Next Article in Journal
Development of a Polyherbal Topical Gel for the Treatment of Acne
Previous Article in Journal
Origin of the Springback Effect in Ambient-Pressure-Dried Silica Aerogels: The Effect of Surface Silylation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polymer Gels: Classification and Recent Developments in Biomedical Applications

by
Mariana Chelu
and
Adina Magdalena Musuc
*
“Ilie Murgulescu” Institute of Physical Chemistry, 202 Spl. Independentei, 060021 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Gels 2023, 9(2), 161; https://doi.org/10.3390/gels9020161
Submission received: 31 January 2023 / Revised: 12 February 2023 / Accepted: 15 February 2023 / Published: 17 February 2023
(This article belongs to the Section Gel Analysis and Characterization)

Abstract

:
Polymer gels are a valuable class of polymeric materials that have recently attracted significant interest due to the exceptional properties such as versatility, soft-structure, flexibility and stimuli-responsive, biodegradability, and biocompatibility. Based on their properties, polymer gels can be used in a wide range of applications: food industry, agriculture, biomedical, and biosensors. The utilization of polymer gels in different medical and industrial applications requires a better understanding of the formation process, the factors which affect the gel’s stability, and the structure-rheological properties relationship. The present review aims to give an overview of the polymer gels, the classification of polymer gels’ materials to highlight their important features, and the recent development in biomedical applications. Several perspectives on future advancement of polymer hydrogel are offered.

1. Introduction

Polymer gels are a versatile, soft, semi-solid class of materials with an intermediate consistency between liquid and solid states. Their cross-linked network can form cavities of different shapes and sizes, in which various molecules and drugs can be trapped [1,2]. The applications of polymer gels are determined by different factors that (i) influence the formation process, (ii) influence the stabilities of gels, and (iii) the relationships between their unique structure and rheological properties. In everyday life, polymer gels are often found in food products (cakes or sausages, ketchup, cheeses), in personal use (cosmetics, shampoos, toothpaste, shaving cream), medical products (tissue engineering, coatings for medical devices, contact lenses, transdermal drug delivery, wound dressing, drug delivery systems), or in various industrial products (adhesives, paints, asphalts), in the sensor industry, and in environmental protection [3,4,5].
In 1894, van Bemmelen introduced the term “hydrogel” [6]. Later, in 1960, Wichterle and Lim described a cross-linked hydrogel based on poly (2-hydroxyethyl methacrylate) (HEMA), which can be applied to the manufacture of contact lenses, drug carriers, and to treat osteoporosis [7]. In 1980, Lim and Sun fabricated a micro capsular membrane composed of cross-linked alginate for use in cell engineering [8]. One year later, a new material for use in wound dressing was developed. This material, based on the natural polymer, collagen, drew attention to the use of polymeric hydrogels for various potential applications [9].
Polymer gels are systems formed by a polymer and a solvent in the arrangement of a three-dimensional (3D) cross-linked polymeric network. Depending on the variation of the external environment, like physical stimuli (temperature, electric and magnetic field, light, pressure) and/or chemical stimuli (pH, ionic strength, molecular species, and solvent composition), the polymer gels can discontinuously and reversibly change their volume [10,11]. Polymeric gels have the capacity to absorb a significant amount of water (tens to hundreds of times greater than the polymer itself) or biological fluids due to the existence of a hydrophilic component [12,13]. The polymer gel can swell until an equilibrium state is established between the osmotic forces and the ability to expand the polymer chains [14,15,16]. The swelling capacity of the functional polymer gels arises from their hydrophilic functional groups that are attached to polymer chains, and from the cross-links between the polymer chains. This leads to a dissolution resistance in polymer gels. Due to a “soft” intermediate state, the polymer gels display a finite shear viscosity [17,18]. This review is focused on analysis of the main characteristics of polymer gels, scrutinizing the results of the latest research, and their biomedical applications as undertaken during the last few years. From Wichterle’s revolutionary work [7] to the newest polymer gel-based developments and tenders, the present review article offers the reader a detailed overview of this area and an outlook regarding further potential developments.

2. Classification of Polymer Gels

Polymer gels can be classified in different categories:
(i) based on their sources: natural or synthetic origin (Figure 1) [15,19].
(ii) based on polymeric composition: homopolymeric, copolymeric, or multipolymeric interpenetrating polymer gels.
(iii) based on type of cross-linking: chemically or physically cross-linked [20].
(iv) based on physical appearance: amorphous (non-crystalline), semicrystalline, or crystalline.
(v) according to network electrical charge: non-ionic (neutral), ionic (anionic or cationic), amphoteric electrolyte (containing both acidic and basic groups), or zwitterionic (containing both anionic and cationic groups in each structural repeating unit).
In Figure 2 is represented the schematic illustration of the different categories used for polymer gels classification.

2.1. Classification Based on the Polymeric Composition

2.1.1. Homopolymeric Polymer Gels

These materials are obtained using a single species of monomer. Depending on the source of the monomer and the polymerization technique, their structure can be skeletal cross-linked. They can be arranged in a block, alternating, or random configuration [21].

2.1.2. Copolymeric Polymer Gels

Copolymeric gels are formed by two or more different monomers that have at least one hydrophilic part [22,23,24,25].

2.1.3. Multipolymer Interpenetrating Polymer Gels (IPN)

This polymer gels type is formed by a cross-linked polymer and a non-cross-linked synthetic and/or natural component polymer [26,27,28].

2.2. Classification Based on Type of Cross-Linking

2.2.1. Gels Physically Cross-Linked

Physical gels present multiple advantages as they are easier to be obtained and no cross-linking agents being necessary. They can be formed by a physical (hydrogen) bond, by crystallization, by ionic bonds, by self-assembly of small molecules, and by mechanical dispersion. Physical cross-linking is preferred to chemical cross-linking, when it is possible, to avoid the residual toxicity of chemical additives. Polymer gels can be obtained in the form of aerogels, cryogels, hydrogels, xerogels, nano and microgels, films, or composite materials with micro- and nanoparticles.
The molecular forces that act between the constituents of the “soft” matter depend on the size of the polymer particles and the nature of the medium in which they are dispersed. They include hydrogen bonds, intermolecular associations through van der Walls bonds, hydrophobic interactions, electrostatic interactions, polymer interchain interactions, or local crystallite formation.
Physically cross-linked gels are reversible gels, with temporary bonds between the polymer chains that appear following changes in temperature, pH, or solvent composition. They can be used in different fields including biological applications (biomedicine, drug administration, diagnostic carriers, joint replacement) or for technological applications (food additives, fuel additives, cosmetics, detergents, lubricants, paints) [29,30,31,32,33,34,35].

2.2.2. Gels Chemically Cross-Linked

Chemical gels are formed by the covalent cross-linking of existing polymer chains, which ensures a permanent bond between them [36]. Chemically cross-linked gels are also called irreversible gels. They are usually obtained by four methods:
(i) cross-linking by polymerization, which can be done by addition, condensation, photopolymerization, with free radicals, with electromagnetic radiation, and with plasma.
(ii) polymerization by condensation.
(iii) addition polymerization.
(iv) cross-linking of the polymer chain in random or end-linking processes.
The advantages of polymer gels obtained by addition and condensation are caused by a multifunctional cross-linking agent reacting with the monomer units, thereby initiating the development of the chain. The polymer gels produced in the presence of electromagnetic radiation have the advantage that they can be made at room temperature and physiological pH, even without the addition of a cross-linking agent.
The gels obtained by anionic or cationic polymerization are sensitive to water and, therefore, their use has the disadvantage that they are limited to non-polar monomers, not being able to obtain hydrogels. The degree and type of cross-linking can induce changes in some properties of the network, such as swelling, elasticity, and transport properties [29,37,38,39,40,41].
The types of “bonds” that are formed and the cross-linking methods used establish the physicochemical characteristics of the polymer gels. They have distinct advantages and disadvantages. Table 1 shows the main advantages and disadvantages of physical and chemical cross-linking.

2.2.3. Gels Cross-Linked by Ionizing Radiation

Ionizing radiation is a useful, effective, and clean tool for obtaining polymer gels for biomedical applications [42,43,44,45]. The main advantage of this process is the efficacy of the ionizing radiation at room temperature, and its ability to process any kind of physical material. Using this technique, no residual toxic chemical reagents remain in the final product. Moreover, polymer gels can be sterilized with the cross-linked process at the same time.

2.3. Classification Based on the Source of the Used Precursor

2.3.1. Synthetic Gels

Synthetic gels show adaptable mechanical and degradation characteristics and have multiple applications in engineering and materials science [46,47,48]. This type of polymer is often used in regenerative medicine, bioprinting, energy storage, or drug delivery. Some of the most common polymer gels are based on PEG (poly(ethylene glycol)) [49], PVA (poly(vinyl alcohol)) [50], PMMA (poly(methyl methacrylate)), PHEMA (poly(hydroxyethylmethyl acrylate)) [51,52], polyurethanes, poly(amino acids), and PVP (poly(vinyl pyrrolidone)) [53].
Synthetic polymer gels are specifically designed to imitate biopolymers from organic forms. They have been configured and developed with well-designed functions for various applications, including industrial ones. Thus, both “stimuli-sensitive” and “environmentally sensitive polymers” can be found. They could have the ability to respond to insignificant changes in the environment. The capability of these polymers can be seen in both the rapid changes of their structure and in the reversibility of the phase transitions. These transitions can be of different types, like sol-gel transitions, changes in solubility, shape, surface properties, or the formation of complex gels [54,55].

2.3.2. Natural Polymer Gels

Natural polymer gels contain biopolymers like polysaccharides (xanthan gum, alginate, starch, chitosan), proteins (fibrin, collagen, or gelatin) or polynucleotides, that are found in living organic systems as main components [56,57,58,59]. Bio-based polymer gels have unique and diverse characteristics, the most important being the biodegradability and biocompatibility compared to synthetic hydrogels. Due to their versatile properties, these polymeric gels have led to a significant and growing interest in the field, due to their connection with the natural environment and new and attractive functionalities.
To make gels with different shapes and structures, several types of gelators with different molecular weights can be combined to obtain supramolecular hybrid hydrogels [60].
The mixture between natural and synthetic polymers leads to hybrid hydrogels. These are functionalized materials with unique attributes capable of incorporating the benefits of both types of included polymers, like biodegradability, a good control of rigidity, viscosity, and high strength [61,62].
A schematic representation of various type of polymeric hydrogels is illustrated in Figure 3 [63].

3. Type of Stimuli-Responsive Polymer Hydrogels

Polymer hydrogel can be considered a smart material in relation to multiple application. It can be synthesized to respond to different stimuli in the human body, like ionic strength, pH, and/or temperature. These triggered mechanisms can be used to release drugs or bioactive compounds. A schematic representation of phase transition of a polymer hydrogels in response to different stimuli is given in Figure 4 [64].

3.1. Thermoresponsive Polymer Hydrogels

It was demonstrated that a small change in temperature can affect the equilibrium between hydrophobic and hydrophilic polymer segments by causing a sol-gel phase transition [65]. These kinds of polymer hydrogels are a category of the supramolecular hydrogels that are transformed in gels through hydrophobic interactions. Due to the property to form gel at higher temperatures and to return to a liquid state at lower temperatures, they can be used as biocompatible injectable thermogels [66].
Lee et al. [67] demonstrated that the administration by subcutaneous injection of human C-peptide conjugated with an elastin-like biopolymer (K9-C-Peptide) develops a hydrogel depot that can slowly release the human C-protein into the circulatory system over 19 days. It was demonstrated the long-term influence on hyperglycemia-induced vascular dysfunction by applying an aortic endothelium prototype in diabetic mice.
Dong et al. [68] synthesized an injectable thermo-sensitive chitosan hydrogel that has been incorporated into a 3D-printed poly(ε-caprolactone) (PCL)-based scaffold in order to create a hybrid scaffold. The obtained materials maintained durable compressive strength and provided a promising micro-environment useful for cell growth and in osteogenesis. The 3D scaffold can be further used as a possible bone defect repair for in vivo and subchondral applications.
Cao et al. [69] reported the first development of a transdermal hydrogel made of 5-aminolevulinic acid for use in photodynamic therapy for skin disease. Two triblock copolymers–poly(d,l-lactide-co-glycolide)-b-poly(ethylene glycol)-b-poly(d,l-lactide-co-glycolide) of different block lengths are produced by changing the blending ratio and using both sol–gel transition and gel–sol transition of a thermogelling arrangement. This research has proved that the formation of the “block blend” biomaterials is possible and this suggests further development of more intelligent drug delivery systems.

