethyl glycidyl ether)PMTEGEPoly (3,4-ethylenedioxythiophene)PEDOT
Poly (acrylamide)PAMPoly (acrylonitrile)PAN
Poly (amino carbonate urethane)PACUPoly (caprolactone)PCL
Poly (ether)PEPoly (ethyl glycidyl ether)PEGE
Poly (ethylene glycol dimethacrylate)PEGDMAPoly (ethylene glycol) diacrylatePEGDA
Poly (ethylene glycol) methacrylatesPEGMAPoly (ethylene glycol) methyl ether acrylatePEGMEA
Poly (ethylene glycole)PEGPoly (ethylene oxide)PEO
Poly (glycidol)PGLPoly (glycidyl methyl ether)PGME
Poly (lactide co-glycoside)PLLA-co-GSPoly (lactide-glycolic acid)PLGA
Poly (L-alanine)PLAPoly (L-aspartic acid)P(L-Asp)
Poly (L-lactide)PLLAPoly (L-lysine)PLL
Poly (methacrylic acid)PMAPoly (methoxydiethylene glycol methacrylate)PMEODEGM
Poly (methyl glycidyl ether)PGMEPoly (N, N-di ethylacrylamide)PDEAAM
Poly (N,N-dimethylacrylamide)PDMAPoly (N-acryloyl-2,2-dimethyl-1,3-oxazolidinePADMO
Poly (N-isopropyl meth acryl amide)PNIPMAMPoly (N-isopropylacrylamide)PNIPAM
Poly (N-n-propylacrylamide)PNNPAMPoly (N-vinyl caprolactam)PVCL
Poly (oligo (ethylene glycol) methacrylates)POEGMAPoly (organo phosphazene)POP
Poly (phenylboronate ester) acrylatePPBDEMAPoly (propylene oxide)PPO
Poly (sodium 2-acrylamido-2-methylpropanesulfonate)PAMPSPoly (sodium styrenesulfonate)PSSNa
Poly (urethane)PUPoly (vinyl ether)PVE
Poly [(3-acrylamidopropyl)-trimethylammonium chloride]PAMPTMAPoly acrylamidePAA
Poly ethyleniminePEIPoly Oligo (ethylene oxide) methyl ether methacrylatePOEOMA
Poly propylene glycolPPGPoly tetra (ethylene glycol) diacrylatePTEGDA
Polyacrylic acidPAAPolyamidoaminePAMAM
PolyanilinePANIPolyglycerolPG
Polyvinyl alcoholPVAPorphyrinPor
PrednisonePnPrilocainePl
PropranololPplPropyl acrylic acidPAA
ProtaminePtProtamine sulfatePS
PullulanPluQuantum dotsQDs
Rhodamine BRhBRicin ARA
Salicylic acidSCASebacic acidSA
Sodium 2-acrylamido-2-methylpropane sulfonateAMPSSodium alginateSA
spiropyranSPStyreneST
Succinic anhydrideSASuccinylated pullulanS-Plu
TemozolomideTZTetraethylene glycol dimethacrylateTEGDMA
Tween 80T80Vascular endothelial growth factorVEGF

References

  1. Hoare, T.R.; Kohane, D.S. Hydrogels in drug delivery: Progress and challenges. Polymer 2008, 49, 1993–2007. [Google Scholar] [CrossRef] [Green Version]
  2. Sarwan, T.; Kumar, P.; Choonara, Y.E.; Pillay, V. Hybrid thermo-responsive polymer systems and their biomedical applications. Front. Mater. 2020, 7, 73. [Google Scholar] [CrossRef]
  3. Fundueanu, G.; Constantin, M.; Bucatariu, S.; Ascenzi, P. Poly(N-isopropylacrylamide-co-N-isopropylmethacrylamide) Thermo-Responsive Microgels as Self-Regulated Drug Delivery System. Macromol. Chem. Phys. 2016, 217, 2525–2533. [Google Scholar] [CrossRef]
  4. Barati, D.; Shariati, S.R.P.; Moeinzadeh, S.; Melero-Martin, J.M.; Khademhosseini, A.; Jabbari, E. Spatiotemporal release of BMP-2 and VEGF enhances osteogenic and vasculogenic differentiation of human mesenchymal stem cells and endothelial colony-forming cells co-encapsulated in a patterned hydrogel. J. Control. Release 2016, 223, 126–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sarvestani, A.S.; Xu, W.; He, X.; Jabbari, E. Gelation and degradation characteristics of in situ photo-crosslinked poly(L-lactide-co-ethylene oxide-co-fumarate) hydrogels. Polymer 2007, 48, 7113–7120. [Google Scholar] [CrossRef]
  6. Phan, V.H.G.; Thambi, T.; Duong, H.T.T.; Lee, D.S. Poly(amino carbonate urethane)-based biodegradable, temperature and pH-sensitive injectable hydrogels for sustained human growth hormone delivery. Sci. Rep. 2016, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
  7. Fundueanu, G.; Constantin, M.; Bucatariu, S.; Ascenzi, P. pH/thermo-responsive poly(N-isopropylacrylamide-co-maleic acid) hydrogel with a sensor and an actuator for biomedical applications. Polymer 2017, 110, 177–186. [Google Scholar] [CrossRef]
  8. Sato, Y.; Yamamoto, K.; Horiguchi, S.; Tahara, Y.; Nakai, K.; Kotani, S.; Oseko, F.; Pezzotti, G.; Yamamoto, T.; Kishida, T.; et al. Nanogel tectonic porous 3D scaffold for direct reprogramming fibroblasts into osteoblasts and bone regeneration. Sci. Rep. 2018, 8, 15824. [Google Scholar] [CrossRef] [Green Version]
  9. Han, L.H.; Suri, S.; Schmidt, C.E.; Chen, S. Fabrication of three-dimensional scaffolds for heterogeneous tissue engineering. Biomed. Microdevices 2010, 12, 721–725. [Google Scholar] [CrossRef]
  10. Shi, L.; Wang, F.; Zhu, W.; Xu, Z.; Fuchs, S.; Hilborn, J.; Zhu, L.; Ma, Q.; Wang, Y.; Weng, X.; et al. Self-Healing Silk Fibroin-Based Hydrogel for Bone Regeneration: Dynamic Metal-Ligand Self-Assembly Approach. Adv. Funct. Mater. 2017, 27, 1–14. [Google Scholar] [CrossRef]
  11. Vilaça, H.; Castro, T.; Costa, F.M.G.; Melle-Franco, M.; Hilliou, L.; Hamley, I.W.; Castanheira, E.M.S.; Martins, J.A.; Ferreira, P.M.T. Self-assembled RGD dehydropeptide hydrogels for drug delivery applications. J. Mater. Chem. B 2017, 5, 8607–8617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Arsalani, N.; Kazeminava, F.; Akbari, A.; Hamishehkar, H.; Jabbari, E.; Kafil, H.S. Synthesis of polyhedral oligomeric silsesquioxane nano-crosslinked poly(ethylene glycol)-based hybrid hydrogels for drug delivery and antibacterial activity. Polym. Int. 2019, 68, 667–674. [Google Scholar] [CrossRef]
  13. Liao, W.C.; Lilienthal, S.; Kahn, J.S.; Riutin, M.; Sohn, Y.S.; Nechushtai, R.; Willner, I. PH-and ligand-induced release of loads from DNA-acrylamide hydrogel microcapsules. Chem. Sci. 2017, 8, 3362–3373. [Google Scholar] [CrossRef] [Green Version]
  14. Fan, C.; Wang, D.A. A biodegradable PEG-based micro-cavitary hydrogel as scaffold for cartilage tissue engineering. Eur. Polym. J. 2015, 72, 651–660. [Google Scholar] [CrossRef]
  15. Kim, Y.S.; Cho, K.; Lee, H.J.; Chang, S.; Lee, H.; Kim, J.H.; Koh, W.G. Highly conductive and hydrated PEG-based hydrogels for the potential application of a tissue engineering scaffold. React. Funct. Polym. 2016, 109, 15–22. [Google Scholar] [CrossRef]
  16. Zhang, Q.; Weber, C.; Schubert, U.S.; Hoogenboom, R. Thermoresponsive polymers with lower critical solution temperature: From fundamental aspects and measuring techniques to recommended turbidimetry conditions. Mater. Horiz. 2017, 4, 109–116. [Google Scholar] [CrossRef]
  17. Dadashi, S.; Boddohi, S.; Soleimani, N. Preparation, characterization, and antibacterial effect of doxycycline loaded kefiran nanofibers. J. Drug Deliv. Sci. Technol. 2019, 52, 979–985. [Google Scholar] [CrossRef]
  18. Molaei, M.J. A review on nanostructured carbon quantum dots and their applications in biotechnology, sensors, and chemiluminescence. Talanta 2019, 196, 456–478. [Google Scholar] [CrossRef] [PubMed]
  19. Zong, H.; Xia, X.; Liang, Y.; Dai, S.; Alsaedi, A.; Hayat, T.; Kong, F.; Pan, J.H. Designing function-oriented artificial nanomaterials and membranes via electrospinning and electrospraying techniques. Mater. Sci. Eng. C 2018, 92, 1075–1091. [Google Scholar] [CrossRef] [PubMed]
  20. Aderibigbe, B.; Naki, T. Design and Efficacy of Nanogels Formulations for Intranasal Administration. Molecules 2018, 23, 1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Park, S.; Choi, D.; Jeong, H.; Heo, J.; Hong, J. Drug Loading and Release Behavior Depending on the Induced Porosity of Chitosan/Cellulose Multilayer Nanofilms. Mol. Pharm. 2017, 14, 3322–3330. [Google Scholar] [CrossRef] [PubMed]
  22. González, E.; Frey, M.W. Synthesis, characterization and electrospinning of poly (vinyl caprolactam-co-hydroxymethyl acrylamide) to create stimuli-responsive nanofibers. Polymer 2017, 108, 154–162. [Google Scholar] [CrossRef] [Green Version]
  23. Shariati, S.R.P.; Moeinzadeh, S.; Jabbari, E. Nanofiber Based Matrices for Chondrogenic Differentiation of Stem Cells. J. Nanosci. Nanotechnol. 2016, 16, 8966–8977. [Google Scholar] [CrossRef]
  24. Jabbari, E.; Yang, X.; Moeinzadeh, S.; He, X. Drug release kinetics, cell uptake, and tumor toxicity of hybrid VVVVVVKK peptide-assembled polylactide nanoparticles. Eur. J. Pharm. Biopharm. 2013, 84, 49–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bordat, A.; Boissenot, T.; Nicolas, J.; Tsapis, N. Thermoresponsive polymer nanocarriers for biomedical applications. Adv. Drug Deliv. Rev. 2019, 138, 167–192. [Google Scholar] [CrossRef]
  26. Wang, H.; Di, J.; Sun, Y.; Fu, J.; Wei, Z.; Matsui, H.; Del, C.; Alonso, A.; Zhou, S. Biocompatible PEG-Chitosan@Carbon Dots Hybrid Nanogels for Two-Photon Fluorescence Imaging, Near-Infrared Light/pH Dual-Responsive Drug Carrier, and Synergistic Therapy. Adv. Funct. Mater. 2015, 25, 5537–5547. [Google Scholar] [CrossRef]
  27. Boddohi, S.; Moore, N.; Johnson, P.A.; Kipper, M.J. Polysaccharide-Based Polyelectrolyte Complex Nanoparticles from Chitosan, Heparin, and Hyaluronan. Biomacromolecules 2009, 10, 1402–1409. [Google Scholar] [CrossRef]
  28. Hussain, M.; Xie, J.; Hou, Z.; Shezad, K.; Xu, J.; Wang, K.; Gao, Y.; Shen, L.; Zhu, J. Regulation of Drug Release by Tuning Surface Textures of Biodegradable Polymer Microparticles. ACS Appl. Mater. Interfaces 2017, 9, 14391–14400. [Google Scholar] [CrossRef]
  29. Bamberger, D.; Hobernik, D.; Konhäuser, M.; Bros, M.; Wich, P.R. Surface Modification of Polysaccharide-Based Nanoparticles with PEG and Dextran and the Effects on Immune Cell Binding and Stimulatory Characteristics. Mol. Pharm. 2017, 14, 4403–4416. [Google Scholar] [CrossRef]
  30. Soni, S.; Babbar, A.K.; Sharma, R.K.; Maitra, A. Delivery of hydrophobised 5-fluorouracil derivative to brain tissue through intravenous route using surface modified nanogels. J. Drug Target. 2006, 14, 87–95. [Google Scholar] [CrossRef]
  31. Murphy, E.A.; Majeti, B.K.; Mukthavaram, R.; Acevedo, L.M.; Barnes, L.A.; Cheresh, D.A. Targeted Nanogels: A Versatile Platform for Drug Delivery to Tumors. Mol. Cancer Ther. 2011, 10, 972–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ryu, J.H.; Bickerton, S.; Zhuang, J.; Thayumanavan, S. Ligand-decorated nanogels: Fast one-pot synthesis and cellular targeting. Biomacromolecules 2012, 13, 1515–1522. [Google Scholar] [CrossRef] [Green Version]
  33. Adamo, G.; Grimaldi, N.; Campora, S.; Bulone, D.; Bondì, M.L.; Al-Sheikhly, M.; Sabatino, M.A.; Dispenza, C.; Ghersi, G. Multi-functional nanogels for tumor targeting and redox-sensitive drug and siRNA delivery. Molecules 2016, 21, 1594. [Google Scholar] [CrossRef] [PubMed]
  34. Multi-Functional Nanogels for Tumor Targeting and Redox-Sensitive Drug and siRNA Delivery—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/27886088/ (accessed on 23 May 2020).