3.2. pH-Responsive Polymer Hydrogels

Polymer hydrogels, which are pH-responsive, are a group of biomaterials that were bonded with polymer chain acidic or basic groups, and which demonstrate suitable physical or/and chemical properties in a certain pH domain. The importance of pH-responsive polymer hydrogels is due to their swelling reaction in response to the pH of various body organs, the digestive system, or fluids [70,71,72]. The variations of pH in different parts of the digestive system are very important. In this regard, the pH-sensitive profile of the marine polysaccharide fucoidan–chitosan (FUC-CS) system prevents degradation under acidic gastric conditions and ensures an efficient drug absorption in the intestine [72,73].
Suhail et al. have developed pH-responsive hydrogels of carbopol, chondroitin sulphate, and polyvinyl alcohol using the free radical polymerization method with acrylic acid in the presence of ammonium persulphate and ethylene glycol dimethylacrylate for oral controlled drug delivery [74]. They demonstrated the capability of the developed pH-sensitive polymer hydrogel to obtain maximum swelling and drug release at two pH values (4.6 and 7.4, respectively). No cytotoxic effect was observed on human cancer cells in the colon. The pH-responsive hydrogels have the property to protect the stomach from harmful drug side effects and to preserve the drug from the acidic medium in the stomach.
Schoener et al. [75] have developed a pH-responsive polymer hydrogel based on poly(methacrylic-grafted-ethylene glycol) with various amounts of hydrophobic PMMA nanoparticles. They demonstrated the ability of the hydrogel to hold different amounts of doxorubicin, and to locally release doxorubicin in the colon for use in the treatment of colon cancers. Low pH was applied to simulate the pH of the stomach and then altered to neutral conditions to mimic the upper small intestine. No cytotoxic effect was detected using gastrointestinal tract and colon cancer cell lines, from either varying concentration or times of exposure.
Another study demonstrated that a developed pH-responsive hydrogel using acrylamide and methyl acrylic acid in the presence of N-N′-methylene bisacrylamide by free radical polymerization could be used as a useful oral site-specific release platform to deliver gastric-sensitive bioactive material to the small intestine route [76].
In Figure 5, the schematic representation of the pH-responsive polymer hydrogel, ascribed to a minimum swelling ability in a simulated gastric pH, and a significant swelling and release of theophylline as drug in the simulated intestinal pH for absorption of the loaded prototype drug, is shown.

3.3. Light and Chemical-Responsive Polymer Hydrogels

Light-responsive polymer gels are promising biomaterials with potential for use as sensors [77] or for drug delivery [78], based on their activation by light.
Anugrah et al. [79] has developed a near-infrared-responsive polymer hydrogel based on alginate cross-linked with tetrazine via the Diels–Alder reaction as a controllable drug carrier. A near-infrared sensitive indocyanine green and doxorubicin were included in the polymer hydrogel matrix through gelation. The obtained hydrogels demonstrated a controlled release profile under simulated physiological conditions and a rapid release profile of doxorubicin under near-infrared irradiation. The near-infrared-light promoted the generation of reactive oxygen species caused by indocyanine green, which subsequently released the entrapped doxorubicin.
Wang et al. [80] has succesfuly incorporated a fluorescent carbon nanoparticle into poly(N-isopropylacrylamide-co-acrylamide) nanogels via the one-pot precipitation copolymerization method. The resultant hybrid nanogels can bring together properties for cell imaging, fluorescent temperature sensing, and near-infrared responsiveness for drug delivery systems by accelerating the drug release and by enhancing its therapeutic efficiency. Subjected to laser excitation, it was proved to light up melanoma B16F10 cells from a mouse.

4. Structure-Effect Relationship

The assessment of the relationship between the structure and its effect is extremely important for selection of specific applications (Figure 6). The swelling behaviour and mechanical properties are very important when determining a relationship between these properties and the structural parameters of polymer hydrogels.

4.1. Mechanical Properties

The cross-linked density and the water content determine the mechanical properties of a polymer gel. The mechanical properties of polymer gels are determined using rheological analysis used to measure the viscoelastic properties. When stress is applied to a sample, the response under deformation is measured.
A dermal filler hydrogel was synthesized using hyaluronic acid cross-linked with polyethylene glycol diglycidyl ether and containing calcium hydroxyapatite, glycine, and proline [60]. The physical-chemical characterization was carried out by measuring G′ (elastic modulus), G″ (viscous modulus), and tan δ (phase angle tangent). This showed similar trends under different thermal conditions. The results showed that the product was not affected by storage conditions. It highlighted that the material has a pseudoplastic behaviour (non-Newtonian shear thinning) and that the viscosity of the dermal filler decreased with the increase of the shear rate, under all the conditions in which it was tested [60]. The characteristics of this hyaluronic acid hydrogel make it recommendable for use in the cosmetic industry as a filling material for facial rejuvenation by forced injection through a needle into the tissues.
A hybrid hydrogel was made using a mixture of synthetic and natural polymer building blocks (gelatin and PAOx used as precursor materials). These polymers were chosen to use a combination of the mechanical stability of synthetic polymers with the cell-interactive properties of natural polymers [81]. Thiolated gelatin (SH-gel) was prepared and a hybrid hydrogel obtained using thiolene radical crosslinking, which ensured the interconnectivity of PAOx and gelatin precursors. Figure 7 shows the formation of the hybrid hydrogel composed by gelatin and functionalized with thiol and PAOx, which have been functionalized with alkene. The obtained material showed an increased mechanical stability as well as a thermosensitive behaviour at temperature variations around 30 °C [81].

4.2. Polymer Hydrogel Swelling Properties

The swelling behaviour of biopolymer hydrogels can be described using simulated biological fluids. The swelling properties depend on pH, ionic strength, and temperature [82]. The free and bounded water in relation to the total water content indicated the swelling properties. The water content can be established using thermal analysis methods. By knowing the swelling properties, the degree of crosslinking, the mechanical properties, and the rate of degradation can be calculated.

4.3. Porosity

Porosity has a significant role in the applications of polymer hydrogels. The swelling and release rate of drugs are influenced by the network porosity of polymer hydrogels [83]. There are several techniques that can be used to assess the polymer gels porosity, like gas absorption, optical microscopy, scanning electron microscopy, transmission electron microscopy, atomic force microscopy, and capillary flow porosity.
The high porosity of the polymer hydrogel made it very permeable to various types of drugs, making it suitable for drug delivery in controlled conditions [84,85,86,87,88,89]. In a drug delivery study, the polymer gel’s ability to release drugs in a sustained manner for a long period represents a considerable advantage, following an increase in a drug’s concentration over a long time. Both physical and chemical methodologies can be employed to increase the affinity between the polymer hydrogel matrix and the drug, and to extend the release time [90,91].

5. Applications of Polymer Gels

Polymer hydrogels possess a broad range of applications due to their distinctive patterns and their capability to be applied and to function in various environments. As a result of the water content of polymer hydrogels, they are sufficiently adaptable for use in a large variety of industrial, pharmaceutical, and biological applications. In Figure 8, some of the biomedical applications of polymer hydrogels are shown [92].
A summary of different categories of polymer gels cross-linked by physical, chemical, or irradiation methods used in biomedical applications is presented in Table 2.

5.1. Drug Delivery

In order to use polymer hydrogels as drug delivery systems, they must have the following properties: (i) a porous structure [113]; (ii) an adequate release rate [114]; (iii) the ability to protect the drug [114], and (iv) biodegradable and biocompatible [115,116,117].
For transdermal drug delivery, novel, non-aqueous, directly-compressed tablets containing drugs formulated from common solid pharmaceutical tablet excipients have been developed for use with arrays of microneedle patches in hydrogel form. These patches were prepared and tested for in vivo delivery of amoxicillin, levodopa, and levofloxacin at therapeutically significant concentrations in rats [118].
Using chitosan as a natural polymer and polyurethane containing azomethine as a synthetic polymer, biodegradable hybrid hydrogels were developed for controlled release applications of drugs like 5-fluorouracil. These hydrogels showed good drug release behaviours of 50% of 5-fluorouracil, proving themselves able to be used for this purpose [119].
For the delivery of a drug with antioxidant and anti-inflammatory properties, an injectable hydrogel depot and tissue adhesive loaded with epigallocatechin-3-gallate was prepared. It showed a good response in the sustained release of the drug and could be used as a promising treatment in tissue degeneration to improve inflammatory disorders. [120].
A new smart hydrogel was developed as a sustained release material in the form of a compressed tablet, based on natural polysaccharides isolated from the seeds of Salvia spinosa. The sustained release potential of this hydrogel was investigated for its pH-dependent and salt-sensitive swelling in two steps, both before and after the tablet was prepared in tablet form. The study on the controlled release of theophylline (<80%) from the seeds of Salvia spinosa was monitored at the pH of the gastrointestinal tract. This pH-sensitive material showed good potential for sustained and targeted drug delivery [121].
Polymer hydrogels are ideal materials to adsorb and store different types of drugs to release them in a predetermined way for a fixed period [122].
For colorectal cancer therapy, guar-chitosan dialdehyde-based hydrogels cross-linked in situ for dual drug release were synthesized. These aimed at both simultaneous chemotherapy and pain relief in colorectal cancer therapy. Hydrogels based on guar gum and chitosan-dialdehyde cross-linked in situ were prepared for controlled dual release of curcumin and aspirin. The hydrogels protected the drugs against absorption in the stomach and small intestine, showing potential as a combined therapy for colorectal cancer [123].

5.2. Wound Healing

The wound healing process comprises four stages, shown in Figure 9 [124].
In order to be efficient and to have the best performance, an ideal wound dressing requires several properties: (i) to protect the wound from infection caused by microorganisms; (ii) to be biocompatible; (iii) to have the capacity of moisture retention; (iv) to have a gas permeability; and (v) to provide a moist environment in order to reduce the formation of scar [125].
Ying et al. obtained an extracellular matrix mimic hydrogel containing collagen I and hyaluronic acid by covalent cross-linking of hyaluronic-acid-tyramine (HA-Tyr) through horseradish peroxidase and H2O2 for use as effective wound dressing [126]. The prepared hydrogel has the capacity of autonomous healing promotion by growing vascular cells and then encouraging the closure of wound.
Ding et al. developed a collagen, chitosan, and dialdehyde-terminated polyethylene glycol self-healing polymer gel based on dynamic imine bonds for wound dressing [127]. The obtained hydrogels have shown good healing capacity, thermal stability, antibacterial activity, exceptional hemostatic ability, and injectability.
Yu et al. [128] developed an injectable hydrogel of carboxymethyl chitosan with γ-polyglutamic acid and polydopamine hydrogel for antibacterial applications and the avoidance of tumor recurrence. The carboxymethyl chitosan-based hydrogel showed a good biocompatibility.

5.3. Bone Regeneration

Bone infections, trauma, or bone diseases caused by aging, fractures, cartilage damage, or bone defects, including osteoarthritis, substantially affect people’s quality of life. Bioactive materials-based hydrogels can stimulate bone regeneration by acting as a bionic extracellular matrix. Recently, biomimetic polymer hydrogel materials have gained attention in bone repair as they facilitate adhesion, proliferation, and differentiation of stem cells [129,130]. An important role in sustaining the balance of the mineral supply in the organism is played by mineral ions like copper, magnesium, calcium, and zinc. These metal ions are linked to the polymer chains in order to form efficient polymer hydrogels which can accelerate the regeneration of the bone.
A macroporous GelMA-structured hydrogel obtained by incorporating MgO nanoparticles based on thiol-ene click reactions was developed [131]. These hydrogels presented good mechanical properties and a porous structure. In vivo experiments revealed an extracellular matrix microenvironment for enhancing the osteogenic differentiation to promote bone tissue regeneration.

5.4. Cancer Treatment

As a worldwide health problem, cancer has become the primarily cause of mortality. The treatment of cancer in different stages is currently done using surgery, radiotherapy, chemotherapy, immunotherapy, and targeted molecular therapy [64]. Chemotherapy is the classic treatment and is usually used in the targeted treatment of certain types of tumors or types of cancer. The main disadvantages are the side effects (which are severe, most of the time) and the low specificity of many antitumor drugs that fail to induce the selective death of tumor cells. Due to the side effects associated with the high cytotoxicity of chemotherapeutic compounds, a significant interest has focused on the design and manufacture of a new effective system for cancer treatment. Among numerous other formulations, like films, suspensions etc., polymer hydrogels represent the most adequate drug delivery systems in cancer therapy. Temperature, pH, and ionic responsive hydrogels are efficient as drug release systems. In response to various stimuli like temperature, pH, light, magnetic, or electric fields, or enzymes, drugs are released from the smart polymer gels. In response to the side effects of anticancer therapies, sustained studies have been carried out in recent years to reduce the amount of their cardiotoxicity. Thus, to improve their biocompatibility, as well as their efficiency, significant effort has been made to develop new delivery systems as alternatives to classic cytotoxic anticancer drugs [132].
A new type of magnetic PVA gel containing nickel nanoparticles was developed through a simple one-step procedure (Figure 10) [133].
The obtained material could be used in anti-cancer drug delivery and biotechnology (Figure 11).
Gao et al. [134] developed a hybrid, injectable, thermosensitive hydrogel system for the simultaneous delivery of co-encapsulated norcantharidin nanoparticles and doxorubicin via intratumoral administration for hepatocellular carcinoma. The in vivo testing on a mice tumor model, inhibited tumor growth and angiogenesis. Recently, different thermosensitive hydrogels for localized cancer therapy were developed, based on Pluronic F127 with titanium carbide [135], poly(d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) with indocyanine green [136], chitosan and silk sericin with tegafur - protoporphyrin heterodimers [137], chitosan and hyaluronic acid with indocyanine green, imiquimod, and cyclophosphamide [138], methylcellulose with IR820 [139], and Pluronic F127 with black phosphate nanosheets and docetaxel [140].

5.5. Hygiene Products

Recently, numerous studies have been conducted on the incorporation of natural polymers like cellulose, starch, alginate, and xanthan gum to produce natural, biodegradable, non-toxic, and biocompatible superabsorbent materials [141,142,143].
As a superabsorbent biopolymer, chitosan has been intensively studied for the cosmetic industry in the manufacture and production of various sanitary products for women and children [144,145,146].
Dry hydrogels in the form of covalently cross-linked sodium carboxymethyl cellulose and hydroxyethyl cellulose films were synthesized, employing citric acid as a cross-linking agent. The films showed excellent water absorption and can be targeted as absorbent materials for personal care [147].
Two new superabsorbent hydrogels based on carboxymethyl guar cross-linked with bentonite borax and fumed silica particle reinforcement were synthesized. The incorporation of silica particles showed a positive effect on water absorption capacity, showing that the hydrogels can be used as disposable hygiene products [148].
In recent years, significant attention was focused on the production and characterization of the stability, formulation, and antimicrobial assessment of hand sanitizers to ensure their stability and efficiency.
The gel formulation as hand sanitizer must fulfil several requirements: (i) to avoid the risk of leakage; (ii) a pleasant smell; (iii) fast absorption; (iv) no reduction of the rate of alcohol (the optimal content being from 60% to 90%) [149].
Due to the significant biocompatibility of cellulose, its use as cellulose-based hydrogel is widely studied [150,151,152]. A gel hand sanitizer was manufactured utilizing silver nanoparticles as the antimicrobial agent and coated with chitosan in different concentrations as the stabilizing agent [153].

5.6. Antimicrobial Applications

Severe wound infections are one of the primary factors that cause disease, disability, and even death. To avoid infections, modern treatment is dependent on antimicrobial drugs like antibiotics, that can act to destroy the pathogens or to inhibit their growth. Antibiotic treatment frequently proves to be ineffective in destroying infections in chronic non-healing wounds due to the development of multiresistant microbes [154,155]. Antimicrobial wound dressings have drawn wide-ranging attention in the last few years.
Gupta et al. reported the fabrication of silver nanoparticles loaded in a biosynthetic bacterial cellulose hydrogel to develop a wound hydrogel dressing [156]. The nanoparticles of silver were obtained by a green procedure using an aqueous solution of curcumin and hydroxypropyl-β-cyclodextrin. The obtained biocompatible hydrogel dressings demonstrated a broad spectrum of antimicrobial activity against three pathogenic microbes that usually infect wounds: Pseudomonas aeruginosa, Staphylococcus aureus, and Candida auris. An injectable hydrogel based on poly(vinyl alcohol), silk sericin loaded with microspheres of poly(vinyl alcohol) containing gentamicin, vancomycin, and their combination was developed to accelerate the healing of burn wounds and for infection prevention. The synthesis method was based on inverse emulsion cross-linking [157]. The results have shown the synergistic antimicrobial effects of gentamicin and vancomycin against Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli. In vivo studies on a rat burn model showed a cell migration and collagen deposition that promoted the early re-epithelialization and burn wound healing.

5.7. Bio-Sensing Applications

Polymer hydrogel-based biosensors have received significant attention in recent years since they are extremely sensitive, easy to fabricate, and can be applied in a range of fields (detection of drugs, diagnosis of many diseases, and environmental domain for detecting the aqueous contaminants).
Their use in the biomedical domain is due to their resemblance to biological tissue and their biocompatibility.
The preparation of hydrogels in the form of porous cell-like films incorporating Prussian blue nanoparticles and various enzymes on electrode surfaces for electrochemical biosensing has been reported. They were made by drop-casting composite hydrogels on the surface of SPE-type screen-printed flexible electrodes. The two amperometric biosensors developed based on hydrogel composites integrating glucose oxidase and alcohol dehydrogenase showed a high sensitivity for the rapid detection of glucose and ethanol in serum. Hydrogels can be considered electrochemical biosensing platforms due to the possibility of the efficient immobilization of some enzymes and nanomaterials in their matrix for the detection of a variety of analytes [158].
A new enzymatic biosensor was prepared for the detection of trimethylamine N-oxide (TMOA) which contains three enzymes: trimethylamine N-oxide-reductase, glucose oxidase, and catalase. It presented a signal linearly dependent on the TMOA concentration in a range between 2 µM and 15 mM, with the lowest detectable concentration being 10 µM TMOA [159].
A new additive manufacturing strategy to efficiently produce layered gelatin hydrogel microfibers bonded to 3D printed thermoplastic structures of various shapes has been developed. This can be applied to optimize the preparation and 3D modeling of electrospun hydrogel fibers using cross-linked hydrophilic polymers and oligomers, for various cell cultures or bio-sensing applications [160].
Different electrochemical biosensors for monitoring microbial metabolites and biomarkers for different types of human microbiome, especially on the gastrointestinal microbiome, have been studied to improve the diagnosis and monitoring of diseases such as gastrointestinal diseases [161].
A new hybrid hydrogel material was developed using mechanochemical incorporation of 3-((7-hydroxy-4-methylcoumarin)methylene)aminophenylboronic acid into the aragose matrix. This hydrogel can selectively and rapidly detect biogenic polyamines spermine and spermidine using a fluorescence turn-on method. The obtained smart platform can be further used to measure the spermine levels in blood plasma and human urine [162].
Wang et al. [163] have produced a new, highly-sensitive, and cost-effective DNA hydrogel sensor for visually (with the naked eye) quantitative detection of miRNAs with potential uses in nucleic acid biosensing. An enzyme polysaccharide hydrogel was designed and manufactured to be targeted by β-mannosidase for delivering bovine serum albumin and lysozyme when it is subjected to β-mannosidase in vivo [164].
An important role in specific biomolecule detection is provided by the three-dimensional (3D) polymer hydrogel network structure. The cross-linked functionalization of DNA with PEG can be used for the detection of short oligonucleotides in complex media [165]. The scheme of the detection assay is presented in Figure 12.
Several protease matrix metalloproteinase responsive hydrogels have been explored as possible protein-responsive drug delivery approaches [166,167], as cancer biomarkers, for the measurement of protein biomarkers [168,169,170,171], and as enzyme-responsive polymer hydrogel for rapid detection of strains such as Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa [172,173,174,175].
Table 3 illustrates some of the latest polymer hydrogels used in possible applications.

6. Conclusions and Future Perspectives

The present review aims to provide an overview of polymer hydrogels, their classification depending on different categories, and the possible applications based on their cross-linking. The biomedical applications of polymer hydrogels are also discussed.
In summary, polymer hydrogels represent a large system made of small monomers physically or chemically cross-linked. The most representative properties, like biocompatibility and biodegradability, make them suitable for pharmaceutical, biological, biomedical, food industry, and environmental applications.
A future development can be seen in the inclusion of various nanomaterials into the polymer hydrogels matrix for use in specific target applications. This possibility can be applied to develop new stimuli-sensitive hydrogels responsive to like light, temperature, pH, and electric field.
The rapid development of the science of polymer gels is a consequence of the importance of these materials and their application in both material and biomedical domains. Nevertheless, there are still various challenging problems that must be addressed in order to fully understand their properties from a more fundamental point of view.
Due to their functional versatility that offers the possibility of their application in many differing fields, including some high-tech disciplines that present a potential for future development, polymer gels deserve sustained efforts towards future study and development.