  35. Li, Y.; Bui, Q.N.; Duy, L.T.M.; Yang, H.Y.; Lee, D.S. One-Step Preparation of pH-Responsive Polymeric Nanogels as Intelligent Drug Delivery Systems for Tumor Therapy. Biomacromolecules 2018, 19, 2062–2071. [Google Scholar] [CrossRef] [PubMed]
  36. Zuo, Y.; Jiao, Z.; Ma, L.; Song, P.; Wang, R.; Xiong, Y. Hydrogen bonding induced UCST phase transition of poly(ionic liquid)-based nanogels. Polymer 2016, 98, 287–293. [Google Scholar] [CrossRef]
  37. Indulekha, S.; Arunkumar, P.; Bahadur, D.; Srivastava, R. Dual responsive magnetic composite nanogels for thermo-chemotherapy. Colloids Surf. B Biointerfaces 2017, 155, 304–313. [Google Scholar] [CrossRef]
  38. Miao, C.; Li, F.; Zuo, Y.; Wang, R.; Xiong, Y. Novel redox-responsive nanogels based on poly(ionic liquid)s for the triggered loading and release of cargos. RSC Adv. 2016, 6, 3013–3019. [Google Scholar] [CrossRef]
  39. Gao, Y.; Dong, C.M. Quadruple thermo-photo-redox-responsive random copolypeptide nanogel and hydrogel. Chin. Chem. Lett. 2018, 29, 927–930. [Google Scholar] [CrossRef]
  40. Lee, J.-Y.; Chung, S.-J.; Cho, H.-J.; Kim, D.-D. Bile acid-conjugated chondroitin sulfate A-based nanoparticles for tumor-targeted anticancer drug delivery. Eur. J. Pharm. Biopharm. 2015, 94, 532–541. [Google Scholar] [CrossRef]
  41. Schmid, D.; Park, C.G.; Hartl, C.A.; Subedi, N.; Cartwright, A.N.; Puerto, R.B.; Zheng, Y.; Maiarana, J.; Freeman, G.J.; Wucherpfennig, K.W.; et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 2017, 8, 1–11. [Google Scholar] [CrossRef]
  42. Zhang, Q.; Colazo, J.; Berg, D.; Mugo, S.M.; Serpe, M.J. Multiresponsive Nanogels for Targeted Anticancer Drug Delivery. Mol. Pharm. 2017, 14, 2624–2628. [Google Scholar] [CrossRef]
  43. Tran, N.; Webster, T.J. Magnetic nanoparticles: Biomedical applications and challenges. J. Mater. Chem. 2010, 20, 8760–8767. [Google Scholar] [CrossRef]
  44. Marek, S.R.; Conn, C.A.; Peppas, N.A. Cationic nanogels based on diethylaminoethyl methacrylate. Polymer 2010, 51, 1237–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Yin, Y.; Hu, B.; Yuan, X.; Cai, L.; Gao, H.; Yang, Q. Nanogel: A Versatile Nano-Delivery System for Biomedical Applications. Pharmaceutics 2020, 12, 290. [Google Scholar] [CrossRef] [Green Version]
  46. Vinogradov, S.V. Nanogels in the race for drug delivery. Nanomedicine 2010, 5, 165–168. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, H.; Ke, F.; Mararenko, A.; Wei, Z.; Banerjee, P.; Zhou, S. Responsive polymer-fluorescent carbon nanoparticle hybrid nanogels for optical temperature sensing, near-infrared light-responsive drug release, and tumor cell imaging. Nanoscale 2014, 6, 7443–7452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Park, K.M.; Son, J.Y.; Choi, J.H.; Kim, I.G.; Lee, Y.; Lee, J.Y.; Park, K.D. Material for the Treatment of Urinary Incontinence. Biomacromolecules 2014, 15, 1979–1984. [Google Scholar] [CrossRef] [PubMed]
  49. Chang, R.; Tsai, W.-B. Fabrication of Photothermo-Responsive Drug-Loaded Nanogel for Synergetic Cancer Therapy. Polymers 2018, 10, 1098. [Google Scholar] [CrossRef] [Green Version]
  50. Song, L.; Liang, X.; Yang, S.; Wang, N.; He, T.; Wang, Y.; Zhang, L.; Wu, Q.; Gong, C. Novel polyethyleneimine-R8-heparin nanogel for high-efficiency gene delivery in vitro and in vivo. Drug Deliv. 2018, 25, 122–131. [Google Scholar] [CrossRef] [Green Version]
  51. Śliwa, T.; Jarzębski, M.; Andrzejewska, E.; Szafran, M.; Gapiński, J. Uptake and controlled release of a dye from thermo-sensitive polymer P(NIPAM-co-Vim). React. Funct. Polym. 2017, 115, 102–108. [Google Scholar] [CrossRef]
  52. Li, Y.; Xiao, K.; Luo, J.; Lee, J.; Pan, S.; Lam, K.S. A novel size-tunable nanocarrier system for targeted anticancer drug delivery. J. Control. Release 2010, 144, 314–323. [Google Scholar] [CrossRef] [Green Version]
  53. Zhou, T.; Xiao, C.; Fan, J.; Chen, S.; Shen, J.; Wu, W.; Zhou, S. A nanogel of on-site tunable pH-response for efficient anticancer drug delivery. Acta Biomater. 2013, 9, 4546–4557. [Google Scholar] [CrossRef]
  54. Bhuchar, N.; Sunasee, R.; Ishihara, K.; Thundat, T.; Narain, R. Degradable thermoresponsive nanogels for protein encapsulation and controlled release. Bioconjug. Chem. 2012, 23, 75–83. [Google Scholar] [CrossRef]
  55. Tan, J.P.K.; Tan, M.B.H.; Tam, M.K.C. Application of nanogel systems in the administration of local anesthetics. Local Reg. Anesth. 2010, 3, 93–100. [Google Scholar] [CrossRef] [Green Version]
  56. Kohli, E.; Han, H.Y.; Zeman, A.D.; Vinogradov, S.V. Formulations of biodegradable Nanogel carriers with 5′-triphosphates of nucleoside analogs that display a reduced cytotoxicity and enhanced drug activity. J. Control. Release 2007, 121, 19–27. [Google Scholar] [CrossRef] [Green Version]
  57. Wang, X.; Cheng, D.; Liu, L.; Li, X. Development of poly(hydroxyethyl methacrylate) nanogel for effective oral insulin delivery. Pharm. Dev. Technol. 2018, 23, 351–357. [Google Scholar] [CrossRef]
  58. De Backer, L.; Braeckmans, K.; Stuart, M.C.A.; Demeester, J.; De Smedt, S.C.; Raemdonck, K. Bio-inspired pulmonary surfactant-modified nanogels: A promising siRNA delivery system. J. Control. Release 2015, 206, 177–186. [Google Scholar] [CrossRef] [Green Version]
  59. Yukia, Y.; Nochi, T.; Kong, I.G.; Takahashi, H.; Sawada, S.I.; Akiyoshi, K.; Kiyono, H. Nanogel-based antigen-delivery system for nasal vaccines. Biotechnol. Genet. Eng. Rev. 2013, 29, 61–72. [Google Scholar] [CrossRef]
  60. Novel Inulin-Based Mucoadhesive Micelles Loaded With Corticosteroids as Potential Transcorneal Permeation Enhancers—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/28512019/ (accessed on 23 May 2020).