Author Contributions

Conceptualization, M.C. and A.M.M.; methodology, M.C. and A.M.M.; data curation, M.C.; and A.M.M.; writing—original draft preparation, M.C. and A.M.M.; writing—review and editing, A.M.M.; visualization, A.M.M.; supervision, A.M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Giuffrida, S.G.; Forysiak, W.; Cwynar, P.; Szweda, R. Shaping Macromolecules for Sensing Applications—From Polymer Hydrogels to Foldamers. Polymers 2022, 14, 580. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, F.; Yao, D.; Guo, R.; Deng, L.; Dong, A.; Zhang, J. Composites of Polymer Hydrogels and Nanoparticulate Systems for Biomedical and Pharmaceutical Applications. Nanomaterials 2015, 5, 2054–2130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Smith, D.K. Molecular Gels: Structure and Dynamics; Weiss, R.G., Ed.; Royal Society of Chemistry: Cambridge, UK, 2018; pp. 300–371. [Google Scholar]
  4. Annabi, N.; Tamayol, A.; Uquillas, J.A.; Akbari, M.; Bertassoni, L.E.; Cha, C.; Camci-Unal, G.; Dokmeci, M.R.; Peppas, N.A.; Khademhosseini, A. 25th Anniversary Article: Rational Design and Applications of Hydrogels in Regenerative Medicine. Adv. Mater. 2014, 26, 85–124. [Google Scholar] [CrossRef] [PubMed]
  5. Seliktar, D. Designing Cell-Compatible Hydrogels for Biomedical Applications. Science 2012, 336, 1124–1128. [Google Scholar] [CrossRef] [PubMed]
  6. van Bemmelen, J.M. The hydrogel and the crystalline hydrate of copper oxide. Z. Anorg. Chem. 1894, 5, 466–483. [Google Scholar] [CrossRef] [Green Version]
  7. Wichterle, O.; Lím, D. Hydrophilic Gels for Biological Use. Nature 1960, 185, 117–118. [Google Scholar] [CrossRef]
  8. Lim, F.; Sun, A.M. Microencapsulated Islets as Bioartificial Endocrine Pancreas. Science 1980, 210, 908–910. [Google Scholar] [CrossRef]
  9. Yannas, I.V.; Gordon, P.L.; Huang, C.; Silver, F.H.; Burke, J.F. Crosslinked Collagen-Mucopolysaccharide Composite Materials. U.S. Patent US4280954A, 28 July 1981. [Google Scholar]
  10. Horkay, F. Polyelectrolyte Gels: A Unique Class of Soft Materials. Gels 2021, 7, 102. [Google Scholar] [CrossRef]
  11. Masuda, T.; Akimoto, A.M.; Yoshida, R. Self-Oscillating Polymer Materials. In Biomaterials Nanoarchitectonics; Ebara, M., Ed.; William Andrew Publishing, Elsevier: Amsterdam, The Netherlands, 2016; pp. 219–236. [Google Scholar] [CrossRef]
  12. Bhattacharya, S.; Shunmugam, R. Polymer based gels and their applications in remediation of dyes from textile effluents. J. Macromol. Sci. Part A 2020, 57, 907–926. [Google Scholar] [CrossRef]
  13. Lu, S.; Bo, Q.; Zhao, G.; Shaikh, A.; Dai, C. Recent advances in enhanced polymer gels for profile control and water shutoff: A review. Front. Chem. 2023, 11, 1067094. [Google Scholar] [CrossRef]
  14. Hassan, P.; Verma, G.; Ganguly, R. Soft Materials—Properties and Applications. In Functional Materials; Elsevier: Amsterdam, The Netherlands, 2012; pp. 1–59. [Google Scholar] [CrossRef]
  15. Nasution, H.; Harahap, H.; Dalimunthe, N.F.; Ginting, M.H.S.; Jaafar, M.; Tan, O.O.H.; Aruan, H.K.; Herfananda, A.L. Hydrogel and Effects of Crosslinking Agent on Cellulose-Based Hydrogels: A Review. Gels 2022, 8, 568. [Google Scholar] [CrossRef] [PubMed]
  16. Hong, W.; Liu, Z.; Suo, Z. Inhomogeneous swelling of a gel in equilibrium with a solvent and mechanical load. Int. J. Solids Struct. 2009, 46, 3282–3289. [Google Scholar] [CrossRef] [Green Version]
  17. Douglas, J.F. Weak and Strong Gels and the Emergence of the Amorphous Solid State. Gels 2018, 4, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Tanpichai, S.; Phoothong, F.; Boonmahitthisud, A. Superabsorbent cellulose-based hydrogels cross-liked with borax. Sci. Rep. 2022, 12, 8920. [Google Scholar] [CrossRef]
  19. Zhao, W.; Jin, X.; Cong, Y.; Liu, Y.; Fu, J. Degradable natural polymer hydrogels for articular cartilage tissue engineering. J. Chem. Technol. Biotechnol. 2013, 88, 327–339. [Google Scholar] [CrossRef]
  20. Park, K.R.; Nho, Y.C. Preparation and characterization by radiation of hydrogels of PVA and PVP containingAloe vera. J. Appl. Polym. Sci. 2004, 91, 1612–1618. [Google Scholar] [CrossRef]
  21. Iizawa, T.; Taketa, H.; Maruta, M.; Ishido, T.; Gotoh, T.; Sakohara, S. Synthesis of porous poly(N-isopropylacrylamide) gel beads by sedimentation polymerization and their morphology. J. Appl. Polym. Sci. 2007, 104, 842–850. [Google Scholar] [CrossRef]
  22. Hu, Y.; Wu, X.; JinRui, X. Self-Assembled Supramolecular Hydrogels Formed by Biodegradable PLA/CS Diblock Copolymers and β-Cyclodextrin for Controlled Dual Drug Delivery. Int. J. Biol. Macromol. 2018, 108, 18–23. [Google Scholar] [CrossRef]
  23. Yapar, E.A.; Ýnal, Ö. Poly(Ethylene Oxide)–Poly(Propylene Oxide)-Based Copolymers for Transdermal Drug Delivery: An Overview. Trop. J. Pharm. Res. 2012, 11, 855–866. [Google Scholar] [CrossRef] [Green Version]
  24. Sen-Britain, S.; Hicks, W.L.; Hard, R.; Gardella, J.A. Differential orientation and conformation of surface-bound keratinocyte growth factor on (hydroxyethyl)methacrylate, (hydroxyethyl)methacrylate/methyl methacrylate, and (hydroxyethyl)methacrylate/methacrylic acid hydrogel copolymers. Biointerphases 2018, 13, 06E406. [Google Scholar] [CrossRef]
  25. Lanzalaco, S.; Armelin, E. Poly(N-isopropylacrylamide) and Copolymers: A Review on Recent Progresses in Biomedical Applications. Gels 2017, 3, 36. [Google Scholar] [CrossRef]
  26. Silverstein, M.S. Interpenetrating polymer networks: So happy together? Polymer 2020, 207, 122929. [Google Scholar] [CrossRef]
  27. Singhal, R.; Gupta, K. A Review: Tailor-made Hydrogel Structures (Classifications and Synthesis Parameters). Polym. Technol. Eng. 2015, 55, 54–70. [Google Scholar] [CrossRef]
  28. Mohite, P.B.; Adhav, S.S. A hydrogels: Methods of preparation and applications. Int. J. Adv. Pharm. 2017, 6, 79–85. [Google Scholar]
  29. Chen, M.; Cui, Y.; Wang, Y.; Chang, C. Triple physically cross-linked hydrogel artificial muscles with high-stroke and high-work capacity. Chem. Eng. J. 2023, 453, 139893. [Google Scholar] [CrossRef]
  30. Tang, Z.; Liu, D.; Lyu, X.; Liu, Y.; Liu, Y.; Yang, W.; Shen, Z.; Fan, X. Ultra-stretchable ion gels based on physically cross-linked polymer networks. J. Mater. Chem. C 2022, 10, 10926–10934. [Google Scholar] [CrossRef]
  31. Mahmood, S.; Khan, N.R.; Razaque, G.; Shah, S.U.; Shahid, M.G.; Albarqi, H.A.; Alqahtani, A.A.; Alasiri, A.; Basit, H.M. Microwave-Treated Physically Cross-Linked Sodium Alginate and Sodium Carboxymethyl Cellulose Blend Polymer Film for Open Incision Wound Healing in Diabetic Animals—A Novel Perspective for Skin Tissue Regeneration Application. Pharmaceutics 2023, 15, 418. [Google Scholar] [CrossRef]
  32. Wu, M.; Chen, X.; Xu, J.; Zhang, H. Freeze-thaw and solvent-exchange strategy to generate physically cross-linked organogels and hydrogels of curdlan with tunable mechanical properties. Carbohydr. Polym. 2022, 278, 119003. [Google Scholar] [CrossRef]
  33. Guo, Y.; Wu, M.; Li, R.; Cai, Z.; Zhang, H. Thermostable physically crosslinked cryogel from carboxymethylated konjac glucomannan fabricated by freeze-thawing. Food Hydrocoll. 2022, 122, 107103. [Google Scholar] [CrossRef]
  34. Sarmah, D.; Karak, N. Physically cross-linked starch/hydrophobically-associated poly(acrylamide) self-healing mechanically strong hydrogel. Carbohydr. Polym. 2022, 289, 119428. [Google Scholar] [CrossRef]
  35. Dong, X.; Yao, F.; Jiang, L.; Liang, L.; Sun, H.; He, S.; Shi, M.; Guo, Z.; Yu, Q.; Yao, M.; et al. Facile preparation of a thermosensitive and antibiofouling physically crosslinked hydrogel/powder for wound healing. J. Mater. Chem. B 2022, 10, 2215–2229. [Google Scholar] [CrossRef] [PubMed]
  36. Caló, E.; Khutoryanskiy, V.V. Biomedical applications of hydrogels: A review of patents and commercial products. Eur. Polym. J. 2015, 65, 252–267. [Google Scholar] [CrossRef] [Green Version]
  37. Horkay, F.; Douglas, J.F. Polymer Gels: Basics, Challenges, and Perspectives. In Gels and Other Soft Amorphous Solids; American Chemical Society: Washington, DC, USA, 2018; Chapter 1; pp. 1–13. [Google Scholar] [CrossRef] [Green Version]
  38. Zhou, Y.; Chu, R.; Fan, L.; Meng, X.; Zhao, J.; Wu, G.; Li, X.; Jiang, X.; Sun, F. Study on the mechanism and performance of polymer gels by TE and PVA chemical cross-linking. J. Appl. Polym. Sci. 2022, 139, 52043. [Google Scholar] [CrossRef]
  39. Khan, R.; Zaman, M.; Salawi, A.; Khan, M.A.; Iqbal, M.O.; Riaz, R.; Ahmed, M.M.; Butt, M.H.; Alvi, M.N.; Almoshari, Y.; et al. Synthesis of chemically cross-linked pH-sensitive hydrogels for the sustained delivery of ezetimibe. Gels 2022, 8, 281. [Google Scholar] [CrossRef]
  40. Barbero, C.A.; Martínez, M.V.; Acevedo, D.F.; Molina, M.A.; Rivarola, C.R. Cross-Linked Polymeric Gels and Nanocomposites: New Materials and Phenomena Enabling Technological Applications. Macromol 2022, 2, 440–475. [Google Scholar] [CrossRef]
  41. Wang, P.; Meng, X.; Wang, R.; Yang, W.; Yang, L.; Wang, J.; Wang, D.; Fan, C. Biomaterial Scaffolds Made of Chemically Cross-Linked Gelatin Microsphere Aggregates (C-GMSs) Promote Vascularized Bone Regeneration. Adv. Healthc. Mater. 2022, 11, 2102818. [Google Scholar] [CrossRef]
  42. Zhao, W.; Dong, Z.; Zhao, L. Radiation synthesis of polyhedral oligomeric silsesquioxanes (POSS) gel polymers. Radiat. Phys. Chem. 2022, 198, 110251. [Google Scholar] [CrossRef]
  43. Sala, L.; Perecko, T.; Mestek, O.; Pinkas, D.; Homola, T.; Kočišek, J. Cisplatin-Cross-Linked DNA Origami Nanostructures for Drug Delivery Applications. ACS Appl. Nano Mater. 2022, 5, 13267–13275. [Google Scholar] [CrossRef]
  44. Siafaka, P.I.; Gündoğdu, E.A.; Cağlar, E.S.; Özgenç, E.; Gonzalez-Alvarez, M.; Gonzalez-Alvarez, I.; Okur, N.Ü. Polymer Based Gels: Recent and Future Applications in Drug Delivery Field. Curr. Drug Deliv. 2022, 19. [Google Scholar] [CrossRef]
  45. Zhang, P.; Jiang, L.; Chen, H.; Hu, L. Recent Advances in Hydrogel-Based Sensors Responding to Ionizing Radiation. Gels 2022, 8, 238. [Google Scholar] [CrossRef]
  46. Ahmad, Z.; Salman, S.; Khan, S.A.; Amin, A.; Rahman, Z.U.; Al-Ghamdi, Y.O.; Akhtar, K.; Bakhsh, E.M.; Khan, S.B. Versatility of Hydrogels: From Synthetic Strategies, Classification, and Properties to Biomedical Applications. Gels 2022, 8, 167. [Google Scholar] [CrossRef] [PubMed]
  47. Guilherme, M.; Silva, R.; Girotto, E.; Rubira, A.; Muniz, E. Hydrogels based on PAAm network with PNIPAAm included: Hydrophilic–hydrophobic transition measured by the partition of Orange II and Methylene Blue in water. Polymer 2003, 44, 4213–4219. [Google Scholar] [CrossRef]
  48. Zhang, X.; Wu, D.; Chu, C.-C. Synthesis and characterization of partially biodegradable, temperature and pH sensitive Dex–MA/PNIPAAm hydrogels. Biomaterials 2004, 25, 4719–4730. [Google Scholar] [CrossRef]
  49. Lin, C.-C.; Anseth, K.S. PEG Hydrogels for the Controlled Release of Biomolecules in Regenerative Medicine. Pharm. Res. 2009, 26, 631–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Peppas, N.A. Turbidimetric studies of aqueous poly(vinyl alcohol) solutions. Die Makromol. Chem. 1975, 176, 3433. [Google Scholar] [CrossRef]
  51. Schacht, E.H. Polymer chemistry and hydrogel systems. J. Phys. Conf. Ser. 2004, 3, 22–28. [Google Scholar] [CrossRef]
  52. Das, N. Preparation methods and properties of hydrogel: A review. Int. J. Pharm. Pharm. Sci. 2013, 5, 112–117. [Google Scholar]
  53. Benamer, S.; Mahlous, M.; Boukrif, A.; Mansouri, B.; Youcef, S.L. Synthesis and characterisation of hydrogels based on poly(vinyl pyrrolidone). Nucl. Instrum. Methods Phys. Res. Sect. B Beam Interact. Mater. At. 2006, 248, 284–290. [Google Scholar] [CrossRef]
  54. Kumara, A.; Srivastavaa, A.; Galaev, I.Y.; Mattiasson, B. Smart polymers: Physical forms and bioengineering applications. Prog. Polym. Sci. 2007, 32, 1205–1237. [Google Scholar] [CrossRef]
  55. Piras, C.C.; Slavik, P.; Smith, D.K. Self-Assembling Supramolecular Hybrid Hydrogel Beads. Angew. Chem. Int. Ed. 2020, 59, 853–859. [Google Scholar] [CrossRef] [Green Version]
  56. Mano, J.F.; Silva, G.A.; Azevedo, H.S.; Malafaya, P.B.; Sousa, R.A.; Silva, S.S.; Boesel, L.F.; Oliveira, J.M.; Santos, T.C.; Marques, A.P.; et al. Natural origin biodegradable systems in tissue engineering and regenerative medicine: Present status and some moving trends. J. R. Soc. Interface 2007, 4, 999–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Balaci, T.D.; Ozon, E.A.; Baconi, D.L.; Nitulescu, G.; Velescu, B.; Balalau, C.; Paunica, I.; Fita, C.A. Study on the formulation and characterization of a photoprotective cream containing a new synthetized compound. J. Mind Med. Sci. 2020, 7, 193–200. [Google Scholar] [CrossRef]
  58. Chelu, M.; Calderon-Moreno, J.; Atkinson, I.; Pandele Cusu, J.; Rusu, A.; Bratan, V.; Aricov, L.; Anastasescu, M.; Seciu-grama, A.-M.; Musuc, A.M. Green synthesis of bioinspired chitosan-ZnO-based polysaccharide gums hydrogels with propolis extract as novel functional natural biomaterials. Int. J. Biol. Macromol. 2022, 211, 410–424. [Google Scholar] [CrossRef] [PubMed]
  59. Singh, S.K.; Dhyani, A.; Juyal, D. Hydrogel: Preparation, characterization and applications. Pharma Innov. 2017, 6, 25–32. [Google Scholar]
  60. Zerbinati, N.; Capillo, M.C.; Sommatis, S.; Maccario, C.; Alonci, G.; Rauso, R.; Galadari, H.; Guida, S.; Mocchi, R. Rheological Investigation as Tool to Assess Physicochemical Stability of a Hyaluronic Acid Dermal Filler Cross-Linked with Polyethylene Glycol Diglycidyl Ether and Containing Calcium Hydroxyapatite, Glycine and L-Proline. Gels 2022, 8, 264. [Google Scholar] [CrossRef] [PubMed]
  61. Shi, Y.; Ma, C.; Peng, L.; Yu, G. Conductive “Smart” Hybrid Hydrogels with PNIPAM and Nanostructured Conductive Polymers. Adv. Funct. Mater. 2015, 25, 1219–1225. [Google Scholar] [CrossRef]
  62. Chafran, L.; Carfagno, A.; Altalhi, A.; Bishop, B. Green Hydrogel Synthesis: Emphasis on Proteomics and Polymer Particle-Protein Interaction. Polymers 2022, 14, 4755. [Google Scholar] [CrossRef]
  63. Madduma-Bandarage, U.S.K.; Madihally, S.V. Synthetic hydrogels: Synthesis, novel trends, and applications. J. Appl. Polym. Sci. 2020, 138, 50376. [Google Scholar] [CrossRef]
  64. Zhao, J.; Wang, L.; Zhang, H.; Liao, B.; Li, Y. Progress of Research in In Situ Smart Hydrogels for Local Antitumor Therapy: A Review. Pharmaceutics 2022, 14, 2028. [Google Scholar] [CrossRef] [PubMed]
  65. Bajpai, A.K.; Shukla, S.K.; Bhanu, S.; Kankane, S. Responsive polymers in controlled drug delivery. Prog. Polym. Sci. 2008, 33, 1088–1118. [Google Scholar] [CrossRef]
  66. Liow, S.S.; Dou, Q.; Kai, D.; Karim, A.A.; Zhang, K.; Xu, F.; Loh, X.J. Thermogels: In Situ Gelling Biomaterial. ACS Biomater. Sci. Eng. 2016, 2, 295–316. [Google Scholar] [CrossRef]
  67. Lee, A.-J.; Lee, Y.-J.; Jeon, H.-Y.; Kim, M.; Han, E.-T.; Park, W.S.; Hong, S.-H.; Kim, Y.-M.; Ha, K.-S. Application of elastin-like biopolymer-conjugated C-peptide hydrogel for systemic long-term delivery against diabetic aortic dysfunction. Acta Biomater. 2020, 118, 32–43. [Google Scholar] [CrossRef] [PubMed]
  68. Dong, L.; Wang, S.J.; Zhao, X.R.; Zhu, Y.F.; Yu, J.K. 3D-Printed Poly(epsilon-caprolactone) Scaffold Integrated with Cell-laden Chitosan Hydrogels for Bone Tissue Engineering. Sci. Rep. 2017, 7, 13412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Cao, D.; Chen, X.; Cao, F.; Guo, W.; Tang, J.; Cai, C.; Cui, S.; Yang, X.; Yu, L.; Su, Y.; et al. An Intelligent Transdermal Formulation of ALA-Loaded Copolymer Thermogel with Spontaneous Asymmetry by Using Temperature-Induced Sol–Gel Transition and Gel–Sol (Suspension) Transition on Different Sides. Adv. Funct. Mater. 2021, 31, 2100349. [Google Scholar] [CrossRef]
  70. Jabeen, S.; Islam, A.; Ghaffar, A.; Gull, N.; Hameed, A.; Bashir, A.; Jamil, T.; Hussain, T. Development of a novel pH sensitive silane crosslinked injectable hydrogel for controlled release of neomycin sulfate. Int. J. Biol. Macromol. 2017, 97, 218–227. [Google Scholar] [CrossRef] [PubMed]
  71. Thambi, T.; Phan, V.H.G.; Kim, S.H.; Le, T.M.D.; Duong, H.T.T.; Lee, D.S. Smart injectable biogels based on hyaluronic acid bioconjugates finely substituted with poly(β-amino ester urethane) for cancer therapy. Biomater. Sci. 2019, 7, 5424–5437. [Google Scholar] [CrossRef] [PubMed]
  72. Zohdy, K.M.; El-Sherif, R.M.; El-Shamy, A.M. Effect of pH fluctuations on the biodegradability of nanocomposite Mg-alloy in simulated bodily fluids. Chem. Pap. 2022. [Google Scholar] [CrossRef]
  73. Dubashynskaya, N.V.; Gasilova, E.R.; Skorik, Y.A. Nano-Sized Fucoidan Interpolyelectrolyte Complexes: Recent Advances in Design and Prospects for Biomedical Applications. Int. J. Mol. Sci. 2023, 24, 2615. [Google Scholar] [CrossRef]
  74. Suhail, M.; Liu, J.-Y.; Hung, M.-C.; Chiu, I.-H.; Minhas, M.U.; Wu, P.-C. Preparation, In Vitro Characterization, and Cytotoxicity Evaluation of Polymeric pH-Responsive Hydrogels for Controlled Drug Release. Pharmaceutics 2022, 14, 1864. [Google Scholar] [CrossRef]
  75. Schoener, C.A.; Hutson, H.N.; Peppas, N.A. pH-responsive hydrogels with dispersed hydrophobic nanoparticles for the oral delivery of chemotherapeutics. J. Biomed. Mater. Res. A 2013, 101, 2229–2236. [Google Scholar] [CrossRef] [Green Version]
  76. Hibbins, A.R.; Kumar, P.; Choonara, Y.E.; Kondiah, P.P.D.; Marimuthu, T.; Pillay, V.; Du Toit, L.C. Design of a Versatile pH-Responsive Hydrogel for Potential Oral Delivery of Gastric-Sensitive Bioactives. Polymers 2017, 9, 474. [Google Scholar] [CrossRef] [Green Version]
  77. Yang, S.-T.; Cao, L.; Luo, P.G.; Lu, F.; Wang, X.; Wang, H.; Meziani, M.J.; Liu, Y.; Qi, G.; Sun, Y.-P. Carbon Dots for Optical Imaging in Vivo. J. Am. Chem. Soc. 2009, 131, 11308–11309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Liu, G.; Dong, C. Photoresponsive Poly(S-(o-nitrobenzyl)-l-cysteine)-b-PEO from a l-Cysteine N-Carboxyanhydride Monomer: Synthesis, Self-Assembly, and Phototriggered Drug Release. Biomacromolecules 2012, 13, 1573–1583. [Google Scholar] [CrossRef]
  79. Anugrah, D.S.B.; Ramesh, K.; Kim, M.; Hyun, K.; Lim, K.T. Near-infrared light-responsive alginate hydrogels based on diselenide-containing cross-linkage for on demand degradation and drug release. Carbohydr. Polym. 2019, 223, 115070. [Google Scholar] [CrossRef]
  80. 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]
  81. Podevyn, A.; Van Vlierberghe, S.; Dubruel, P.; Hoogenboom, R. Design and Synthesis of Hybrid Thermo-Responsive Hydrogels Based on Poly(2-oxazoline) and Gelatin Derivatives. Gels 2022, 8, 64. [Google Scholar] [CrossRef] [PubMed]
  82. Boral, S.; Gupta, A.N.; Bohidar, H. Swelling and de-swelling kinetics of gelatin hydrogels in ethanol–water marginal solvent. Int. J. Biol. Macromol. 2006, 39, 240–249. [Google Scholar] [CrossRef] [PubMed]
  83. Salerno, A.; Borzacchiello, R.; Netti, P.A. Pore structure and swelling behavior of porous hydrogels prepared via a thermal reverse-casting technique. J. Appl. Polym. Sci. 2011, 122, 3651–3660. [Google Scholar] [CrossRef]
  84. Ahmed, E.M. Hydrogel: Preparation, characterization, and applications: A review. J. Adv. Res. 2015, 6, 105–121. [Google Scholar] [CrossRef] [Green Version]
  85. Hoare, T.R.; Kohane, D.S. Hydrogels in drug delivery: Progress and challenges. Polymer 2008, 49, 1993–2007. [Google Scholar] [CrossRef] [Green Version]
  86. Ghasemiyeh, P.; Mohammadi-Samani, S. Hydrogels as Drug Delivery Systems; Pros and Cons. Trends Pharm. Sci. 2019, 5, 7–24. [Google Scholar] [CrossRef]
  87. Sepulveda, A.F.; Kumpgdee-Vollrath, M.; Franco, M.K.; Yokaichiya, F.; de Araujo, D.R. Supramolecular structure organization and rheological properties modulate the performance of hyaluronic acid-loaded thermosensitive hydrogels as drug-delivery systems. J. Colloid Interface Sci. 2023, 630, 328–340. [Google Scholar] [CrossRef] [PubMed]
  88. Li, Q.; Ma, W.; Ma, H.; Shang, H.; Qiao, N.; Sun, X. Synthesis and Characterization of Temperature-/pH-Responsive Hydrogels for Drug Delivery. ChemistrySelect 2023, 8, e202204270. [Google Scholar] [CrossRef]
  89. Tang, Z.; Yu, M.; Mondal, A.K.; Lin, X. Porous Scaffolds Based on Polydopamine/Chondroitin Sulfate/Polyvinyl Alcohol Composite Hydrogels. Polymers 2023, 15, 271. [Google Scholar] [CrossRef] [PubMed]
  90. Barclay, T.G.; Day, C.M.; Petrovsky, N.; Garg, S. Review of polysaccharide particle-based functional drug delivery. Carbohydr. Polym. 2019, 221, 94–112. [Google Scholar] [CrossRef] [PubMed]
  91. Kim, M.; Cha, C. Integrative control of mechanical and degradation properties of in situ crosslinkable polyamine-based hydrogels for dual-mode drug release kinetics. Polymer 2018, 145, 272–280. [Google Scholar] [CrossRef]
  92. Varaprasad, K.; Raghavendra, G.M.; Jayaramudu, T.; Yallapu, M.M.; Sadiku, R. A mini review on hydrogels classification and recent developments in miscellaneous applications. Mater. Sci. Eng. C 2017, 79, 958–971. [Google Scholar] [CrossRef]
  93. Lobban, R.; Biswas, A.; Ruiz-Márquez, K.J.; Bellan, L.M. Leveraging the gel-to-sol transition of physically crosslinked thermoresponsive polymer hydrogels to enable reactions induced by lowering temperature. RSC Adv. 2022, 12, 21885–21891. [Google Scholar] [CrossRef]
  94. Grosskopf, A.K.; Mann, J.L.; Baillet, J.; Hernandez, H.L.; Autzen, A.A.A.; Yu, A.C.; Appel, E.A. Extreme Extensibility in Physically Cross-Linked Nanocomposite Hydrogels Leveraging Dynamic Polymer–Nanoparticle Interactions. Macromolecules 2022, 55, 7498–7511. [Google Scholar] [CrossRef]
  95. Rochani, A.; Agrahari, V.; Chandra, N.; Singh, O.N.; McCormick, T.J.; Doncel, G.F.; Clark, M.R.; Kaushal, G. Development and Preclinical Investigation of Physically Cross-Linked and pH-Sensitive Polymeric Gels as Potential Vaginal Contraceptives. Polymers 2022, 14, 1728. [Google Scholar] [CrossRef]
  96. Hinsenkamp, A.; Fülöp, Á.; Hricisák, L.; Pál, É.; Kun, K.; Majer, A.; Varga, V.; Lacza, Z.; Hornyák, I. Application of Injectable, Crosslinked, Fibrin-Containing Hyaluronic Acid Scaffolds for In Vivo Remodeling. J. Funct. Biomater. 2022, 13, 119. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, Z.; Cui, H.; Liu, M.; Grage, S.L.; Hoffmann, M.; Sedghamiz, E.; Wenzel Levkin, P.A. Tough, Transparent, 3D-Printable, and Self-Healing Poly(ethylene glycol)-Gel (PEGgel). Adv. Mater. 2022, 34, 2107791. [Google Scholar] [CrossRef] [PubMed]