  61. Asghar, K.; Qasim, M.; Dharmapuri, G.; Das, D. Investigation on a smart nanocarrier with a mesoporous magnetic core and thermo-responsive shell for co-delivery of doxorubicin and curcumin: A new approach towards combination therapy of cancer. RSC Adv. 2017, 7, 28802–28818. [Google Scholar] [CrossRef] [Green Version]
  62. Luan, S.; Zhu, Y.; Wu, X.; Wang, Y.; Liang, F.; Song, S. Hyaluronic-Acid-Based pH-Sensitive Nanogels for Tumor-Targeted Drug Delivery. ACS Biomater. Sci. Eng. 2017, 3, 2410–2419. [Google Scholar] [CrossRef]
  63. Mohtashamian, S.; Boddohi, S.; Hosseinkhani, S. Preparation and optimization of self-assembled chondroitin sulfate-nisin nanogel based on quality by design concept. Int. J. Biol. Macromol. 2018, 107, 2730–2739. [Google Scholar] [CrossRef]
  64. Wu, Z.; Zhang, X.; Guo, H.; Li, C.; Yu, D. An injectable and glucose-sensitive nanogel for controlled insulin release. J. Mater. Chem. 2012, 22, 22788–22796. [Google Scholar] [CrossRef]
  65. Abandansari, H.S.; Abuali, M.; Nabid, M.R.; Niknejad, H. Enhance chemotherapy efficacy and minimize anticancer drug side effects by using reversibly pH- and redox-responsive cross-linked unimolecular micelles. Polymer 2017, 116, 16–26. [Google Scholar] [CrossRef]
  66. Pikabea, A.; Ramos, J.; Papachristos, N.; Stamopoulos, D.; Forcada, J. Synthesis and characterization of PDEAEMA-based magneto-nanogels: Preliminary results on the biocompatibility with cells of human peripheral blood. J. Polym. Sci. Part Polym. Chem. 2016, 54, 1479–1494. [Google Scholar] [CrossRef]
  67. Chen, S.; Bian, Q.; Wang, P.; Zheng, X.; Lv, L.; Dang, Z.; Wang, G. Photo, pH and redox multi-responsive nanogels for drug delivery and fluorescence cell imaging. Polym. Chem. 2017, 8, 6150–6157. [Google Scholar] [CrossRef]
  68. Deng, L.; Zhai, Y.; Lin, X.; Jin, F.; He, X.; Dong, A. Investigation on properties of re-dispersible cationic hydrogel nanoparticles. Eur. Polym. J. 2008, 44, 978–986. [Google Scholar] [CrossRef]
  69. Dong, H.; Xu, Q.; Li, Y.; Mo, S.; Cai, S.; Liu, L. The synthesis of biodegradable graft copolymer cellulose-graft-poly(L-lactide) and the study of its controlled drug release. Colloids Surf. B Biointerfaces 2008, 66, 26–33. [Google Scholar] [CrossRef]
  70. Guo, Y.; Wang, X.; Shu, X.; Shen, Z.; Sun, R.-C. Self-Assembly and Paclitaxel Loading Capacity of Cellulose-graft-poly(lactide) Nanomicelles. J. Agric. Food Chem. 2012, 60, 3900–3908. [Google Scholar] [CrossRef]
  71. Cho, J.-K.; Park, W.; Na, K. Self-organized nanogels from pullulan- g -poly(L-lactide) synthesized by one-pot method: Physicochemical characterization and in vitro doxorubicin release. J. Appl. Polym. Sci. 2009, 113, 2209–2216. [Google Scholar] [CrossRef]
  72. Schauperl, M.; Podewitz, M.; Waldner, B.J.; Liedl, K.R. Enthalpic and Entropic Contributions to Hydrophobicity. J. Chem. Theory Comput. 2016, 12, 4600–4610. [Google Scholar] [CrossRef]
  73. Dhanya, S.; Bahadur, D.; Kundu, G.C.; Srivastava, R. Maleic acid incorporated poly-(N-isopropylacrylamide) polymer nanogels for dual-responsive delivery of doxorubicin hydrochloride. Eur. Polym. J. 2013, 49, 22–32. [Google Scholar] [CrossRef]
  74. Qian, K.; Ma, Y.; Wan, J.; Geng, S.; Li, H.; Fu, Q.; Peng, X.; Kan, X.; Zhou, G.; Liu, W.; et al. The studies about doxorubicin-loaded p (N-isopropyl-acrylamide-co-butyl methylacrylate) temperature-sensitive nanogel dispersions on the application in TACE therapies for rabbit VX2 liver tumor. J. Control. Release 2015, 212, 41–49. [Google Scholar] [CrossRef]
  75. Yu, T.; Geng, S.; Li, H.; Wan, J.; Peng, X.; Liu, W.; Zhao, Y.; Yang, X.; Xu, H. The stimuli-responsive multiphase behavior of core-shell nanogels with opposite charges and their potential application in in situ gelling system. Colloids Surf. B Biointerfaces 2015, 136, 99–104. [Google Scholar] [CrossRef]
  76. Quan, C.Y.; Sun, Y.X.; Cheng, H.; Cheng, S.X.; Zhang, X.Z.; Zhuo, R.X. Thermosensitive P (NIPAAm-co-PAAc-co-HEMA) nanogels conjugated with transferrin for tumor cell targeting delivery. Nanotechnology 2008, 19. [Google Scholar] [CrossRef]
  77. Aguirre, G.; Villar-Alvarez, E.; González, A.; Ramos, J.; Taboada, P.; Forcada, J. Biocompatible stimuli-responsive nanogels for controlled antitumor drug delivery. J. Polym. Sci. Part Polym. Chem. 2016, 54, 1694–1705. [Google Scholar] [CrossRef]
  78. Zhou, L.; Zhang, F. Thermo-sensitive and photoluminescent hydrogels: Synthesis, characterization, and their drug-release property. Mater. Sci. Eng. C 2011, 31, 1429–1435. [Google Scholar] [CrossRef]
  79. Molina, M.; Wedepohl, S.; Calderón, M. Polymeric near-infrared absorbing dendritic nanogels for efficient in vivo photothermal cancer therapy. Nanoscale 2016, 8, 5852–5856. [Google Scholar] [CrossRef] [Green Version]
  80. Kubota, K.; Hamano, K.; Kuwahara, N.; Fujishige, S.; Ando, I. Characterization of Poly(N-isopropylmethacrylamide) in Water. Polym. J. 1990, 22, 1051–1057. [Google Scholar] [CrossRef]
  81. Djokpé, E.; Vogt, W. N-isopropylacrylamide and N-isopropylmethacrylamide: Cloud points of mixtures and copolymers. Macromol. Chem. Phys. 2001, 202, 750–757. [Google Scholar] [CrossRef]
  82. Cors, M.; Wrede, O.; Genix, A.C.; Anselmetti, D.; Oberdisse, J.; Hellweg, T. Core-Shell Microgel-Based Surface Coatings with Linear Thermoresponse. Langmuir 2017, 33, 6804–6811. [Google Scholar] [CrossRef] [Green Version]
  83. Peters, J.T.; Hutchinson, S.S.; Lizana, N.; Verma, I.; Peppas, N.A. Synthesis and characterization of poly(N-isopropyl methacrylamide) core/shell nanogels for controlled release of chemotherapeutics. Chem. Eng. J. 2018, 340, 58–65. [Google Scholar] [CrossRef]
  84. Deshpande, S.; Sharma, S.; Koul, V.; Singh, N. Core-shell nanoparticles as an efficient, sustained, and triggered drug-delivery system. ACS Omega 2017, 2, 6455–6463. [Google Scholar] [CrossRef]
  85. Idziak, I.; Avoce, D.; Lessard, D.; Gravel, D.; Zhu, X.X. Thermosensitivity of Aqueous Solutions of Poly( N,N -diethylacrylamide). Macromolecules 1999, 32, 1260–1263. [Google Scholar] [CrossRef]
  86. Colonne, M.; Chen, Y.; Wu, K.; Freiberg, S.; Giasson, S.; Zhu, X.X. Binding of streptavidin with biotinylated thermosensitive nanospheres based on poly(N,N-diethylacrylamide-co-2-hydroxyethyl methacrylate). Bioconjug. Chem. 2007, 18, 999–1003. [Google Scholar] [CrossRef]
  87. Kishi, R.; Matsuda, A.; Miura, T.; Matsumura, K.; Iio, K. Fast responsive poly(N, N-diethylacrylamide) hydrogels with interconnected microspheres and bi-continuous structures. Colloid Polym. Sci. 2009, 287, 505–512. [Google Scholar] [CrossRef]
  88. Horák, D.; Matulka, K.; Hlídková, H.; Lapčíková, M.; Beneš, M.J.; Jaroš, J.; Hampl, A.; Dvořák, P. Pentapeptide-modified poly(N,N-diethylacrylamide) hydrogel scaffolds for tissue engineering. J. Biomed. Mater. Res. Part B Appl. Biomater. 2011, 98 B, 54–67. [Google Scholar] [CrossRef]
  89. Scherzinger, C.; Lindner, P.; Keerl, M.; Richtering, W. Cononsolvency of poly(N, N-diethylacrylamide) (PDEAAM) and poly(N-isopropylacrylamide) (PNIPAM) based microgels in water/methanol mixtures: Copolymer vs core-shell microgel. Macromolecules 2010, 43, 6829–6833. [Google Scholar] [CrossRef]
  90. Ngadaonye, J.I.; Geever, L.M.; Killion, J.; Higginbotham, C.L. Development of novel chitosan-poly(N,N-diethylacrylamide) IPN films for potential wound dressing and biomedical applications. J. Polym. Res. 2013, 20. [Google Scholar] [CrossRef]
  91. Blasco, E.; Schmidt, B.V.K.J.; Barner-Kowollik, C.; Piñol, M.; Oriol, L. Dual thermo- and photo-responsive micelles based on miktoarm star polymers. Polym. Chem. 2013, 4, 4506–4514. [Google Scholar] [CrossRef]
  92. Delaittre, G.; Save, M.; Gaborieau, M.; Castignolles, P.; Rieger, J.; Charleux, B. Synthesis by nitroxide-mediated aqueous dispersion polymerization, characterization, and physical core-crosslinking of pH- and thermoresponsive dynamic diblock copolymer micelles. Polym. Chem. 2012, 3, 1526–1538. [Google Scholar] [CrossRef]
  93. Li, X.; Li, X.; Shi, X.; Qiu, G.; Lu, X. Thermosensitive DEA/DMA copolymer nanogel: Low initiator induced synthesis and structural colored colloidal array’s optical properties. Eur. Polym. J. 2017, 96, 484–493. [Google Scholar] [CrossRef]
  94. Grazon, C.; Rieger, J.; Sanson, N.; Charleux, B. Study of poly(N,N-diethylacrylamide) nanogel formation by aqueous dispersion polymerization of N,N-diethylacrylamide in the presence of poly(ethylene oxide)-b-poly(N,N-dimethylacrylamide) amphiphilic macromolecular RAFT agents. Soft Matter 2011, 7, 3482–3490. [Google Scholar] [CrossRef]
  95. Lu, X.; Sun, M.; Barron, A.E. Non-ionic, thermo-responsive DEA/DMA nanogels: Synthesis, characterization, and use for DNA separations by microchip electrophoresis. J. Colloid Interface Sci. 2011, 357, 345–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rieger, J.; Grazon, C.; Charleux, B.; Alaimo, D.; Jérôme, C. Pegylated thermally responsive block copolymer micelles and nanogels via in situ RAFT aqueous dispersion polymerization. J. Polym. Sci. Part Polym. Chem. 2009, 47, 2373–2390. [Google Scholar] [CrossRef]
  97. Yu, Z.; Gu, H.; Tang, D.; Lv, H.; Ren, Y.; Gu, S. Fabrication of PVCL-co-PMMA nanofibers with tunable volume phase transition temperatures and maintainable shape for anti-cancer drug release. RSC Adv. 2015, 5, 64944–64950. [Google Scholar] [CrossRef]
  98. Roh, Y.H.; Moon, J.Y.; Hong, E.J.; Kim, H.U.; Shim, M.S.; Bong, K.W. Microfluidic fabrication of biocompatible poly(N-vinylcaprolactam)-based microcarriers for modulated thermo-responsive drug release. Colloids Surf. B Biointerfaces 2018, 172, 380–386. [Google Scholar] [CrossRef] [PubMed]
  99. Lynch, B.; Crawford, K.; Baruti, O.; Abdulahad, A.; Webster, M.; Puetzer, J.; Ryu, C.; Bonassar, L.J.; Mendenhall, J. The effect of hypoxia on thermosensitive poly(N-vinylcaprolactam) hydrogels with tunable mechanical integrity for cartilage tissue engineering. J. Biomed. Mater. Res. Part B Appl. Biomater. 2017, 105, 1863–1873. [Google Scholar] [CrossRef]
  100. Kutsevol, N.; Glamazda, A.; Chumachenko, V.; Harahuts, Yu.; Stepanian, S.G.; Plokhotnichenko, A.M.; Karachevtsev, V.A. Behavior of hybrid thermosensitive nanosystem dextran-graft-PNIPAM/gold nanoparticles: Characterization within LCTS. J. Nanoparticle Res. 2018, 20, 236. [Google Scholar] [CrossRef]
  101. Chen, Y.; Cheng, W.; Teng, L.; Jin, M.; Lu, B.; Ren, L.; Wang, Y. Graphene Oxide Hybrid Supramolecular Hydrogels with Self-Healable, Bioadhesive and Stimuli-Responsive Properties and Drug Delivery Application. Macromol. Mater. Eng. 2018, 303, 1–11. [Google Scholar] [CrossRef]
  102. Wang, J.; Chen, G.; Zhao, Z.; Sun, L.; Zou, M.; Ren, J.; Zhao, Y. Responsive graphene oxide hydrogel microcarriers for controllable cell capture and release. Sci. China Mater. 2018, 61, 1314–1324. [Google Scholar] [CrossRef] [Green Version]
  103. Callejas-Fernández, J.; Ramos, J.; Forcada, J.; Moncho-Jordá, A. On the scattered light by dilute aqueous dispersions of nanogel particles. J. Colloid Interface Sci. 2015, 450, 310–315. [Google Scholar] [CrossRef] [PubMed]
  104. Kehren, D.; Molano Lopez, A.C.; Pich, A. Nanogel-modified polycaprolactone microfibres with controlled water uptake and degradability. Polymer 2014, 55, 2153–2162. [Google Scholar] [CrossRef]
  105. Madhusudana Rao, K.; Mallikarjuna, B.; Krishna Rao, K.S.V.; Siraj, S.; Chowdoji Rao, K.; Subha, M.C.S. Novel thermo/pH sensitive nanogels composed from poly(N-vinylcaprolactam) for controlled release of an anticancer drug. Colloids Surf. B Biointerfaces 2013, 102, 891–897. [Google Scholar] [CrossRef] [PubMed]
  106. Tang, J.; Cui, X.; Caranasos, T.G.; Hensley, M.T.; Vandergriff, A.C.; Hartanto, Y.; Shen, D.; Zhang, H.; Zhang, J.; Cheng, K. Heart Repair Using Nanogel-Encapsulated Human Cardiac Stem Cells in Mice and Pigs with Myocardial Infarction. ACS Nano 2017, 11, 9738–9749. [Google Scholar] [CrossRef] [PubMed]
  107. Xu, N.; Huang, X.; Yin, G.; Bu, M.; Pu, X.; Chen, X.; Liao, X.; Huang, Z. Thermosensitive star polymer pompons with a core-arm structure as thermo-responsive controlled release drug carriers. RSC Adv. 2018, 8, 15604–15612. [Google Scholar] [CrossRef] [Green Version]
  108. Cao, P.; Li, W.; Sun, X.; Liang, Y.; Gao, X.; Li, X.; Song, Z.; Liang, G. Gene delivery by a cationic and thermosensitive nanogel promoted established tumor growth inhibition. Nanomed. 2015, 10, 1585–1597. [Google Scholar] [CrossRef]
  109. Shakoori, Z.; Ghanbari, H.; Omidi, Y.; Pashaiasl, M.; Akbarzadeh, A.; Jomeh Farsangi, Z.; Rezayat, S.M.; Davaran, S. Fluorescent multi-responsive cross-linked P(N-isopropylacrylamide)-based nanocomposites for cisplatin delivery. Drug Dev. Ind. Pharm. 2017, 43, 1283–1291. [Google Scholar] [CrossRef]
  110. Zhang, L.; Yang, Z.; Zhu, W.; Ye, Z.; Yu, Y.; Xu, Z.; Ren, J.; Li, P. Dual-Stimuli-Responsive, Polymer-Microsphere-Encapsulated CuS Nanoparticles for Magnetic Resonance Imaging Guided Synergistic Chemo-Photothermal Therapy. ACS Biomater. Sci. Eng. 2017, 3, 1690–1701. [Google Scholar] [CrossRef]
  111. Poorgholy, N.; Massoumi, B.; Jaymand, M. A novel starch-based stimuli-responsive nanosystem for theranostic applications. Int. J. Biol. Macromol. 2017, 97, 654–661. [Google Scholar] [CrossRef]
  112. Le, P.N.; Pham, D.C.; Nguyen, D.H.; Tran, N.Q.; Dimitrov, V.; Ivanov, P.; Xuan, C.N.; Nguyen, H.N.; Nguyen, C.K. Poly (N-isopropylacrylamide)-functionalized dendrimer as a thermosensitive nanoplatform for delivering malloapelta B against HepG2 cancer cell proliferation. Adv. Nat. Sci. Nanosci. Nanotechnol. 2017, 8. [Google Scholar] [CrossRef]
  113. Wu, Y.; Li, H.; Rao, Z.; Li, H.; Wu, Y.; Zhao, J.; Rong, J. Controlled protein adsorption and delivery of thermosensitive poly (N-isopropylacrylamide) nanogels. J. Mater. Chem. B 2017, 5, 7974–7984. [Google Scholar] [CrossRef] [PubMed]
  114. Le, P.N.; Nguyen, N.H.; Nguyen, C.K.; Tran, N.Q. Smart dendrimer-based nanogel for enhancing 5-fluorouracil loading efficiency against MCF7 cancer cell growth. Bull. Mater. Sci. 2016, 39, 1493–1500. [Google Scholar] [CrossRef]
  115. Verma, N.K.; Purohit, M.P.; Equbal, D.; Dhiman, N.; Singh, A.; Kar, A.K.; Shankar, J.; Tehlan, S.; Patnaik, S. Targeted Smart pH and Thermoresponsive N,O-Carboxymethyl Chitosan Conjugated Nanogels for Enhanced Therapeutic Efficacy of Doxorubicin in MCF-7 Breast Cancer Cells. Bioconjug. Chem. 2016, 27, 2605–2619. [Google Scholar] [CrossRef] [PubMed]
  116. Chiang, W.; Huang, W.; Chang, Y.; Shen, M.; Chen, H.; Chern, C.; Chiu, H. Doxorubicin-Loaded Nanogel Assemblies with pH/Thermo-triggered Payload Release for Intracellular Drug Delivery. Macromol. Chem. Phys. 2014, 215, 1332–1341. [Google Scholar] [CrossRef]
  117. Li, L.; Fu, L.; Ai, X.; Zhang, J.; Zhou, J. Design and Fabrication of Temperature-Sensitive Nanogels with Controlled Drug Release Properties for Enhanced Photothermal Sterilization. Chem. Eur. J. 2017, 23, 18180–18186. [Google Scholar] [CrossRef]
  118. Bardajee, G.R.; Hooshyar, Z. A novel thermo-sensitive nanogel composing of poly(N-isopropylacrylamide) grafted onto alginate-modified graphene oxide for hydrophilic anticancer drug delivery. J. Iran. Chem. Soc. 2018, 15, 121–129. [Google Scholar] [CrossRef]
  119. Xu, X.; Wang, X.; Luo, W.; Qian, Q.; Li, Q.; Han, B.; Li, Y. Triple cell-responsive nanogels for delivery of drug into cancer cells. Colloids Surf. B Biointerfaces 2018, 163, 362–368. [Google Scholar] [CrossRef]
  120. Zhang, H.; Li, Q.; Zhang, Y.; Xia, Y.; Yun, L.; Zhang, Q.; Zhang, T.; Chen, X.; Chen, H.; Li, W. A nanogel with passive targeting function and adjustable polyplex surface properties for efficient anti-tumor gene therapy. RSC Adv. 2016, 6, 84445–84456. [Google Scholar] [CrossRef]
  121. Naga Sravan Kumar Varma, V.; Shivakumar, S.; Fathima, S.J.; Radha, V.; Khanum, F. PH and thermosensitive 5-fluorouracil loaded poly (NIPAM-: Co -AAc) nanogels for cancer therapy. RSC Adv. 2016, 6, 105495–105507. [Google Scholar] [CrossRef]
  122. Lee, S.H.; Bui, H.T.; Vales, T.P.; Cho, S.; Kim, H.J. Multi-color fluorescence of pNIPAM-Based nanogels modulated by dual stimuli-responsive FRET processes. Dyes Pigments 2017, 145, 216–221. [Google Scholar] [CrossRef]
  123. Liu, D.; Ma, L.; An, Y.; Li, Y.; Liu, Y.; Wang, L.; Guo, J.; Wang, J.; Zhou, J. Thermoresponsive Nanogel-Encapsulated PEDOT and HSP70 Inhibitor for Improving the Depth of the Photothermal Therapeutic Effect. Adv. Funct. Mater. 2016, 26, 4749–4759. [Google Scholar] [CrossRef]
  124. Zhang, W.; Jiang, P.; Chen, J.; Zhu, C.; Mao, Z.; Gao, C. Application of melatonin-loaded poly(N-isopropylacrylamide) hydrogel particles to reduce the toxicity of airborne pollutes to RAW264.7 cells. J. Colloid Interface Sci. 2017, 490, 181–189. [Google Scholar] [CrossRef] [PubMed]
  125. Bardajee, G.R.; Hooshyar, Z. Drug release study by a novel thermo sensitive nanogel based on salep modified graphene oxide. J. Polym. Res. 2017, 24. [Google Scholar] [CrossRef]
  126. Pikabea, A.; Ramos, J.; Forcada, J. Production of cationic nanogels with potential use in controlled drug delivery. Part. Part. Syst. Charact. 2014, 31, 101–109. [Google Scholar] [CrossRef]
  127. Chen, Z.; Zhang, K.Y.; Tong, X.; Liu, Y.; Hu, C.; Liu, S.; Yu, Q.; Zhao, Q.; Huang, W. Phosphorescent Polymeric Thermometers for In Vitro and In Vivo Temperature Sensing with Minimized Background Interference. Adv. Funct. Mater. 2016, 26, 4386–4396. [Google Scholar] [CrossRef]
  128. Cao, Z.; Zhou, X.; Wang, G. Selective Release of Hydrophobic and Hydrophilic Cargos from Multi-Stimuli-Responsive Nanogels. ACS Appl. Mater. Interfaces 2016, 8, 28888–28896. [Google Scholar] [CrossRef]
  129. Carmona-Moran, C.A.; Zavgorodnya, O.; Penman, A.D.; Kharlampieva, E.; Bridges, S.L.; Hergenrother, R.W.; Singh, J.A.; Wick, T.M. Development of gellan gum containing formulations for transdermal drug delivery: Component evaluation and controlled drug release using temperature responsive nanogels. Int. J. Pharm. 2016, 509, 465–476. [Google Scholar] [CrossRef]
  130. Aguirre, G.; Ramos, J.; Forcada, J. Synthesis of new enzymatically degradable thermo-responsive nanogels. Soft Matter 2013, 9, 261–270. [Google Scholar] [CrossRef]
  131. Gonzalez-Ayon, M.A.; Cortez-Lemus, N.A.; Zizumbo-Lopez, A.; Licea-Claverie, A. Nanogels of poly(N-vinylcaprolactam) core and polyethyleneglycol shell by surfactant free emulsion polymerization. Soft Mater. 2014, 12, 315–325. [Google Scholar] [CrossRef]
  132. Rejinold, N.S.; Baby, T.; Chennazhi, K.P.; Jayakumar, R. Multi drug loaded thermo-responsive fibrinogen-graft-Poly (N-vinyl caprolactam) nanogels for breast cancer drug delivery. J. Biomed. Nanotechnol. 2015, 11, 392–402. [Google Scholar] [CrossRef]
  133. Peng, H.; Xu, W.; Pich, A. Temperature and pH dual-responsive Poly (vinyl lactam) copolymers functionalized with amine side groups: Via RAFT polymerization. Polym. Chem. 2016, 7, 5011–5022. [Google Scholar] [CrossRef]