  98. Liang, Y.; Sun, X.; Lv, Q.; Shen, Y.; Liang, H. Fully physically cross-linked hydrogel as highly stretchable, tough, self-healing and sensitive strain sensors. Polymer 2020, 210, 123039. [Google Scholar] [CrossRef]
  99. Di Palma, T.M.; Migliardini, F.; Gaele, M.F.; Corbo, P. Physically cross-linked xanthan hydrogels as solid electrolytes for Al/air batteries. Ionics 2019, 25, 4209–4217. [Google Scholar] [CrossRef]
  100. Hurst, J.; Rickmann, A.; Heider, N.; Hohenadl, C.; Reither, C.; Schatz, A.; Schnichels, S.; Januschowski, K.; Spitzer, M.S. Long-Term Biocompatibility of a Highly Viscously Thiol-Modified Cross-Linked Hyaluronate as a Novel Vitreous Body Substitute. Front. Pharmacol. 2022, 13, 817353. [Google Scholar] [CrossRef]
  101. Hosoya, N.; Nishiguchi, K.; Saito, H.; Maeda, S. Chemically cross-linked gel storage for fuel to realize evaporation suppression. Chem. Eng. J. 2022, 444, 136506. [Google Scholar] [CrossRef]
  102. Tang, C.; Brodie, P.; Brunsting, M.; Tam, K.C. Carboxylated cellulose cryogel beads via a one-step ester crosslinking of maleic anhydride for copper ions removal. Carbohydr. Polym. 2022, 242, 116397. [Google Scholar] [CrossRef]
  103. Lo, Y.-W.; Sheu, M.-T.; Chiang, W.-H.; Chiu, Y.-L.; Tu, C.-M.; Wang, W.-Y.; Wu, M.-H.; Wang, Y.-C.; Lu, M.; Ho, H.-O. In situ chemically crosslinked injectable hydrogels for the subcutaneous delivery of trastuzumab to treat breast cancer. Acta Biomater. 2019, 86, 280–290. [Google Scholar] [CrossRef]
  104. Zanata, D.D.M.; Battirola, L.C.; Gonçalves, M.D.C. Chemically cross-linked aerogels based on cellulose nanocrystals and polysilsesquioxane. Cellulose 2018, 25, 7225–7238. [Google Scholar] [CrossRef]
  105. Bukhari, S.M.H.; Khan, S.; Rehanullah, M.; Ranjha, N.M. Synthesis and Characterization of Chemically Cross-Linked Acrylic Acid/Gelatin Hydrogels: Effect of pH and Composition on Swelling and Drug Release. Int. J. Polym. Sci. 2015, 2015, 187961. [Google Scholar] [CrossRef]
  106. Kakugo, A.; Sugimoto, S.; Shikinaka, K.; Gong, J.P.; Osada, Y. Characteristics of chemically cross-linked myosin gels. J. Biomater. Sci. Polym. Ed. 2005, 16, 203–218. [Google Scholar] [CrossRef] [PubMed]
  107. Vaghi, L.; Monti, M.; Marelli, M.; Motto, E.; Papagni, A.; Cipolla, L. Photoinduced Porcine Gelatin Cross-Linking by Homobi- and Homotrifunctional Tetrazoles. Gels 2021, 7, 124. [Google Scholar] [CrossRef] [PubMed]
  108. Demeter, M.; Călina, I.; Scărișoreanu, A.; Micutz, M. E-Beam Cross-Linking of Complex Hydrogels Formulation: The Influence of Poly(Ethylene Oxide) Concentration on the Hydrogel Properties. Gels 2021, 8, 27. [Google Scholar] [CrossRef]
  109. de Oliviera, M.J.; Moreira, E.G.; Salvador, P.A.; Alcântara, M.T.; Lugão, A.B. Synthesis of polymeric gels crosslinked by ionizing radiation for treatment of cutaneous leishmaniasis. In Proceedings of the 2019 International Nuclear Atlantic Conference—INAC 2019, Santos, SP, Brazil, 21–25 October 2019. [Google Scholar]
  110. Nadtoka, O.; Kutsevol, N. Thermal analysis of cross-linked hydrogels based on PVA and D-g-PAA obtained by various methods. Mol. Cryst. Liq. Cryst. 2018, 661, 52–57. [Google Scholar] [CrossRef]
  111. Yu, Y.; Cui, S. Facile Preparation of Chemically Cross-Linked Microgels by Irradiation of Visible Light at Room Temperature. Langmuir 2009, 25, 11272–11275. [Google Scholar] [CrossRef] [PubMed]
  112. Wei, S.-M.; Pei, M.-Y.; Pan, W.-L.; Thissen, H.; Tsai, S.-W. Gelatin Hydrogels Reinforced by Absorbable Nanoparticles and Fibrils Cured In Situ by Visible Light for Tissue Adhesive Applications. Polymers 2020, 12, 1113. [Google Scholar] [CrossRef] [PubMed]
  113. Loessner, D.; Meinert, C.; Kaemmerer, E.; Martine, L.C.; Yue, K.; Levett, P.A.; Klein, T.J.; Melchels, F.P.W.; Khademhosseini, A.; Hutmacher, D.W. Functionalization, preparation and use of cell-laden gelatin methacryloyl–based hydrogels as modular tissue culture platforms. Nat. Protoc. 2016, 11, 727–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, J.; Mooney, D.J. Designing hydrogels for controlled drug delivery. Nat. Rev. Mater. 2016, 1, 16071. [Google Scholar] [CrossRef]
  115. Qiu, Y.; Park, K. Environment-sensitive hydrogels for drug delivery. Adv. Drug Deliv. Rev. 2001, 53, 321–339. [Google Scholar] [CrossRef]
  116. Gupta, P.; Vermani, K.; Garg, S. Hydrogels: From controlled release to pH-responsive drug delivery. Drug Discov. Today 2002, 7, 569–579. [Google Scholar] [CrossRef]
  117. Chai, Q.; Jiao, Y.; Yu, X. Hydrogels for Biomedical Applications: Their Characteristics and the Mechanisms behind Them. Gels 2017, 3, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. McAlister, E.; Dutton, B.; Vora, L.K.; Zhao, L.; Ripolin, A.; Zahari, D.S.Z.B.P.H.; Quinn, H.L.; Tekko, I.A.; Courtenay, A.J.; Kelly, S.A.; et al. Directly Compressed Tablets: A Novel Drug-Containing Reservoir Combined with Hydrogel-Forming Microneedle Arrays for Transdermal Drug Delivery. Adv. Healthc. Mater. 2020, 10, e2001256. [Google Scholar] [CrossRef] [PubMed]
  119. Kamaci, M.; Kaya, I. Chitosan based hybrid hydrogels for drug delivery: Preparation, biodegradation, thermal, and mechanical properties. Polym. Adv. Technol. 2023, 34, 779–788. [Google Scholar] [CrossRef]
  120. He, Z.; Luo, H.; Wang, Z.; Chen, D.; Feng, Q.; Cao, X. Injectable and tissue adhesive EGCG-laden hyaluronic acid hydrogel depot for treating oxidative stress and inflammation. Carbohydr. Polym. 2023, 299, 120180. [Google Scholar] [CrossRef]
  121. Hussain, M.A.; Bukhari, S.N.A.; Ali, A.; Haseeb, M.T.; Muhammad, G.; Sheikh, F.A.; Farid-Ul-Haq, M.; Ahmad, N. A Smart Hydrogel from Salvia spinosa Seeds: pH Responsiveness, On-off Switching, Sustained Drug Release, and Transit Detection. Curr. Drug Deliv. 2023, 20, 292–305. [Google Scholar] [CrossRef] [PubMed]
  122. Dattilo, M.; Patitucci, F.; Prete, S.; Parisi, O.I.; Puoci, F. Polysaccharide-Based Hydrogels and Their Application as Drug Delivery Systems in Cancer Treatment: A Review. J. Funct. Biomater. 2023, 14, 55. [Google Scholar] [CrossRef]
  123. Dalei, G.; Das, S.; Jena, S.R.; Jena, D.; Nayak, J.; Samanta, L. In situ crosslinked dialdehyde guar gum-chitosan Schiff-base hydrogels for dual drug release in colorectal cancer therapy. Chem. Eng. Sci. 2023, 269, 118482. [Google Scholar] [CrossRef]
  124. Brumberg, V.; Astrelina, T.; Malivanova, T.; Samoilov, A. Modern Wound Dressings: Hydrogel Dressings. Biomedicines 2021, 9, 1235. [Google Scholar] [CrossRef]
  125. Junker, J.P.E.; Kamel, R.A.; Caterson, E.J.; Eriksson, E. Clinical impact upon wound healing and inflammation in moist, wet, and dry environments. Adv. Wound Care 2013, 2, 348–356. [Google Scholar] [CrossRef] [Green Version]
  126. Ying, H.; Zhou, J.; Wang, M.; Su, D.; Ma, Q.; Lv, G.; Chen, J. In situ formed collagen-hyaluronic acid hydrogel as biomimetic dressing for promoting spontaneous wound healing. Mater. Sci. Eng. C 2019, 101, 487–498. [Google Scholar] [CrossRef]
  127. Ding, C.; Tian, M.; Feng, R.; Dang, Y.; Zhang, M. Novel Self-Healing Hydrogel with Injectable, pH-Responsive, Strain-Sensitive, Promoting Wound-Healing, and Hemostatic Properties Based on Collagen and Chitosan. ACS Biomater. Sci. Eng. 2020, 6, 3855–3867. [Google Scholar] [CrossRef] [PubMed]
  128. Yu, Y.; Zheng, X.; Liu, X.; Zhao, J.; Wang, S. Injectable carboxymethyl chitosan-based hydrogel for simultaneous anti-tumor recurrence and anti-bacterial applications. Int. J. Biol. Macromol. 2023, 230, 123196. [Google Scholar] [CrossRef] [PubMed]
  129. Chaudhuri, O.; Gu, L.; Klumpers, D.; Darnell, M.; Bencherif, S.A.; Weaver, J.C.; Huebsch, N.; Lee, H.-P.; Lippens, E.; Duda, G.N.; et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nat. Mater. 2016, 15, 326–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Xue, X.; Hu, Y.; Wang, S.; Chen, X.; Jiang, Y.; Su, J. Fabrication of physical and chemical crosslinked hydrogels for bone tissue engineering. Bioact. Mater. 2022, 12, 327–339. [Google Scholar] [CrossRef] [PubMed]
  131. Pan, H.; Gao, H.; Li, Q.; Lin, Z.; Feng, Q.; Yu, C.; Zhang, X.; Dong, H.; Chen, D.; Cao, X. Engineered macroporous hydrogel scaffolds via pickering emulsions stabilized by MgO nanoparticles promote bone regeneration. J. Mater. Chem. B 2020, 8, 6100–6114. [Google Scholar] [CrossRef]
  132. Hanf, A.; Oelze, M.; Manea, A.; Li, H.; Münzel, T.; Daiber, A. The anti-cancer drug doxorubicin induces substantial epigenetic changes in cultured cardiomyocytes. Chem. Interact. 2019, 313, 108834. [Google Scholar] [CrossRef]
  133. Li, J.; Lee, K.-P.; Gopalan, A.I. One-Step Preparation of Nickel Nanoparticle-Based Magnetic Poly(Vinyl Alcohol) Gels. Coatings 2019, 9, 744. [Google Scholar] [CrossRef] [Green Version]
  134. Gao, B.; Luo, J.; Liu, Y.; Su, S.; Fu, S.; Yang, X.; Li, B. Intratumoral Administration of Thermosensitive Hydrogel Co-Loaded with Norcantharidin Nanoparticles and Doxorubicin for the Treatment of Hepatocellular Carcinoma. Int. J. Nanomed. 2021, 16, 4073–4085. [Google Scholar] [CrossRef]
  135. Yao, J.; Zhu, C.; Peng, T.; Ma, Q.; Gao, S. Injectable and Temperature-Sensitive Titanium Carbide-Loaded Hydrogel System for Photothermal Therapy of Breast Cancer. Front. Bioeng. Biotechnol. 2021, 9, 791891. [Google Scholar] [CrossRef]
  136. Jia, Y.P.; Shi, K.; Yang, F.; Liao, J.F.; Han, R.X.; Yuan, L.P.; Hao, Y.; Pan, M.; Xiao, Y.; Qian, Z.Y.; et al. Multifunctional Nanoparticle Loaded Injectable Thermoresponsive Hydrogel as NIR Controlled Release Platform for Local Photothermal Immunotherapy to Prevent Breast Cancer Postoperative Recurrence and Metastases. Adv. Funct. Mater. 2020, 30, 2001059. [Google Scholar] [CrossRef]
  137. Zhang, Z.; Li, A.; Min, X.; Zhang, Q.; Yang, J.; Chen, G.; Zou, M.; Sun, W.; Cheng, G. An ROS-sensitive tegafur-PpIX-heterodimerloaded in situ injectable thermosensitive hydrogel for photodynamic therapy combined with chemotherapy to enhance the tegafur-based treatment of breast cancer. Biomater. Sci. 2021, 9, 221–237. [Google Scholar] [CrossRef]
  138. Mei, E.; Chen, C.; Li, C.; Ding, X.; Chen, J.; Xi, Q.; Zhou, S.; Liu, J.; Li, Z. Injectable and Biodegradable Chitosan Hydrogel-Based Drug Depot Contributes to Synergistic Treatment of Tumors. Biomacromolecules 2021, 22, 5339–5348. [Google Scholar] [CrossRef] [PubMed]
  139. Yang, X.; Gao, L.; Wei, Y.; Tan, B.; Wu, Y.; Yi, C.; Liao, J. Photothermal hydrogel platform for prevention of post-surgical tumor recurrence and improving breast reconstruction. J. Nanobiotechnol. 2021, 19, 307. [Google Scholar] [CrossRef] [PubMed]
  140. Li, R.; Shan, L.; Yao, Y.; Peng, F.; Jiang, S.; Yang, D.; Ling, G.; Zhang, P. Black phosphorus nanosheets and docetaxel micelles co-incorporated thermoreversible hydrogel for combination chemo-photodynamic therapy. Drug Deliv. Transl. Res. 2021, 11, 1133–1143. [Google Scholar] [CrossRef] [PubMed]