  134. Agrawal, G.; Agrawal, R.; Pich, A. Dual Responsive Poly (N-vinylcaprolactam) Based Degradable Microgels for Drug Delivery. Part. Part. Syst. Charact. 2017, 34, 1–9. [Google Scholar] [CrossRef]
  135. Wu, J.Z.; Bremner, D.H.; Li, H.Y.; Sun, X.Z.; Zhu, L.M. Synthesis and evaluation of temperature- and glucose-sensitive nanoparticles based on phenylboronic acid and N-vinylcaprolactam for insulin delivery. Mater. Sci. Eng. C 2016, 69, 1026–1035. [Google Scholar] [CrossRef] [Green Version]
  136. Etchenausia, L.; Deniau, E.; Brûlet, A.; Forcada, J.; Save, M. Cationic Thermoresponsive Poly (N-vinylcaprolactam) Microgels Synthesized by Emulsion Polymerization Using a Reactive Cationic Macro-RAFT Agent. Macromolecules 2018, 51, 2551–2563. [Google Scholar] [CrossRef]
  137. Lou, S.; Gao, S.; Wang, W.; Zhang, M.; Zhang, J.; Wang, C.; Li, C.; Kong, D.; Zhao, Q. Galactose-functionalized multi-responsive nanogels for hepatoma-targeted drug delivery. Nanoscale 2015, 7, 3137–3146. [Google Scholar] [CrossRef]
  138. Dong, X.; Wei, C.; Lu, L.; Liu, T.; Lv, F. Fluorescent nanogel based on four-arm PEG-PCL copolymer with porphyrin core for bioimaging. Mater. Sci. Eng. C 2016, 61, 214–219. [Google Scholar] [CrossRef] [PubMed]
  139. Góis, J.R.; Serra, A.C.; Coelho, J.F.J. Synthesis and characterization of new temperature-responsive nanocarriers based on POEOMA-b-PNVCL prepared using a combination of ATRP, RAFT and CuAAC. Eur. Polym. J. 2016, 81, 224–238. [Google Scholar] [CrossRef]
  140. Berger, S.; Ornatsky, O.; Baranov, V.; Winnik, M.A.; Pich, A. Hybrid nanogels by encapsulation of lanthanide-doped LaF3nanoparticles as elemental tags for detection by atomic mass spectrometry. J. Mater. Chem. 2010, 20, 5141–5150. [Google Scholar] [CrossRef]
  141. Ye, Z.; Li, Y.; An, Z.; Wu, P. Exploration of Doubly Thermal Phase Transition Process of PDEGA-b-PDMA-b-PVCL in Water. Langmuir 2016, 32, 6691–6700. [Google Scholar] [CrossRef]
  142. Cui, Q.; Wu, F.; Wang, E. Thermosensitive behavior of poly(ethylene Glycol)-based block copolymer (peg-b-PADMO) controlled via self-assembled microstructure. J. Phys. Chem. B 2011, 115, 5913–5922. [Google Scholar] [CrossRef]
  143. Felberg, L.E.; Doshi, A.; Hura, G.L.; Sly, J.; Piunova, V.A.; Swope, W.C.; Rice, J.E.; Miller, R.; Head-Gordon, T. Structural transition of nanogel star polymers with pH by controlling PEGMA interactions with acid or base copolymers. Mol. Phys. 2016, 114, 3221–3231. [Google Scholar] [CrossRef] [Green Version]
  144. Peters, J.T.; Verghese, S.; Subramanian, D.; Peppas, N.A. Surface hydrolysis-mediated PEGylation of poly(N-isopropyl acrylamide) based nanogels. Regen. Biomater. 2017, 4, 281–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Kadlubowski, S.; Matusiak, M.; Jenczyk, J.; Olejniczak, M.N.; Kozanecki, M.; Okrasa, L. Radiation-induced synthesis of thermo-sensitive, gradient hydrogels based on 2-(2-methoxyethoxy)ethyl methacrylate. Radiat. Phys. Chem. 2014, 100, 23–31. [Google Scholar] [CrossRef]
  146. París, R.; Quijada-Garrido, I. Temperature- and pH-responsive behaviour of poly (2-(2-methoxyethoxy) ethyl methacrylate-co-N,N-dimethylaminoethyl methacrylate) hydrogels. Eur. Polym. J. 2010, 46, 2156–2163. [Google Scholar] [CrossRef]
  147. Shen, W.; Chang, Y.; Liu, G.; Wang, H.; Cao, A.; An, Z. Biocompatible, Antifouling, and Thermosensitive Core−Shell Nanogels Synthesized by RAFT Aqueous Dispersion Polymerization. Macromolecules 2011, 44, 2524–2530. [Google Scholar] [CrossRef]
  148. Biglione, C.; Sousa-Herves, A.; Menger, M.; Wedepohl, S.; Calderón, M.; Strumia, M.C. Facile ultrasonication approach for the efficient synthesis of ethylene glycol-based thermoresponsive nanogels. RSC Adv. 2015, 5, 15407–15413. [Google Scholar] [CrossRef] [Green Version]
  149. Zhang, C.; Maric, M. Statistical terpolymers with thermo-responsive fluorescence response in an ionic liquid: Effects of solvatophilicity on LCST phase separation and reversibility. Polym. Chem. 2014, 5, 4926–4938. [Google Scholar] [CrossRef]
  150. Rajan, R.; Matsumura, K. Tunable Dual-Thermoresponsive Core–Shell Nanogels Exhibiting UCST and LCST Behavior. Macromol. Rapid Commun. 2017, 38, 1–6. [Google Scholar] [CrossRef] [Green Version]
  151. Cao, H.; Guo, F.; Chen, Z.; Kong, X.Z. Preparation of Thermoresponsive Polymer Nanogels of Oligo (Ethylene Glycol) Diacrylate-Methacrylic Acid and Their Property Characterization. Nanoscale Res. Lett. 2018, 13, 1–10. [Google Scholar] [CrossRef] [Green Version]
  152. Lutz, J.-F.; Akdemir, Ö.; Hoth, A. Point by Point Comparison of Two Thermosensitive Polymers Exhibiting a Similar LCST: Is the Age of Poly (NIPAM) Over? J. Am. Chem. Soc. 2006, 128, 13046–13047. [Google Scholar] [CrossRef]
  153. Fang, Q.; Chen, T.; Zhong, Q.; Wang, J. Thermoresponsive polymers based on oligo (ethylene glycol) methyl ether methacrylate and modified substrates with thermosensitivity. Macromol. Res. 2017, 25, 206–213. [Google Scholar] [CrossRef]
  154. Guo, Y.; Dong, X.; Ruan, W.; Shang, Y.; Liu, H. A thermo-sensitive OEGMA-based polymer: Synthesis, characterization and interactions with surfactants in aqueous solutions with and without salt. Colloid Polym. Sci. 2017, 295, 327–340. [Google Scholar] [CrossRef]
  155. Alejo, T.; Prieto, M.; García-Juan, H.; Andreu, V.; Mendoza, G.; Sebastián, V.; Arruebo, M. A facile method for the controlled polymerization of biocompatible and thermoresponsive oligo (ethylene glycol) methyl ether methacrylate copolymers. Polym. J. 2018, 50, 203–211. [Google Scholar] [CrossRef]
  156. Zhu, C.; Xiao, J.; Tang, M.; Feng, H.; Chen, W.; Du, M. Platinum covalent shell cross-linked micelles designed to deliver doxorubicin for synergistic combination cancer therapy. Int. J. Nanomed. 2017, 12, 3697–3710. [Google Scholar] [CrossRef] [Green Version]
  157. Li, X.; Qian, Y.; Liu, T.; Hu, X.; Zhang, G.; You, Y.; Liu, S. Biomaterials Amphiphilic multiarm star block copolymer-based multifunctional unimolecular micelles for cancer targeted drug delivery and MR imaging. Biomaterials 2011, 32, 6595–6605. [Google Scholar] [CrossRef]
  158. Wang, L.H.; Xu, X.M.; Hong, C.Y.; Wu, D.C.; Yu, Z.Q.; You, Y.Z. Biodegradable large compound vesicles with controlled size prepared via the self-assembly of branched polymers in nanodroplet templates. Chem. Commun. 2014, 50, 9676–9678. [Google Scholar] [CrossRef]
  159. Qiao, Z.-Y.; Zhang, R.; Du, F.-S.; Liang, D.-H.; Li, Z.-C. Multi-responsive nanogels containing motifs of ortho ester, oligo(ethylene glycol) and disulfide linkage as carriers of hydrophobic anti-cancer drugs. J. Control. Release 2011, 152, 57–66. [Google Scholar] [CrossRef] [PubMed]
  160. Xia, M.; Cheng, Y.; Theato, P.; Zhu, M. Thermo-induced double phase transition behavior of physically cross-linked hydrogels based on oligo(ethylene glycol) methacrylates. Macromol. Chem. Phys. 2015, 216, 2230–2240. [Google Scholar] [CrossRef]
  161. Alejo, T.; Andreu, V.; Mendoza, G.; Sebastian, V.; Arruebo, M. Controlled release of bupivacaine using hybrid thermoresponsive nanoparticles activated via photothermal heating. J. Colloid Interface Sci. 2018, 523, 234–244. [Google Scholar] [CrossRef] [Green Version]
  162. Sheng, W.; Liu, T.; Liu, S.; Wang, Q.; Li, X.; Guang, N. Temperature and pH responsive hydrogels based on polyethylene glycol analogues and poly (methacrylic acid) via click chemistry. Polym. Int. 2015, 64, 1415–1424. [Google Scholar] [CrossRef]
  163. Cazares-Cortes, E.; Espinosa, A.; Guigner, J.M.; Michel, A.; Griffete, N.; Wilhelm, C.; Ménager, C. Doxorubicin Intracellular Remote Release from Biocompatible Oligo(ethylene glycol) Methyl Ether Methacrylate-Based Magnetic Nanogels Triggered by Magnetic Hyperthermia. ACS Appl. Mater. Interfaces 2017, 9, 25775–25788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Hayashi, K.; Ono, K.; Suzuki, H.; Sawada, M.; Moriya, M.; Sakamoto, W.; Yogo, T. High-Frequency, Magnetic-Field-Responsive Drug Release from Magnetic Nanoparticle/Organic Hybrid Based on Hyperthermic Effect. ACS Appl. Mater. Interfaces 2010, 2, 1903–1911. [Google Scholar] [CrossRef]
  165. Liu, L.; Li, W.; Liu, K.; Yan, J.; Hu, G.; Zhang, A. Comblike Thermoresponsive Polymers with Sharp Transitions: Synthesis, Characterization, and Their Use as Sensitive Colorimetric Sensors. Macromolecules 2011, 44, 8614–8621. [Google Scholar] [CrossRef]
  166. Wu, W.; Shen, J.; Banerjee, P.; Zhou, S. A multifuntional nanoplatform based on responsive fluorescent plasmonic ZnO-Au@PEG hybrid nanogels. Adv. Funct. Mater. 2011, 21, 2830–2839. [Google Scholar] [CrossRef]
  167. Zhao, A.; Zhou, S.; Zhou, Q.; Chen, T. Thermosensitive micelles from PEG-based ether-anhydride triblock copolymers. Pharm. Res. 2010, 27, 1627–1643. [Google Scholar] [CrossRef] [PubMed]
  168. Yang, Z.; Ding, J. A thermosensitive and biodegradable physical gel with chemically crosslinked nanogels as the building block. Macromol. Rapid Commun. 2008, 29, 751–756. [Google Scholar] [CrossRef]
  169. Wu, J.; Liu, X.Q.; Wang, Y.C.; Wang, J. Template-free synthesis of biodegradable nanogels with tunable sizes as potential carriers for drug delivery. J. Mater. Chem. 2009, 19, 7856–7863. [Google Scholar] [CrossRef]
  170. Ko, D.Y.; Moon, H.J.; Jeong, B. Temperature-sensitive polypeptide nanogels for intracellular delivery of a biomacromolecular drug. J. Mater. Chem. B 2015, 3, 3525–3530. [Google Scholar] [CrossRef]
  171. Liu, G.; Qiu, Q.; An, Z. Development of thermosensitive copolymers of poly (2-methoxyethyl acrylate-co-poly (ethylene glycol) methyl ether acrylate) and their nanogels synthesized by RAFT dispersion polymerization in water. Polym. Chem. 2012, 3, 504–513. [Google Scholar] [CrossRef]
  172. Ahmed, M.; Narain, R. Intracellular delivery of DNA and enzyme in active form using degradable carbohydrate-based nanogels. Mol. Pharm. 2012, 9, 3160–3170. [Google Scholar] [CrossRef]
  173. Ulasan, M.; Yavuz, E.; Bagriacik, E.U.; Cengeloglu, Y.; Yavuz, M.S. Biocompatible thermoresponsive PEGMA nanoparticles crosslinked with cleavable disulfide-based crosslinker for dual drug release. J. Biomed. Mater. Res. Part A 2015, 103, 243–251. [Google Scholar] [CrossRef] [PubMed]
  174. Aktan, B.; Chambre, L.; Sanyal, R.; Sanyal, A. “Clickable” Nanogels via Thermally Driven Self-Assembly of Polymers: Facile Access to Targeted Imaging Platforms using Thiol-Maleimide Conjugation. Biomacromolecules 2017, 18, 490–497. [Google Scholar] [CrossRef] [PubMed]
  175. Quan, S.; Wang, Y.; Zhou, A.; Kumar, P.; Narain, R. Galactose-based thermosensitive nanogels for targeted drug delivery of iodoazomycin arabinofuranoside (IAZA) for theranostic management of hypoxic hepatocellular carcinoma. Biomacromolecules 2015, 16, 1978–1986. [Google Scholar] [CrossRef] [PubMed]
  176. Cheng, W.; Chen, Y.; Teng, L.; Lu, B.; Ren, L.; Wang, Y. Antimicrobial colloidal hydrogels assembled by graphene oxide and thermo-sensitive nanogels for cell encapsulation. J. Colloid Interface Sci. 2018, 513, 314–323. [Google Scholar] [CrossRef]
  177. Wang, X.; Wu, Z.; Li, J.; Pan, G.; Shi, D.; Ren, J. Preparation, characterization, biotoxicity, and biodistribution of thermo-responsive magnetic complex micelles formed by Mn0.6Zn0.4Fe2O4and a PCL/PEG analogue copolymer for controlled drug delivery. J. Mater. Chem. B 2017, 5, 296–306. [Google Scholar] [CrossRef] [PubMed]
  178. Wen, Y.; Oh, J.K. Intracellular delivery cellulose-based bionanogels with dual temperature/pH-response for cancer therapy. Colloids Surf. B Biointerfaces 2015, 133, 246–253. [Google Scholar] [CrossRef]
  179. Wu, W.; Shen, J.; Banerjee, P.; Zhou, S. Core-shell hybrid nanogels for integration of optical temperature-sensing, targeted tumor cell imaging, and combined chemo-photothermal treatment. Biomaterials 2010, 31, 7555–7566. [Google Scholar] [CrossRef]
  180. Liras, M.; Quijada-Garrido, I.; García, O. QDs decorated with thiol-monomer ligands as new multicrosslinkers for the synthesis of smart luminescent nanogels and hydrogels. Polym. Chem. 2017, 8, 5317–5326. [Google Scholar] [CrossRef]
  181. Wu, W.; Shen, J.; Banerjee, P.; Zhou, S. Water-dispersible multifunctional hybrid nanogels for combined curcumin and photothermal therapy. Biomaterials 2011, 32, 598–609. [Google Scholar] [CrossRef]
  182. Asadian-Birjand, M.; Bergueiro, J.; Rancan, F.; Cuggino, J.C.; Mutihac, R.C.; Achazi, K.; Dernedde, J.; Blume-Peytayi, U.; Vogt, A.; Calderón, M. Engineering thermoresponsive polyether-based nanogels for temperature dependent skin penetration. Polym. Chem. 2015, 6, 5827–5831. [Google Scholar] [CrossRef] [Green Version]
  183. Fernandes Stefanello, T.; Szarpak-Jankowska, A.; Appaix, F.; Louage, B.; Hamard, L.; De Geest, B.G.; Van Der Sanden, B.; Nakamura, C.V.; Auzély-Velty, R. Thermoresponsive hyaluronic acid nanogels as hydrophobic drug carrier to macrophages. Acta Biomater. 2014, 10, 4750–4758. [Google Scholar] [CrossRef] [PubMed]
  184. Nakai, T.; Hirakura, T.; Sakurai, Y.; Shimoboji, T.; Ishigai, M.; Akiyoshi, K. Injectable Hydrogel for Sustained Protein Release by Salt-Induced Association of Hyaluronic Acid Nanogel. Macromol. Biosci. 2012, 12, 475–483. [Google Scholar] [CrossRef] [PubMed]
  185. Tahara, Y.; Sakiyama, M.; Takeda, S.; Nishimura, T.; Mukai, S.A.; Sawada, S.I.; Sasaki, Y.; Akiyoshi, K. Self-Assembled Nanogels of Cholesterol-Bearing Hydroxypropyl Cellulose: A Thermoresponsive Building Block for Nanogel Tectonic Materials. Langmuir 2016, 32, 12283–12289. [Google Scholar] [CrossRef] [PubMed]
  186. Lee, C.-T.; Huang, C.; Lee, Y. Biomimetic Porous Scaffolds Made from Poly(L-lactide)-g-chondroitin Sulfate Blend with Poly(L-lactide) for Cartilage Tissue Engineering. Biomacromolecules 2006, 7, 2200–2209. [Google Scholar] [CrossRef] [PubMed]
  187. Nagahama, K.; Ouchi, T.; Ohya, Y. Biodegradable nanogels prepared by self-assembly of poly(L-lactide)-grafted dextran: Entrapment and release of proteins. Macromol. Biosci. 2008, 8, 1044–1052. [Google Scholar] [CrossRef]
  188. Na, K.; Lee, K.H.; Lee, D.H.; Bae, Y.H. Biodegradable thermo-sensitive nanoparticles from poly (L-lactic acid)/poly (ethylene glycol) alternating multi-block copolymer for potential anti-cancer drug carrier. Eur. J. Pharm. Sci. 2006, 27, 115–122. [Google Scholar] [CrossRef]
  189. Scaffaro, R.; Re, G.L.; Rigogliuso, S.; Ghersi, G. 3D polylactide-based scaffolds for studying human hepatocarcinoma processes in vitro. Sci. Technol. Adv. Mater. 2012, 13, 045003. [Google Scholar] [CrossRef]
  190. Wu, J.; Su, Z.-G.; Ma, G.-H. A thermo- and pH-sensitive hydrogel composed of quaternized chitosan/glycerophosphate. Int. J. Pharm. 2006, 315. [Google Scholar] [CrossRef]
  191. Yun, Q.; Wang, S.S.; Xu, S.; Yang, J.P.; Fan, J.; Yang, L.L.; Chen, Y.; Fu, S.Z.; Wu, J.B. Use of 5-Fluorouracil Loaded Micelles and Cisplatin in Thermosensitive Chitosan Hydrogel as an Efficient Therapy against Colorectal Peritoneal Carcinomatosis. Macromol. Biosci. 2017, 17, 1–12. [Google Scholar] [CrossRef]
  192. Won, D.A.; Kim, M.; Tae, G. Systemic modulation of the stability of pluronic hydrogel by a small amount of graphene oxide. Colloids Surf. B Biointerfaces 2015, 128, 515–521. [Google Scholar] [CrossRef]
  193. Sharma, G.; Kamboj, S.; Thakur, K.; Negi, P.; Raza, K.; Katare, O.P. Delivery of Thermoresponsive-Tailored Mixed Micellar Nanogel of Lidocaine and Prilocaine with Improved Dermatokinetic Profile and Therapeutic Efficacy in Topical Anaesthesia. AAPS Pharm. Sci.Technol. 2017, 18, 790–802. [Google Scholar] [CrossRef] [PubMed]
  194. Kobayashi, H.; Katakura, O.; Morimoto, N.; Akiyoshi, K.; Kasugai, S. Effects of cholesterol-bearing pullulan (CHP)-nanogels in combination with prostaglandin E1 on wound healing. J. Biomed. Mater. Res. Part B Appl. Biomater. 2009, 91, 55–60. [Google Scholar] [CrossRef] [PubMed]
  195. Akiyama, E.; Morimoto, N.; Kujawa, P.; Ozawa, Y.; Winnik, F.M.; Akiyoshi, K. Self-assembled nanogels of cholesteryl-modified polysaccharides: Effect of the polysaccharide structure on their association characteristics in the dilute and semidilute regimes. Biomacromolecules 2007, 8, 2366–2373. [Google Scholar] [CrossRef] [PubMed]
  196. Morimoto, N.; Hirano, S.; Takahashi, H.; Loethen, S.; Thompson, D.H.; Akiyoshi, K. Self-Assembled pH-Sensitive Cholesteryl Pullulan Nanogel As a Protein Delivery Vehicle. Biomacromolecules 2013, 14, 56–63. [Google Scholar] [CrossRef]
  197. Fujioka-Kobayashi, M.; Ota, M.S.; Shimoda, A.; Nakahama, K.I.; Akiyoshi, K.; Miyamoto, Y.; Iseki, S. Cholesteryl group- and acryloyl group-bearing pullulan nanogel to deliver BMP2 and FGF18 for bone tissue engineering. Biomaterials 2012, 33, 7613–7620. [Google Scholar] [CrossRef]
  198. Jung, Y.S.; Park, W.; Na, K. Succinylated polysaccharide-based thermosensitive polyelectrostatic complex for protein drug delivery. J. Bioact. Compat. Polym. 2014, 29, 81–92. [Google Scholar] [CrossRef]
  199. Gioffredi, E.; Boffito, M.; Calzone, S.; Giannitelli, S.M.; Rainer, A.; Trombetta, M.; Mozetic, P.; Chiono, V. Pluronic F127 Hydrogel Characterization and Biofabrication in Cellularized Constructs for Tissue Engineering Applications. Procedia CIRP 2016, 49, 125–132. [Google Scholar] [CrossRef] [Green Version]
  200. Dou, Q.; Karim, A.A.; Loh, X.J. Modification of thermal and mechanical properties of PEG-PPG-PEG copolymer (F127) with MA-POSS. Polymers 2016, 8, 341. [Google Scholar] [CrossRef] [Green Version]
  201. Basak, R.; Bandyopadhyay, R. Encapsulation of hydrophobic drugs in pluronic F127 micelles: Effects of drug hydrophobicity, solution temperature, and pH. Langmuir 2013, 29, 4350–4356. [Google Scholar] [CrossRef] [Green Version]
  202. Al Khateb, K.; Ozhmukhametova, E.K.; Mussin, M.N.; Seilkhanov, S.K.; Rakhypbekov, T.K.; Lau, W.M.; Khutoryanskiy, V.V. In situ gelling systems based on Pluronic F127/Pluronic F68 formulations for ocular drug delivery. Int. J. Pharm. 2016, 502, 70–79. [Google Scholar] [CrossRef]
  203. Yin, Q.Q.; Wu, L.; Gou, M.L.; Qian, Z.Y.; Zhang, W.S.; Liu, J. Long-lasting infiltration anaesthesia by lidocaine-loaded biodegradable nanoparticles in hydrogel in rats. Acta Anaesthesiol. Scand. 2009, 53, 1207–1213. [Google Scholar] [CrossRef] [PubMed]
  204. Choi, W.I.; Sahu, A.; Vilos, C.; Kamaly, N.; Jo, S.M.; Lee, J.H.; Tae, G. Bioinspired Heparin Nanosponge Prepared by Photo-crosslinking for Controlled Release of Growth Factors. Sci. Rep. 2017, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Park, M.R.; Seo, B.B.; Song, S.C. Dual ionic interaction system based on polyelectrolyte complex and ionic, injectable, and thermosensitive hydrogel for sustained release of human growth hormone. Biomaterials 2013, 34, 1327–1336. [Google Scholar] [CrossRef] [PubMed]