  141. Teli, M.D.; Mallick, A. Utilization of Waste Sorghum Grain for Producing Superabsorbent for Personal Care Products. J. Polym. Environ. 2018, 26, 1393–1404. [Google Scholar] [CrossRef]
  142. Reshma, G.; Reshmi, C.R.; Nair, S.V.; Menon, D. Superabsorbent sodium carboxymethyl cellulose membranes based on a new cross-linker combination for female sanitary napkin applications. Carbohydr. Polym. 2020, 248, 116763. [Google Scholar]
  143. Mitura, S.; Sionkowska, A.; Jaiswal, A. Biopolymers for hydrogels in cosmetics. J. Mater. Sci. Mater. Med. 2020, 31, 50. [Google Scholar]
  144. Shibly, M.M.H.; Hossain, M.A.; Hossain, M.F.; Nur, M.G.; Hossain, M.B. Development of biopolymer-based menstrual pad and quality analysis against commercial merchandise. Bull. Natl. Res. Cent. 2021, 45, 50. [Google Scholar] [CrossRef]
  145. Achmad, H.; Ramadhany, Y.F. Effectiveness of chitosan tooth paste from white shrimp (Litopenaeusvannamei) to reduce number of Streptococcus mutans in the case of early childhood caries. J. Intern. Dent. Med. Res. 2017, 10, 358–363. [Google Scholar]
  146. Gonçalves, M.M.; Lobsinger, K.L.; Carneiro, J.; Picheth, G.F.; Pires, C.; Saul, C.K.; Maluf, D.F.; Pontarolo, R. Morphological study of electrospun chitosan/poly(vinyl alcohol)/glycerol nanofibres for skin care applications. Int. J. Biol. Macromol. 2022, 194, 172–178. [Google Scholar] [CrossRef]
  147. Kang, J.; Yun, S.I. Double-Network Hydrogel Films Based on Cellulose Derivatives and κ-Carrageenan with Enhanced Mechanical Strength and Superabsorbent Properties. Gels 2023, 9, 20. [Google Scholar] [CrossRef] [PubMed]
  148. Bachra, Y.; Grouli, A.; Damiri, F.; Zhu, X.X.; Talbi, M.; Berrada, M. Synthesis, Characterization, and Swelling Properties of a New Highly Absorbent Hydrogel Based on Carboxymethyl Guar Gum Reinforced with Bentonite and Silica Particles for Disposable Hygiene Products. ACS Omega 2022, 7, 39002–39018. [Google Scholar] [CrossRef] [PubMed]
  149. Sommatis, S.; Capillo, M.C.; Maccario, C.; Rauso, R.; D’Este, E.; Herrera, M.; Castiglioni, M.; Mocchi, R.; Zerbinati, N. Antimicrobial Efficacy Assessment and Rheological Investigation of Two Different Hand Sanitizers Compared with the Standard Reference WHO Formulation 1. Gels 2023, 9, 108. [Google Scholar] [CrossRef]
  150. Mistry, P.A.; Konar, M.N.; Latha, S.; Chadha, U.; Bhardwaj, P.; Eticha, T.K. Chitosan Superabsorbent Biopolymers in Sanitary and Hygiene Applications. Int. J. Polym. Sci. 2023, 2023, 4717905. [Google Scholar] [CrossRef]
  151. Bhaladhare, S.; Das, D. Cellulose: A fascinating biopolymer for hydrogel synthesis. J. Mater. Chem. B 2022, 10, 1923–1945. [Google Scholar] [CrossRef]
  152. Mudiyanselage, T.K.; Weerasinghe, N.; Karunaratna, M.; Withanage, N. Highly porous double network hydrogel having fast responding time and high mechanical strength via emulsion template polymerization. J. Appl. Polym. Sci. 2022, 139, e53048. [Google Scholar] [CrossRef]
  153. Wulandari, I.O.; Pebriatin, B.E.; Valiana, V.; Hadisaputra, S.; Ananto, A.D.; Sabarudin, A. Green Synthesis of Silver Nanoparticles Coated by Water Soluble Chitosan and Its Potency as Non-Alcoholic Hand Sanitizer Formulation. Materials 2022, 15, 4641. [Google Scholar] [CrossRef]
  154. Wu, Y.-K.; Cheng, N.-C.; Cheng, C.-M. Biofilms in Chronic Wounds: Pathogenesis and Diagnosis. Trends Biotechnol. 2019, 37, 505–517. [Google Scholar] [CrossRef]
  155. Williams, H.; Campbell, L.; Crompton, R.A.; Singh, G.; McHugh, B.J.; Davidson, D.J.; McBain, A.J.; Cruickshank, S.M.; Hardman, M.J. Microbial Host Interactions and Impaired Wound Healing in Mice and Humans: Defining a Role for BD14 and NOD2. J. Investig. Dermatol. 2018, 138, 2264–2274. [Google Scholar] [CrossRef] [Green Version]
  156. Gupta, A.; Briffa, S.M.; Swingler, S.; Gibson, H.; Kannappan, V.; Adamus, G.; Kowalczuk, M.M.; Martin, C.; Radecka, I. Synthesis of Silver Nanoparticles Using Curcumin-Cyclodextrins Loaded into Bacterial Cellulose-Based Hydrogels for Wound Dressing Applications. Biomacromolecules 2020, 21, 1802–1811. [Google Scholar] [CrossRef]
  157. Bakadia, B.M.; Zhong, A.; Li, X.; Boni, B.O.O.; Ahmed, A.A.Q.; Souho, T.; Zheng, R.; Shi, Z.; Shi, D.; Lamboni, L.; et al. Biodegradable and injectable poly(vinyl alcohol) microspheres in silk sericin-based hydrogel for the controlled release of antimicrobials: Application to deep full-thickness burn wound healing. Adv. Compos. Hybrid Mater. 2022, 5, 2847–2872. [Google Scholar] [CrossRef]
  158. Baretta, R.; Raucci, A.; Cinti, S.; Frasconi, M. Porous hydrogel scaffolds integrating Prussian Blue nanoparticles: A versatile strategy for electrochemical (bio)sensing. Sens. Actuators B Chem. 2023, 376, 132985. [Google Scholar] [CrossRef]
  159. Waffo, A.; Mitrova, B.; Tiedemann, K.; Iobbi-Nivol, C.; Leimkühler, S.; Wollenberger, U. Electrochemical Trimethylamine N-Oxide Biosensor with Enzyme-Based Oxygen-Scavenging Membrane for Long-Term Operation under Ambient Air. Biosensors 2021, 11, 98. [Google Scholar] [CrossRef] [PubMed]
  160. Gill, E.L.; Wang, W.; Liu, R.; Huang, Y.Y.S. Additive batch electrospinning patterning of tethered gelatin hydrogel fibres with swelling-induced fibre curling. Addit. Manuf. 2020, 36, 101456. [Google Scholar] [CrossRef]
  161. Sánchez-Tirado, E.; Agüí, L.; González-Cortés, A.; Campuzano, S.; Yáñez-Sedeño, P.; Pingarrón, J.M. Electrochemical (Bio)Sensing Devices for Human-Microbiome-Related Biomarkers. Sensors 2023, 23, 837. [Google Scholar] [CrossRef]
  162. Nair, R.R.; Debnath, S.; Das, S.; Wakchaure, P.; Ganguly, B.; Chatterjee, P.B. A Highly Selective Turn-On Biosensor for Measuring Spermine/Spermidine in Human Urine and Blood. ACS Appl. Bio Mater. 2019, 2, 2374–2387. [Google Scholar] [CrossRef]
  163. Wang, H.; Wang, H.; Li, Y.; Jiang, C.; Chen, D.; Wen, Y.; Li, Z. Capillarity self-driven DNA hydrogel sensor for visual quantification of microRNA. Sens. Actuators B Chem. 2020, 313, 128036. [Google Scholar] [CrossRef]
  164. Kono, H.; Otaka, F.; Ozaki, M. Preparation and characterization of guar gum hydrogels as carrier materials for controlled protein drug delivery. Carbohydr. Polym. 2014, 111, 830–840. [Google Scholar] [CrossRef]
  165. Mazzarotta, A.; Caputo, T.; Raiola, L.; Battista, E.; Netti, P.; Causa, F. Small Oligonucleotides Detection in Three-Dimensional Polymer Network of DNA-PEG Hydrogels. Gels 2021, 7, 90. [Google Scholar] [CrossRef]
  166. Chandrawati, R. Enzyme-responsive polymer hydrogels for therapeutic delivery. Exp. Biol. Med. 2016, 241, 972–979. [Google Scholar] [CrossRef]
  167. Purcell, B.P.; Lobb, D.; Charati, M.B.; Dorsey, S.M.; Wade, R.J.; Zellars, K.N.; Doviak, H.; Pettaway, S.; Logdon, C.B.; Shuman, J.A.; et al. Injectable and bioresponsive hydrogels for on-demand matrix metalloproteinase inhibition. Nat. Mater. 2014, 13, 653–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Shohatee, D.; Keifer, J.; Schimmel, N.; Mohanty, S.; Ghosh, G. Hydrogel-based suspension array for biomarker detection using horseradish peroxidase-mediated silver precipitation. Anal. Chim. Acta 2018, 999, 132–138. [Google Scholar] [CrossRef] [PubMed]
  169. Wang, W.; Han, R.; Chen, M.; Luo, X. Antifouling Peptide Hydrogel Based Electrochemical Biosensors for Highly Sensitive Detection of Cancer Biomarker HER2 in Human Serum. Anal. Chem. 2021, 93, 7355–7361. [Google Scholar] [CrossRef]
  170. Wechsler, M.E.; Dang, H.K.H.J.; Simmonds, S.P.; Bahrami, K.; Wyse, J.M.; Dahlhauser, S.D.; Reuther, J.F.; VandeWalle, A.N.; Anslyn, E.V.; Peppas, N.A. Electrostatic and Covalent Assemblies of Anionic Hydrogel-Coated Gold Nanoshells for Detection of Dry Eye Biomarkers in Human Tears. Nano Lett. 2021, 21, 8734–8740. [Google Scholar] [CrossRef] [PubMed]
  171. Culver, H.R.; Wechsler, M.E.; Peppas, N.A. Label-Free Detection of Tear Biomarkers Using Hydrogel-Coated Gold Nanoshells in a Localized Surface Plasmon Resonance-Based Biosensor. ACS Nano 2018, 12, 9342–9354. [Google Scholar] [CrossRef]
  172. Jia, Z.; Sukker, I.; Müller, M.; Schönherr, H. Selective Discrimination of Key Enzymes of Pathogenic and Nonpathogenic Bacteria on Autonomously Reporting Shape-Encoded Hydrogel Patterns. ACS Appl. Mater. Interfaces 2018, 10, 5175–5184. [Google Scholar] [CrossRef]
  173. Kaur, K.; Chelangat, W.; Druzhinin, S.I.; Karuri, N.W.; Müller, M.; Schönherr, H. Quantitative E. coli Enzyme Detection in Reporter Hydrogel-Coated Paper Using a Smartphone Camera. Biosensors 2021, 11, 25. [Google Scholar] [CrossRef]
  174. Horcajada, J.P.; Montero, M.; Oliver, A.; Sorlí, L.; Luque, S.; Gómez-Zorrilla, S.; Benito, N.; Grau, S. Epidemiology and treatment of multidrug-resistant and extensively drug-resistant Pseudomonas aeruginosa infections. Clin. Microbiol. Rev. 2019, 32, e00031-19. [Google Scholar] [CrossRef]
  175. Jia, Z.; Gwynne, L.; Sedgwick, A.C.; Müller, M.; Williams, G.T.; Jenkins, A.T.A.; James, T.D.; Schönherr, H. Enhanced Colorimetric Differentiation between Staphylococcus aureus and Pseudomonas aeruginosa Using a Shape-Encoded Sensor Hydrogel. ACS Appl. Bio Mater. 2020, 3, 4398–4407. [Google Scholar] [CrossRef]
  176. Hu, Y.; Shin, Y.; Park, S.; Jeong, J.-P.; Kim, Y.; Jung, S. Multifunctional Oxidized Succinoglycan/Poly(N-isopropylacrylamide-co-acrylamide) Hydrogels for Drug Delivery. Polymers 2023, 15, 122. [Google Scholar] [CrossRef]
  177. de Lima, H.H.C.; Santos, G.M.; da Silva, C.T.P.; Mori, J.C.; Rinaldi, J.D.C.; Cotica, S.K.; Tambourgi, E.B.; Guilherme, M.R.; Rinaldi, A.W. Synthesis and characterization of a hydrophobic association hydrogel for drug delivery. J. Mol. Liq. 2023, 372, 120709. [Google Scholar] [CrossRef]
  178. Wu, M.; Lin, M.; Li, P.; Huang, X.; Tian, K.; Li, C. Local anesthetic effects of lidocaine-loaded carboxymethyl chitosan cross-linked with sodium alginate hydrogels for drug delivery system, cell adhesion, and pain management. J. Drug Deliv. Sci. Technol. 2023, 79, 104007. [Google Scholar] [CrossRef]
  179. Li, Y.; Yao, M.; Luo, Y.; Li, J.; Wang, Z.; Liang, C.; Qin, C.; Huang, C.; Yao, S. Polydopamine-Reinforced Hemicellulose-Based Multifunctional Flexible Hydrogels for Human Movement Sensing and Self-Powered Transdermal Drug Delivery. ACS Appl. Mater. Interfaces 2023, 15, 5883–5896. [Google Scholar] [CrossRef]
  180. Zhou, C.; Xu, R.; Han, X.; Tong, L.; Xiong, L.; Liang, J.; Sun, Y.; Zhang, X.; Fan, Y. Protocatechuic acid-mediated injectable antioxidant hydrogels facilitate wound healing. Compos. Part B Eng. 2023, 250, 110451. [Google Scholar] [CrossRef]
  181. Cao, H.; Xiang, D.; Zhou, X.; Yue, P.; Zou, Y.; Zhong, Z.; Ma, Y.; Wang, L.; Wu, S.; Ye, Q. High-strength, antibacterial, antioxidant, hemostatic, and biocompatible chitin/PEGDE-tannic acid hydrogels for wound healing. Carbohydr. Polym. 2023, 307, 120609. [Google Scholar] [CrossRef]
  182. Qin, M.; Guo, Y.; Su, F.; Huang, X.; Qian, Q.; Zhou, Y.; Pan, J. High-strength, fatigue-resistant, and fast self-healing antibacterial nanocomposite hydrogels for wound healing. Chem. Eng. J. 2023, 455, 140854. [Google Scholar] [CrossRef]
  183. Bock, N.; Forouz, F.; Hipwood, L.; Clegg, J.; Jeffery, P.; Gough, M.; van Wyngaard, T.; Pyke, C.; Adams, M.N.; Bray, L.J.; et al. GelMA, Click-Chemistry Gelatin and Bioprinted Polyethylene Glycol-Based Hydrogels as 3D Ex Vivo Drug Testing Platforms for Patient-Derived Breast Cancer Organoids. Pharmaceutics 2023, 15, 261. [Google Scholar] [CrossRef] [PubMed]
  184. Sun, J.; Wu, X.; Xiao, J.; Zhang, Y.; Ding, J.; Jiang, J.; Chen, Z.; Liu, X.; Wei, D.; Zhou, L.; et al. Hydrogel-Integrated Multimodal Response as a Wearable and Implantable Bidirectional Interface for Biosensor and Therapeutic Electrostimulation. ACS Appl. Mater. Interfaces 2023, 15, 5897–5909. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Type of materials used for synthesis of hydrogel based on their origin [15].
Figure 1. Type of materials used for synthesis of hydrogel based on their origin [15].
Gels 09 00161 g001
Figure 2. Classification of polymer gels.
Figure 2. Classification of polymer gels.
Gels 09 00161 g002
Figure 3. (a) Representation of various categories of hydrogels. (b) Preparation by cross-linking between polymer chains [63].
Figure 3. (a) Representation of various categories of hydrogels. (b) Preparation by cross-linking between polymer chains [63].
Gels 09 00161 g003
Figure 4. A graphical representation of polymer gels phase transition of representative hydrogels. (a) temperature responsive; (b) pH responsive; (c) tonic strength responsive [64].
Figure 4. A graphical representation of polymer gels phase transition of representative hydrogels. (a) temperature responsive; (b) pH responsive; (c) tonic strength responsive [64].
Gels 09 00161 g004
Figure 5. Mechanism of pH-responsive hydrogel system [76].
Figure 5. Mechanism of pH-responsive hydrogel system [76].
Gels 09 00161 g005
Figure 6. Schematic representation of structure-effect relationship of polymer gels.
Figure 6. Schematic representation of structure-effect relationship of polymer gels.
Gels 09 00161 g006
Figure 7. Schematic representation of the formation of a hybrid hydrogel [81].
Figure 7. Schematic representation of the formation of a hybrid hydrogel [81].
Gels 09 00161 g007
Figure 8. Schematic representation of biomedical applications of polymer gels [92].
Figure 8. Schematic representation of biomedical applications of polymer gels [92].
Gels 09 00161 g008
Figure 9. Stages of wound healing process [124].
Figure 9. Stages of wound healing process [124].
Gels 09 00161 g009
Figure 10. The schematic representation of simultaneous formation of the magnetic gel containing Ni nanoparticles [133].
Figure 10. The schematic representation of simultaneous formation of the magnetic gel containing Ni nanoparticles [133].
Gels 09 00161 g010
Figure 11. Images of the synthesized material (a) PVA gel; (b) PVA/Ni magnetic gel; (c) Ni-nanoparticles (NPs) in the magnetic field; (d) PVA/Ni magnetic gel in the magnetic field [133].
Figure 11. Images of the synthesized material (a) PVA gel; (b) PVA/Ni magnetic gel; (c) Ni-nanoparticles (NPs) in the magnetic field; (d) PVA/Ni magnetic gel in the magnetic field [133].
Gels 09 00161 g011
Figure 12. The mechanism of DNA-PEG hydrogels detection assay [165].
Figure 12. The mechanism of DNA-PEG hydrogels detection assay [165].
Gels 09 00161 g012
Table 1. Advantages and disadvantages of physical and chemical cross-linking.
Table 1. Advantages and disadvantages of physical and chemical cross-linking.
Type of Cross-LinkingAdvantages of Polymer GelsDisadvantages of Polymer Gels
Physical cross-linking
is a homogenous reversible gel.
is formed by molecular entanglements, H-bonding, ionic, or hydrophobic forces.
it can be dissolved by changing the environmental conditions (ionic strength, pH, temperature).
the gelation process occurs under mild conditions.
absence of any chemical cross-linking agents.
the preparation method does not use chemical modification.
is less toxic.
led to inconsistent in vivo performance.
the presence of hydrophilic and hydrophobic areas.
can easily incorporate bioactive molecules.
lower bond energy.
lower cross-linking degree.
weak viscoelastic properties.
weak bonds.
less stable against degradation.
poor mechanical properties.
Chemical cross-linking
is a non-homogeneous permanent or irreversible gel.
covalent cross-linking bonds.
it may be charged or non-charged depending on the nature of functional groups from their structure. The charged polymer gels (i) generally reveal changes in swelling at pH variation, or (ii) they can suffer changes in shape when it is subjected to an electric field.
forms strong polymer gel bonds.
satisfactory viscoelastic properties.
an increased resistance to degradation.
it is prepared using chemical modification.
it is flexible to dissolution, degradation, and chemical modification.
better mechanical properties.
it can be used in cosmetics, pharmaceuticals, medicine, food industry, and agriculture.
the presence of toxic agents in the synthesis process.
it must be washed in order to remove the residue.
Table 2. Applications of polymer gels cross-linked by physical, chemical or irradiation methods.
Table 2. Applications of polymer gels cross-linked by physical, chemical or irradiation methods.
Type of Polymer GelsGels Physically Cross-LinkedGels Chemically Cross-LinkedGels
Cross-Linked by Irradiation
ApplicationsReferences
Methylcellulose hydrogelyes__Thermoresponsive materials [93]
Nanocomposite hydrogel materials (cellulose polymers and biodegradable nanoparticles)yes__3D printing, adhesives, injectable biomaterials, and foods[94]
iota-carrageenan (Ci) phenylboronic acid functionalized hydroxylpropylmethyacrylate copolymer (PBA)-based (Ci-PBA) gelyes__Gels for contraception[95]
Hyaluronic acid (HA) cross-linked using DVS or BDDE, alone or in combination with fibrinyes__In vivo remodelling processes[96]
Gel platform based on poly(ethylene glycol) (PEG) with poly(hydroxyethyl methacrylate-acrylic co-acid)yes__Multi-functional gel for wearable electronics, soft actuators, and robotics (inclusive 3D-printing)[97]
polyvinyl alcohol (PVA), acrylic acid (AA), ammonium persulfate
(APS) and Fe3+
yes__High-performance strain sensors[98]
Xanthan hydrogels with both alkaline and acid solutions as new solid electrolytesyes__High-conductivity solid electrolytes for Al-air primary
cells
[99]
Highly viscously thiol-modified cross-linked hyaluronate (TCHA)-_yes_Clinical field (Vitreous Body Substitute)[100]
Poly(N-isopropylacrylamide) (PNIPPAm) gel with ethanol _yes_polymeric gel storage for liquid fuels[101]
Highly carboxylated cellulose nanofibril (CNF) cryogel beads using maleic anhydride (MA)_yes_Heavy metal ions (Cu (II)) removal[102]
Maleimide-modified c-polyglutamic acid (c-PGA-MA) and thiol end-functionalized 4-arm poly (ethylene glycol) (4-arm PEG-SH) hydrogel _yes_Clinical field (for the subcutaneous delivery of trastuzumab to treat breast cancer)[103]
Cellulose nanocrystals (CNCs) and
polysilsesquioxane (PSS) aerogels with a porous hybrid structure
_yes_Biomedical area (Absorbents)[104]
Acrylic Acid/Gelatin Hydrogels_yes_Study of the effect of pH and composition on swelling and drug release (pheniramine maleate used for allergy treatment was loaded as model drug)[105]
Scallop myosin with 1-ethyl-3-(3-dimethylaminoprolyl) carbodiimide hydrochloride (EDC), glutaraldehyde (GA), or transglutaminase (TG) gels_yes_Study of the effects of cross-linking on enzyme activity of myosin and of morphological features of myosin gel on the actins movement[106]
Photoinduced cross-linked porcine skin gelatin with bi- and trifunctional tetrazoles__yesApplications of polyfunctional tetrazoles in photoinduced cross-linking of biological polymers[107]
collagen poly(vinyl pyrrolidone) (PVP)-poly(ethylene oxide) (PEO) cross-linked by e-beam irradiation in an
aqueous polymeric solution
__yesDevelopment of a new class of superabsorbent hydrogels[108]
Polymeric gels (cream) with Glucantime (Sb V) and gel (cream) with silver nanoparticles__yesBiomedical for alternative treatment of cutaneous Leishmaniasis[109]
Polyvinyl alcohol (PVA) cross-linked with N,N′-methylene bis-acrylamide for the synthesis of branched polymer Dextran-graft Polyacrylaamide (D-g-PAA)__yesComparative study of thermal behaviour of the hydrogels for their further use in medicine[110]
Sodium alginate microgels modified by the partial grafting of phenol groups on the backbone, in the presence of the Ru(II) catalyst complex_yesyesApplications in
biology
[111]
Gelatin hydrogels cross-linked by γ -ray irradiation using 60Co__yesAbsorption of cationic dyes and their controlled release[112]
Table 3. Most recent biomedical applications of polymer hydrogels.
Table 3. Most recent biomedical applications of polymer hydrogels.
ApplicationPolymer HydrogelDescriptionReference
Drug deliveryoxidized succinoglycan (OSG) and a poly (N-isopropyl acrylamide-co-acrylamide) [P(NIPAM-AM)] copolymer.stimuli-responsive drug delivery systems[176]
vinyl alginate (SA), acrylamide, and the hydrophobic molecule N,N′-(disulfanediylbis(4,1-phenylene))diacrylamide (SPDAAm)controlled release of poorly water-soluble molecules[177]
lidocaine (LID) loaded with carboxymethyl chitosan (CCS) cross-linked with sodium alginate (SA) hydrogelslocal anesthetic effect[178]
nano-polydopamine-reinforced hemicellulose-based hydrogelsnext generation of flexible materials proper for health monitoring and self-administration[179]
Wound healingprotocatechuic acid (PA)-mediated carboxylated chitosan (CCS) conjugated with oxidized hyaluronic acid (OHA)antibacterial properties against common pathogens[180]
chitin/PEGDE-tannic acid (CPT) hydrogelsgood antibacterial, antioxidant, and hemostatic activities[181]
incorporated gold nanorods and Ca2+ into polyacrylic acid and polyvinyl alcoholthe obtained hydrogel dressing could remove the wound bacterial biofilm and promote infected wound healing in vivo[182]
Cancer treatmentbio-printed polyethylene glycol-derived hydrogels (PEG), functionalized with adhesion peptides (RGD, GFOGER and DYIGSR) and gelatin-derived and thiolated-gelatin crosslinked with PEG-4MAL)therapy assessment in patient-derived breast cancer organoids[183]
cross-linked chitosan-dialdehyde guar gum Schiff-base hydrogelschemotherapy and pain relief in CRC therapy[123]
Biosensingureido pyrimidinone/tyramine (Upy/Tyr) difunctionalization of gelatina bidirectional neural interface for both neural recording and therapeutic electrostimulation[184]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chelu, M.; Musuc, A.M. Polymer Gels: Classification and Recent Developments in Biomedical Applications. Gels 2023, 9, 161. https://doi.org/10.3390/gels9020161

AMA Style

Chelu M, Musuc AM. Polymer Gels: Classification and Recent Developments in Biomedical Applications. Gels. 2023; 9(2):161. https://doi.org/10.3390/gels9020161

Chicago/Turabian Style

Chelu, Mariana, and Adina Magdalena Musuc. 2023. "Polymer Gels: Classification and Recent Developments in Biomedical Applications" Gels 9, no. 2: 161. https://doi.org/10.3390/gels9020161

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