  206. Ghaeini-Hesaroeiye, S.; Boddohi, S.; Vasheghani-Farahani, E. Dual responsive chondroitin sulfate based nanogel for antimicrobial peptide delivery. Int. J. Biol. Macromol. 2020, 143, 297–304. [Google Scholar] [CrossRef]
  207. Lučovnik, M.; Mandić, N.T.; Lozar Krivec, J.; Kolenc, U.; Jeverica, S. Prevalenca kolonizacije z bakterijo Streptococcus agalactiae pri nosečnicah v Sloveniji v obdobju 2013-2014. Zdr. Vestn. 2016, 85, 393–400. [Google Scholar] [CrossRef]
  208. Guo, W.; Yang, C.; Lin, H.; Qu, F. P(EO-co-LLA) functionalized Fe3O4@mSiO2nanocomposites for thermo/pH responsive drug controlled release and hyperthermia. Dalton Trans. 2014, 43, 18056–18065. [Google Scholar] [CrossRef]
  209. Seo, S.; Lee, C.S.; Jung, Y.S.; Na, K. Thermo-sensitivity and triggered drug release of polysaccharide nanogels derived from pullulan-g-poly(L-lactide) copolymers. Carbohydr. Polym. 2012, 87, 1105–1111. [Google Scholar] [CrossRef]
  210. Hsiao, M.H.; Larsson, M.; Larsson, A.; Evenbratt, H.; Chen, Y.Y.; Chen, Y.Y.; Liu, D.M. Design and characterization of a novel amphiphilic chitosan nanocapsule-based thermo-gelling biogel with sustained in vivo release of the hydrophilic anti-epilepsy drug ethosuximide. J. Control. Release 2012, 161, 942–948. [Google Scholar] [CrossRef] [Green Version]
  211. Edlich, A.; Gerecke, C.; Giulbudagian, M.; Neumann, F.; Hedtrich, S.; Schäfer-Korting, M.; Ma, N.; Calderon, M.; Kleuser, B. Specific uptake mechanisms of well-tolerated thermoresponsive polyglycerol-based nanogels in antigen-presenting cells of the skin. Eur. J. Pharm. Biopharm. 2017, 116, 155–163. [Google Scholar] [CrossRef]
  212. Tong, N.A.N.; Nguyen, T.P.; Cuu Khoa, N.; Tran, N.Q. Aquated cisplatin and heparin-pluronic nanocomplexes exhibiting sustainable release of active platinum compound and NCI-H460 lung cancer cell antiproliferation. J. Biomater. Sci. Polym. Ed. 2016, 27, 709–720. [Google Scholar] [CrossRef]
  213. Wang, G.; Nie, Q.; Zang, C.; Zhang, B.; Zhu, Q.; Luo, G.; Wang, S. Self-Assembled Thermoresponsive Nanogels Prepared by Reverse Micelle Positive Micelle Method for Ophthalmic Delivery of Muscone, a Poorly Water-Soluble Drug. J. Pharm. Sci. 2016, 105, 2752–2759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Zhu, H.; Li, Y.; Qiu, R.; Shi, L.; Wu, W.; Zhou, S. Responsive fluorescent Bi2O3@PVA hybrid nanogels for temperature-sensing, dual-modal imaging, and drug delivery. Biomaterials 2012, 33, 3058–3069. [Google Scholar] [CrossRef]
  215. Gandhi, S.S.; Yan, H.; Kim, C. Thermoresponsive Gelatin Nanogels. ACS Macro Lett. 2014, 3, 1210–1214. [Google Scholar] [CrossRef]
  216. An, D.; Zhao, D.; Li, X.; Lu, X.; Qiu, G.; Shea, K.J. Synthesis of surfactant-free hydroxypropylcellulose nanogel and its dual-responsive properties. Carbohydr. Polym. 2015, 134, 385–389. [Google Scholar] [CrossRef] [PubMed]
  217. Hassanpour, S.; Bagheri, M. Dual-responsive semi-IPN copolymer nanogels based on poly (itaconic acid) and hydroxypropyl cellulose as a carrier for controlled drug release. J. Polym. Res. 2017, 24, 1–9. [Google Scholar] [CrossRef]
  218. Sun, W.; An, Z.; Wu, P. UCST or LCST? Composition-Dependent Thermoresponsive Behavior of Poly(N-acryloylglycinamide-co-diacetone acrylamide). Macromolecules 2017, 50, 2175–2182. [Google Scholar] [CrossRef]
  219. Nagahama, K.; Hashizume, M.; Yamamoto, H.; Ouchi, T.; Ohya, Y. Hydrophobically modified biodegradable poly (ethylene glycol) copolymers that form temperature-responsive nanogels. Langmuir 2009, 25, 9734–9740. [Google Scholar] [CrossRef]
Figure 1. Schematic degradation of thermosensitive hydrogel below and above LCST.
Figure 1. Schematic degradation of thermosensitive hydrogel below and above LCST.
Gels 06 00020 g001
Figure 2. (a) Infrared-responsive nanocarriers for in-situ forming hydrogels as a drug delivery system and tissue scaffold and (b) magnetic fields and temperature-responsive nanocarriers for increasing drug release.
Figure 2. (a) Infrared-responsive nanocarriers for in-situ forming hydrogels as a drug delivery system and tissue scaffold and (b) magnetic fields and temperature-responsive nanocarriers for increasing drug release.
Gels 06 00020 g002
Figure 3. Scheme of different stimuli-responsive nanogels in response to temperature, enzyme, the magnetic field, and pH in drug delivery applications.
Figure 3. Scheme of different stimuli-responsive nanogels in response to temperature, enzyme, the magnetic field, and pH in drug delivery applications.
Gels 06 00020 g003
Figure 4. In-situ mechanism for near-infrared absorbing nanogels based on PNIPAM-dPG-PANI for photothermal cancer therapy. Adapted from Ref. [79] reproduced by permission of The Royal Society of Chemistry.
Figure 4. In-situ mechanism for near-infrared absorbing nanogels based on PNIPAM-dPG-PANI for photothermal cancer therapy. Adapted from Ref. [79] reproduced by permission of The Royal Society of Chemistry.
Gels 06 00020 g004
Figure 5. NIPAM and NIPMAM-based thermosensitive core–shell nanogels for triggered and sustained release of DOX. (a) Schematic diagram showing the trigger-based release and (b) sustained release of DOX from the nanogels. Adopted from Ref. [84] reproduced by permission of American Chemical Society. Further permissions related to the material excerpted should be directed to the ACS.
Figure 5. NIPAM and NIPMAM-based thermosensitive core–shell nanogels for triggered and sustained release of DOX. (a) Schematic diagram showing the trigger-based release and (b) sustained release of DOX from the nanogels. Adopted from Ref. [84] reproduced by permission of American Chemical Society. Further permissions related to the material excerpted should be directed to the ACS.
Gels 06 00020 g005
Figure 6. Dual-responsive nanogels based on N-vinyl caprolactam (VCL), which illustrate an increase in drug release at high pH, and the polymer network structure is collapsed at high temperature resulting in lower drug release. Adapted from Ref. [105] reproduced by permission of Elsevier.
Figure 6. Dual-responsive nanogels based on N-vinyl caprolactam (VCL), which illustrate an increase in drug release at high pH, and the polymer network structure is collapsed at high temperature resulting in lower drug release. Adapted from Ref. [105] reproduced by permission of Elsevier.
Gels 06 00020 g006
Figure 7. Thermosensitive fluorescent nanogels based on the four-arm PEG–PCL copolymer for co-delivery of hydrophobic and hydrophilic drugs.
Figure 7. Thermosensitive fluorescent nanogels based on the four-arm PEG–PCL copolymer for co-delivery of hydrophobic and hydrophilic drugs.
Gels 06 00020 g007
Figure 8. Thermosensitive nanocarriers based on MEO2MA and OEGMA with TEGDMA as the crosslinker. Adapted from Ref. [148] reproduced by permission of The Royal Society of Chemistry.
Figure 8. Thermosensitive nanocarriers based on MEO2MA and OEGMA with TEGDMA as the crosslinker. Adapted from Ref. [148] reproduced by permission of The Royal Society of Chemistry.
Gels 06 00020 g008
Figure 9. Temperature- and magnetic-field-responsive nanogels based on OEGMA for intracellular remote release of DOX. Adapted from Ref. [163] reproduced by permission of American Chemical Society.
Figure 9. Temperature- and magnetic-field-responsive nanogels based on OEGMA for intracellular remote release of DOX. Adapted from Ref. [163] reproduced by permission of American Chemical Society.
Gels 06 00020 g009
Figure 10. Cholesterol-bearing pullulan biodegradable nanogels for bone morphogenetic protein 2 (BMP2) and fibroblast growth factor 18 (FGF18) delivery for bone regeneration. (a) Synthesis of the acryloyl-group-bearing CHP (CHPOA)/hydrogel block and CHPOA nanogel and the chemical structure of pentaerythritol tetra (mercaptoethyl) polyoxyethylene (PEGSH). (‘R’ in CHPOA is H (glucose), cholesterol, and acryloyl). (b) Schematic diagram shows the release of FGF18 and BMP2 from cholesterol-bearing pullulan nanogels (green arrow represents the protein exchange reaction of serum albumin with FGF18 and BMP2). Adapted from Ref. [197] reproduced by permission of The Elsevier.
Figure 10. Cholesterol-bearing pullulan biodegradable nanogels for bone morphogenetic protein 2 (BMP2) and fibroblast growth factor 18 (FGF18) delivery for bone regeneration. (a) Synthesis of the acryloyl-group-bearing CHP (CHPOA)/hydrogel block and CHPOA nanogel and the chemical structure of pentaerythritol tetra (mercaptoethyl) polyoxyethylene (PEGSH). (‘R’ in CHPOA is H (glucose), cholesterol, and acryloyl). (b) Schematic diagram shows the release of FGF18 and BMP2 from cholesterol-bearing pullulan nanogels (green arrow represents the protein exchange reaction of serum albumin with FGF18 and BMP2). Adapted from Ref. [197] reproduced by permission of The Elsevier.
Gels 06 00020 g010
Table 1. Components and applications of investigated thermosensitive nanogels containing polymers bearing amide groups. “-“ means there is no therapeutic agent introduced in the reference paper and only thermosensitive drug carrier was synthesized and prepared.
Table 1. Components and applications of investigated thermosensitive nanogels containing polymers bearing amide groups. “-“ means there is no therapeutic agent introduced in the reference paper and only thermosensitive drug carrier was synthesized and prepared.
Component *Thermosensitive
Part
Size (nm)TherapeuticsApplication/PropertiesInteractionsRef.
P(NIPAM-AA)NIPAM125–325-heart repairinghydrophobic and electrostatic[106]
PTEGDA-b-
P(NIPAM-co-NMA)
NIPPAM300–480DOXthermo-responsivehydrophilic[107]
PEI-g-PNIPAMNIPAM200–350plasmid gene P53pH sensitive shell/temperature
sensitive core
ionic[108]
P(NIPAM-co-
DMAEMA-co-AFA)
NIPAM100Cisthermo-responsivehydrophobic and hydrophilic[109]
CS-NIPAM
-MAA-
NIPAM235DOXpH-/thermo-sensitive electrostatic[110]
starch-g
-PNIPAM/
Fe3O4
NIPAM67–79MTXmagnetic and
temperature
responsive
hydrophobic and hydrophilic[111]
NIPAM-
(PAMAM)
NIPAM200Mall Bdrug delivery system
against cancer cells
hydrophobic and hydrophilic[112]
(PNIPAM)NIPAM356BSAcontrolled protein
delivery
hydrophobic and hydrophilic[113]
PAMAM G3
–PNIPAM
NIPAM2005-Fuenhancing
5-fluorouracil
loading;
cancer therapy
hydrophobic and hydrophilic[114]
P(NIPPAM-AMPS)-
TEGDMA
NIPAM199–2211DOXpH-/thermo-sensitivecovalent[115]
NIPAM- (dPG)
-PANI
NIPAM155–240Anti-cancer drugefficient in-vivo photothermal cancer therapyhydrophobic and hydrophilic[79]
mPEG-NIPAM- AA-MEANIPPAM52–144DOXpH-/thermo-sensitiveelectrostatic[116]
PEDOT-NIPAMNIPPAM264Curthermo-responsiveionic[117]
PNIPAM/(SA-GO)NIPPAM75–375DOXthermo-responsiveelectrostatic[118]
Alg-NIPAMNIPPAM180DOXredox-, pH-
and thermo-sensitive
electrostatic[119]
PNIPAM-g-PEINIPAM300Toxic protein
Ricin A (RA) encoding
plasmid DNA
(pRA-EGFR)
thermo-responsivehydrophobic and hydrophilic[120]
(NIPAM-co-AA)NIPAM70–1305-FupH-/thermo-sensitivehydrophobic and hydrophilic[121]
P(NIPAM-NBD-SP)NIPAM90–130-thermo-responsivecovalent[122]
NIPAM-
PEDOT-PES
NIPPAM195–295DOXthermo-responsivehydrophobic and hydrophilic[123]
NIPAM-AA-PEGDANIPAM178–954Mtthermo-responsivehydrophobic and hydrophilic[124]
Salep-GO-NIPAMNIPAM93Df and DOXthermo-responsivehydrophobic and hydrophilic[125]
PDEAEMA-Fe3O4PDEAAM150–320-magnetic
and thermo-sensitive
electrostatic[66]
PDEAEMA - EGDMAPDEAAM160–360-pH-/thermo-sensitiveelectrostatic[126]
PAA-
b-PDEAAM
PDEAAM10–110-thermo-responsivehydrophobic and hydrophilic[92]
(DEA)/(DMA)PDEAAM165–288-thermo-responsivehydrophobic and hydrophilic[95]
(DEA)/(DMA)PDEAAM280–440-thermo-responsiveionic[93]
(PDEAAM)PDEAAM65–185-thermo-responsivehydrophobic and hydrophilic[127]
PEO-b-PDEAAMPDEAAM30–150-thermo-responsivehydrophobic and hydrophilic[94]
PDEAEMAPDEAAM200–800Coumarinthermo-responsivecovalent[128]
PVCL-PAAPVCL175–300Diclofenacthermo-responsive [129]
PVCL-Dex-MAPVCL100–400-thermo-responsivehydrophobic[130]
PVCL-PEGMAPVCL80–420-thermo-responsivehydrophilic[131]
Fib-g-PVCLPVCL150–1705-Fu and Megthermo-responsiveionic[132]
PVCLPVCL140–280-nanogel with microfiberhydrophobic and hydrophilic[104]
PVCL-co-VFA
and
P(VP-co-VFA)
PVCL70–180-pH-/thermo-sensitiveionic[133]
PDEAEMA/PVCL
Dex-MA
PVCL700–500DOX- [77]
PVCL-AGAPVCL50–1005-FupH-/thermo-sensitivehydrophobic and hydrophilic[105]
PVCL-co-IAPVCL140–360DOXpH-/thermo-sensitivehydrophobic and hydrophilic[134]
P(VCL-co-AAPBA)PVCL120–250Insulinthermo-responsivehydrophobic and hydrophilic and electrostatic[135]
PVCL-PEGDAPVCL50–120-thermo-responsivehydrophobic and hydrophilic[103]
P(AETAC-X)
- PNVCL
PVCL155–770-thermo-responsiveionic[136]
P(ODGal-
VCL-MAA)
PVCL100–190DOXredox-, pH- and thermo-responsivehydrophobic and hydrophilic[137]
Por–PEG–PCLPVCL100–250-thermo-responsiveelectrostatic attraction and hydrophobic interaction[138]
POEOMA-
b-PVCL
PVCL150–920NR as drug modelthermo-responsivehydrophobic and hydrophilic[139]
P(VCL/AAEMA
/OEGMA)
PVCL90–135-thermo-responsivehydrophobic and hydrophilic[140]
PDEGA-b-
PDMA-b-PVCL
PVCL20–400-thermo-responsivecovalent[141]
* Full names of abbreviations are available in Appendix A.
Table 2. Components and applications of investigated thermosensitive nanogels containing polymers bearing vinyl ether groups. ”-“means there is no therapeutic agent introduced in the reference paper and only thermosensitive drug carrier was synthesized and prepared.
Table 2. Components and applications of investigated thermosensitive nanogels containing polymers bearing vinyl ether groups. ”-“means there is no therapeutic agent introduced in the reference paper and only thermosensitive drug carrier was synthesized and prepared.
Component *Thermosensitive
Part
Size (nm)TherapeuticsApplication/PropertiesInteractionsRef.
ZnO-Au @PEGPEG15–57TZthermo-responsivehydrophobic and hydrophilic[166]
(PEG-b-PADMO)PEG10–80-thermo-responsivecovalent[142]
P(PEG-CPP-SA)PEG80–215DOXthermo-responsivehydrophobic and hydrophilic[167]
PEG-PPG-PEGPEG12*322-thermo-responsivehydrophobic and hydrophilic[168]
PEEP-PEG-PEEPPEG150–650DOXthermo-responsivehydrophobic and hydrophilic[169]
PEG-PLL-PLA-HAPEG160–220BSAthermo-responsivehydrophobic and hydrophilic[170]
P(MEA-co-PEGMEA)PEGMEA28–100 thermo-responsiveionic[171]
LAEMA-b-(PEGMA-co-LAEMA)PEGMA34–315Pt, BSA, BGthermo-responsivehydrophobic and hydrophilic[172]
PEGMA-CVPPEGMA85–205RhBthermo-responsiveionic[173]
PEGMA-
Maleimide-dithiol
PEGMA10–192-thermo-responsivehydrophilic[174]
Hg NPs@
P(MEO2MA
-co-OEGMA)
MEO2MA, OEGMA65Bupivacainethermo-responsivecovalent and electrostatic[161]
MEO2MA-PEGMAMEO2MA40–80-thermo-responsivehydrophilic[147]
DMDEA-
OEGMA-BADS
OEGMA17–58Paclitaxel, DOXthermo-responsivehydrophobic[159]
P[(LAEMA-MA)-b
-(DEGMA-MBAM-LAEMA)]
DEGMA60–180IAZAthermo-responsivehydrophobic[175]
MEO2MA –
OEGMA-HEMA
MEO2MA, OEGMA71–180RhB as labelthermo-responsivecovalent[148]
MEO2MA-OEGMAMEO2MA, OEGMA45DOXthermo-responsivehydrophobic and hydrophilic[176]
PCL-b-P(MEO2MA-co-OEGMA)
Mn-Zn-Fe2O4
MEO2MA, OEGMA33–129DOXtemperature and magnetic responsivehydrophobic and hydrophilic[177]
Clay/
P(MEO2MA -co- POEGMA)
MEO2MA, OEGMA200–400-nanogel/hydrogel nanocompositehydrophobic and hydrophilic[160]
MEO2MA-ChS @Carbon QDsMEO2MA125–350DOXpH-/thermo-sensitiveelectrostatic[26]
CMC-MEO2MA-OEOMA-DMAMEO2MA10DOXpH-/thermo-sensitiveelectrostatic[178]
Ag-Au @
MEO2MA-HA
MEO2MA10*60TZHA as targeting, bimetallic NP as imaginghydrophobic and hydrophilic[179]
QDs-SEMA-
PMEO2MA
MEO2MA6-smart luminescenthydrophobic and hydrophilic[180]
Ag/Au @PS-
MEO2MA-co-
MEO5MA
MEO2MA20–40Curthermo-responsivehydrophobic and hydrophilic[181]
P(MEO2MA-co-OEGMA-co-MAA)OEGMA260–650DOXtemperature and magnetic sensitivehydrophobic and hydrophilic and electrostatic [163]
dPG-OEGMA-DEGMAOEGMA50–200-thermo-responsivehydrophobic and hydrophilic[182]
HA-P(DEGMA-co-OEGMA)OEGMA150–214hydrophobic dyethermo-responsivehydrophobic and hydrophilic[183]
P(MEODEGM-
AEMA-MPC)
MEODEGM45–282insulinthermo-responsiveIonic and electrostatic [54]
* Full names of abbreviations are available in Appendix A.
Table 3. Components and applications of investigated thermosensitive nanogels containing hydrophilic polymers bearing hydrophobic groups. ”-“means there is no therapeutic agent introduced in the reference paper and only thermosensitive drug carrier was synthesized and prepared.
Table 3. Components and applications of investigated thermosensitive nanogels containing hydrophilic polymers bearing hydrophobic groups. ”-“means there is no therapeutic agent introduced in the reference paper and only thermosensitive drug carrier was synthesized and prepared.
Component *Thermosensitive
Part
Size (nm)TherapeuticsApplication/PropertiesInteractionsRef.
POP-PSPOP250–600hGHthermo-responsiveionic and hydrophobic and hydrophilic[205]
cholesterol
bearing
HP-Clu
Chl29–82-thermo-responsivehydrophobic and hydrophilic[185]
PLLA-ChS
Nisin
PLLA180–300nisintarget delivery for infection diseaseesterification[206]
Succinylated
pullulan
-g- PLLA
PLLA190–520lysozymethermo-responsiveelectrostatic and hydrophobic interactions[198]
S-Plu-g-OLLAPLLA250–450amino acidsthermo-responsiveelectrostatic[207]
Plu-g-PLLAPLLA202–341DOXthermo-responsivehydrophobic and hydrophilic[71]
Fe3O4@mSiO2-PEO-PLAPLLA85–150DOXthermo-responsiveesterification[208]
Plu-g- PLLAPLLA120–160DOXthermo-responsivehydrophobic[209]
F-127 and HepF-12750–525bFGF, HGF VEGF,
BMP-2,
thermo-responsiveionic[204]
ChS - β-GPβ-GP100–500ethosuximidethermo-responsivehydrophobic[210]
P(GME-co-EGE)P(GME-co-EGE)110–160-thermo-responsivehydrophobic[211]
F-127 and T80F-12732.5Lid and Plthermo-responsivehydrophobic and hydrophilic[193]
F-127 and HepF-127133Cisthermo-responsivehydrophobic and hydrophilic[212]
PEO-PPO-
PEO
PPO60–360Mcthermo-responsivehydrophobic and hydrophilic[213]
Bi2O3 @PVAPVA80–185TZthermo-responsivehydrophobic and hydrophilic and covalent[214]
Gel A-GAGel A60–250-thermo-responsive-[215]
HP-Clu and PMMAHP-Clu150–240-pH-/thermo-sensitivehydrophobic and hydrophilic[216]
HP-Clu- (PIA-co-PMA)HP-Clu100–610DOXpH-/thermo-sensitiveelectrostatic[217]
NAGA -DAAMNAGA50–600-thermo-responsivehydrophobic and hydrophilic[218]
P(L-Asp-co- PEG)- caprylcaprylic acid7–180-thermo-responsivehydrophobic interaction[219]
* Full names of abbreviations are available in Appendix A.

Share and Cite

MDPI and ACS Style

Ghaeini-Hesaroeiye, S.; Razmi Bagtash, H.; Boddohi, S.; Vasheghani-Farahani, E.; Jabbari, E. Thermoresponsive Nanogels Based on Different Polymeric Moieties for Biomedical Applications. Gels 2020, 6, 20. https://0-doi-org.brum.beds.ac.uk/10.3390/gels6030020

AMA Style

Ghaeini-Hesaroeiye S, Razmi Bagtash H, Boddohi S, Vasheghani-Farahani E, Jabbari E. Thermoresponsive Nanogels Based on Different Polymeric Moieties for Biomedical Applications. Gels. 2020; 6(3):20. https://0-doi-org.brum.beds.ac.uk/10.3390/gels6030020

Chicago/Turabian Style

Ghaeini-Hesaroeiye, Sobhan, Hossein Razmi Bagtash, Soheil Boddohi, Ebrahim Vasheghani-Farahani, and Esmaiel Jabbari. 2020. "Thermoresponsive Nanogels Based on Different Polymeric Moieties for Biomedical Applications" Gels 6, no. 3: 20. https://0-doi-org.brum.beds.ac.uk/10.3390/gels6030020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop