Next Article in Journal
Advances in Neuroprotection in Glaucoma: Pharmacological Strategies and Emerging Technologies
Next Article in Special Issue
Development of Scaffolds with Chitosan Magnetically Activated with Cobalt Nanoferrite: A Study on Physical-Chemical, Mechanical, Cytotoxic and Antimicrobial Behavior
Previous Article in Journal
Synthesis, Absolute Configuration, Biological Profile and Antiproliferative Activity of New 3,5-Disubstituted Hydantoins
Previous Article in Special Issue
Freeze-Dried Camelina Lipid Droplets Loaded with Human Basic Fibroblast Growth Factor-2 Formulation for Transdermal Delivery: Breaking through the Cuticle Barrier to Accelerate Deep Second-Degree Burn Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Chitosan-Based Smart Hydrogels for Colorectal Cancer Treatment

by
Urszula Piotrowska
* and
Klaudia Orzechowska
Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(10), 1260; https://doi.org/10.3390/ph17101260
Submission received: 4 September 2024 / Revised: 16 September 2024 / Accepted: 19 September 2024 / Published: 25 September 2024
(This article belongs to the Special Issue Biodegradable Polymeric Nanosystems for Drug Delivery)

Abstract

:
Despite advancements in early detection and treatment in developed countries, colorectal cancer (CRC) remains the third most common malignancy and the second-leading cause of cancer-related deaths worldwide. Conventional chemotherapy, a key option for CRC treatment, has several drawbacks, including poor selectivity and the development of multiple drug resistance, which often lead to severe side effects. In recent years, the use of polysaccharides as drug delivery systems (DDSs) to enhance drug efficacy has gained significant attention. Among these polysaccharides, chitosan (CS), a linear, mucoadhesive polymer, has shown promise in cancer treatment. This review summarizes current research on the potential applications of CS-based hydrogels as DDSs for CRC treatment, with a particular focus on smart hydrogels. These smart CS-based hydrogel systems are categorized into two main types: stimuli-responsive injectable hydrogels that undergo sol-gel transitions in situ, and single-, dual-, and multi-stimuli-responsive CS-based hydrogels capable of releasing drugs in response to various triggers. The review also discusses the structural characteristics of CS, the methods for preparing CS-based hydrogels, and recent scientific advances in smart CS-based hydrogels for CRC treatment.

1. Introduction

According to the American Cancer Society, there will be over 2 million new cancer cases and over 611 thousand cancer deaths in the US in 2024. Improved treatment and earlier detection for some cancers have led to a continued decline in cancer mortality through 2021, averting over 4 million deaths since 1991 [1].
Colorectal cancer (CRC) is the third most common malignancy worldwide, accounting for approximately 10% of all cancer cases. Despite improvements in early detection and treatment in developed countries, the lifetime risk of CRC development is around 5%. The 5-year survival rate for CRC is less than 10%, making it the second-leading cause of cancer-related deaths globally [2]. CRC-related mortality is a result of the progressive accumulation of multiple genetic and epigenetic aberrations within cells [3]. CRC usually emerges from the glandular, epithelial cells of the large intestine. Normally, new epithelial cells are replenished every 2–7 days to replace those continually shed by the passage of food, which makes intestinal epithelium the most vigorously self-renewing tissue of adult mammals [4]. Any dysregulation occurring from proliferation to differentiation stages, and even post-differentiation, may lead to uncontrolled cell division and the formation of tumors. Various factors can contribute to this dysregulation, including specific hereditary gene mutations (such as hereditary non-polyposis colorectal cancer and fibroblast activation protein) and mutagenic and carcinogenic factors (high-fat diets, excessive alcohol consumption) [5]. The human large intestine is one of the most densely populated microbial communities on Earth, so the impact of infectious factor is also very important. Studies in animal models revealed the role of several bacteria in carcinogenesis (Helicobacter pylori, Streptococcus bovis, Fusobacterium nucleatum, certain strains of Escherichia coli, and producing enterotoxins Bacteroides fragilis strains) [6,7]. What is more, colon cancer has been divided into left-sided colon cancer, which is the more common type of cancer, and right-sided colon cancer, found in the colonic spleen [8]. The differences in the gut microbiota on both the left and right side of the colon are important factors in determining the occurrence and prognosis of CRC [9]. Moreover, the treatment of the right- and left-sided colon cancer are also different [10].
The principal modes of cancer therapy are surgery, radiotherapy, and chemotherapy, either alone or in combination [11]. Currently, chemotherapy with cytostatic drugs remains the most common method for cancer treatment [12]. Chemotherapeutic drugs induce apoptosis of rapidly growing cancer cells by interfering with DNA synthesis and mitosis in a non-selective manner, while causing undesirable side effects in normal tissues that reduce patient survival [13].
An interesting approach to the targeted delivery of chemotherapeutic agents into tumors is drug delivery systems (DDSs) [14,15]. DDSs could improve the effectiveness of the therapy by controlled release, while simultaneously protecting active substances from enzymatic degradation, enhancing drug accumulation in tumor tissues, meeting immunogenicity requirements, and reducing the systemic side effects of the therapy.
Of all the approaches, hydrogels have played a significant role as a carrier in drug 66 delivery, as they are biodegradable and biocompatible materials [16]. The range of their applications has been expanding to include a variety of drug and dosage forms. In the case of CRC, hydrogels facilitate localized treatment due to their adhesive properties or ability to gel in situ in response to temperature changes, reducing systemic toxicity and improving patient outcomes. They can be designed to release drugs in response to specific stimuli, such as pH changes in the gastrointestinal tract, making them highly adaptable for colon-targeted therapies in precision oncology. Hydrogels consist of cross-linked polymer chains forming a three-dimensional network structure, which allows them to absorb substantial amounts of fluid. Due to their high water content, soft texture, and porosity, they closely mimic the characteristics of living tissues [17]. Hydrogels can be synthesized from natural or synthetic polymers. The use of natural polymers such as polysaccharides (chitosan (CS), alginate, dextran, and cellulose) has many advantages associated with their biocompatibility and biological properties [18]. These polymers can be converted into 3D hydrogel structures through physical or chemical cross-linking approaches and become promising biomaterials for tissue engineering, wound repair, and controlled delivery of the drugs. Among the above-mentioned polymers, CS, natural, polysaccharide-based polymer has been widely exploited for the delivery of drugs, peptides, protein, and genes to the colon [19].
In this paper, state-of-the-art CS-based colon-targeted hydrogel drug delivery systems that have potential applications in the treatment of CRC were reviewed.

2. Physicochemical and Biological Properties of Chitosan

CS is a linear, semi-crystalline polymer obtained by the partial deacetylation of chitin [20]. This process, which can be achieved through enzymatic or chemical methods [21,22], was first discovered by Rouget in 1859 [23]. Crustacean shells are a well-established industrial source for the production of commercially available CS. Recently, however, there has been growing interest in CS extracted from agricultural waste products, such as fungal sources. This shift is largely driven by the demand for vegan-friendly products. Moreover, fungal CS has a low polydispersity index and is biocompatible due to the absence of allergenic animal-derived proteins [24,25]. CS available on the market exists in the form of dry flakes that range in color from white to yellow, solution, and fine powder [26].
The chemical structure of CS is composed of two subunits: D-glucosamine and N-acetyl-D-glucosamine, interconnected by β-1,4-glycosidic bonds. As a result, CS is chemically known as poly(N-acetyl-2-amino-2-deoxy-D-glucopyranose) (Figure 1) [27].
Each deacetylated subunit of CS contains a primary amine group with a pKa value of approximately 6.5. The cationic charge of these amino groups is responsible for CS’s mucoadhesive effect with the negatively charged mucosal surfaces of the intestinal tract. This property enables CS-based DDSs to enhance the absorption of chemotherapeutic agents, prolong their circulation, and improve therapeutic efficacy. Additionally, protonated CS transiently opens the tight junctions among epithelial cells, increasing cancer cell permeability, which makes it an excellent carrier for the local treatment of CRC [28,29]. CS can also be metabolized in vivo by enzymes such as lysozyme through enzymatic reactions [30].
CS can be characterized by its molecular weight, typically ranging from 50 kDa to 2000 kDa [31], and its degree of deacetylation (DD), which varies from 40% to 98% [32]. These two parameters significantly influence the physicochemical and biological properties of CS and can be controlled during production. Molecular weight affects the solubility, biodegradability, and antioxidant activity of the polymer. The degree of deacetylation is directly proportional to the solubility, viscosity, crystallinity, biocompatibility, mucoadhesiveness, analgesic, antibacterial, and hemostatic activity of CS [33,34]. Among various techniques, including Fourier transform infrared spectroscopy (FTIR), nuclear magnetic resonance (NMR), and UV spectroscopy, FTIR is the simplest and primary method for evaluating the DD of CS [35]. While the ideal CS structure would have a 100% DD, achieving a high DD remains challenging, limiting the use of CS in biomedical applications.
CS is soluble in aqueous acidic solution, such as acetic acid [36], citric acid, glutamic acid [37], aspartic acid, hydrochloric acid, or lactic acid [38], at pH levels below 6.5. This solubility occurs by protonating the –NH2 groups in the glucosamine units to form R-NH3+ [39]. This protonation leads to repulsion between the positively charged macrochains, allowing water molecules to diffuse in and subsequently solvate the macromolecules [40]. CS with a low DD (around 40%) remains soluble up to a pH of 9.0, whereas highly deacetylated CS (≥85%) is soluble only up to a pH of 6.5 [41]. The addition of salts to the CS solution further affects its solubility. As the ionic strength increases, the solubility decreases. In solution, CS initially adopts an extended conformation because each positively charged deacetylated unit repels the neighboring glucosamine unit. However, the addition of an electrolyte reduces this repelling effect, leading the molecule to adopt a more random coil-like conformation. As a result, increasing the electrolyte concentration triggers a salting-out effect, causing CS to precipitate from the solution [32,42,43].
Increasing the DD increases the viscosity of CS. This can be explained by the fact that CS with different levels of deacetylation adopts different conformations in aqueous solution. Highly deacetylated CS has an extended conformation with a more flexible chain due to charge repulsion within the molecule. In contrast, CS with a low degree of deacetylation tends to have a rod-like or coiled shape, resulting from the lower charge density along the polymer chain [41].
Protonated CS can complex with various anionic molecules, forming nanoparticles (NPs) through polyelectrolyte interactions that lead to self-assembly. These hydrophilic CS NPs exhibit longer retention times in the bloodstream. Hu et al. demonstrated that chondroitin sulfate-chitosan NPs were effectively endocytosed by Caco-2 fibroblasts without causing significant cytotoxic effects, even at high concentrations [44]. In Caco-2 cells, a reversible opening of tight junctions (TJs) by CS led to a decrease in transepithelial electrical resistance and an increase in paracellular marker fluxes [45,46]. Some studies reported that CS or its derivatives induced reorganization of the actin cytoskeleton in Caco-2 cells, while others found no morphological changes in the actin cytoskeleton [29,47]. In Caco-2 cells, CS appears to activate a PKC-dependent signaling pathway that affects TJ integrity [48].
CS and its oligomers, chitooligosaccharides (COS), act as prebiotics by promoting the growth of beneficial bacteria in the colon while suppressing harmful, proinflammatory bacteria. This modulation of gut microbiota can improve microbial balance and contribute to CRC prevention [49]. For instance, Wu et al. demonstrated that COS could protect mice models from CRC by rebalancing bacterial and fungal populations, specifically reducing harmful bacteria like EscherichiaShigella, Enterococcus, and Turicibacter, and increasing beneficial microbes such as Akkermansia and Cladosporium [50]. Additionally, Calinescu et al. proposed innovative CS-based DDSs for targeted probiotic colon delivery, further highlighting the therapeutic potential of CS in gut health management [51].
Finally, the functional groups in CS, such as primary amine and hydroxyl groups, act as electron donors, facilitating CS’s cross-linking with other polymers and making it well-suited for DDSs.

3. Preparation of Chitosan-Based Hydrogels

CS hydrogels, as natural polymer hydrogels, are formed through various physicochemical interactions that result in a cross-linked network structure. The amino groups present in the CS chains play a crucial role in the formation of these hydrogels. The properties of CS hydrogels, including mechanical strength, swelling behavior, and biodegradability, can be finely tuned by manipulating several factors. These include the synthesis method, CS concentration, temperature, pH, and the duration of cross-linking. By adjusting these parameters, it is possible to customize the hydrogel to meet specific needs in various applications [52].
There are two main methods for preparing CS-based hydrogels based on their attachment mechanisms: chemical and physical cross-linking [53,54]. Chemical cross-linking involves the formation of covalent bonds between molecules, resulting in hydrogels with excellent mechanical strength. A covalently cross-linked gel is characterized by a permanent network structure due to its irreversible chemical bonds. However, most of the cross-linkers used to date are either relatively toxic or have an unclear fate in the human body, with limited data available on their biocompatibility. In contrast, physical cross-linking involves the formation of noncovalent bonds, such as van der Waals forces, hydrogen bonds, ionic bonds, and hydrophobic interactions forming “reversible” or “physical” gels [55,56].

3.1. Chemical Cross-Linking

Chemically cross-linked CS-based hydrogels, often referred to as “chemical gels,” are composed of a network of covalent bonds that link the polymer chains together. These hydrogels are formed when the multiple amino and hydroxyl functional groups on the chitosan chain interact with a cross-linking agent, resulting in a stable and more rigid three-dimensional (3D) gel structure. This covalent cross-linking imparts enhanced mechanical strength and stability to the hydrogels, making them suitable for various applications where durability and rigidity are required (Table 1) [57].
CS hydrogels have been synthesized using a variety of chemical cross-linkers, including glutaraldehyde [58] or epoxy-based molecules [59]. However, the use of these synthetic cross-linking agents often results in some degree of cytotoxicity, which can negatively affect the biocompatibility of CS-based DDSs. This limitation has prompted research into safer and more biocompatible cross-linking alternatives for biomedical applications. Examples of such biocompatible cross-linkers include genipin [60,61] or citric acid [62]. Interestingly, citric acid can form both ionic and covalent bonds with CS. This dual bonding capability enhances the stability and mechanical properties of the CS hydrogel, making citric acid a versatile and biocompatible cross-linker for various biomedical applications [63].
Another method to induce chemical cross-linking is through photocross-linking. During the process, light (usually UV or visible light) activates a photoinitiator or directly excites specific functional groups within the polymer chains, leading to the formation of covalent bonds among the chains [64]. This technique, used both in vivo and in vitro, allows for the regulation of drug release from the hydrogel based on the duration of light exposure. By adjusting the light-exposure time, researchers can precisely control the release profile of the drug, making this method highly effective for targeted and controlled drug delivery applications [65,66].
Table 1. Characterization of chemical cross-linked chitosan-based hydrogels.
Table 1. Characterization of chemical cross-linked chitosan-based hydrogels.
Chemical Cross-LinkingCharacteristics of the HydrogelsRef.
Schiff base reactionHigh stability
Self-adapting ability
pH/swelling dependence
[61,67]
Diels–Alder reactionInjectability
Self-healing
High mechanical qualities
[68]
Michael addition reactionGood mechanical characteristics
Structural stability
Good in vivo degradability
Thermal stability
[69]
Thiol-ene click chemistryImproved mechanical strength
Surface roughness
Biocompatibility
pH-sensitive, CS-based hydrogels
[70]
PhotopolymerizationBiocompatibility (enhance cell adherence, proliferation, and differentiation)
Biodegradability
Wound healing
Mechanically resilient
Elastic hydrogel
Controlled drug delivery
[71,72,73]
Graft copolymerizationHighly elastic hydrogels
Bioscaffolds
[74]

3.2. Physical Cross-Linking

Physically cross-linked CS hydrogels, formed through non-covalent interactions, exhibit remarkable mechanical strength, reduced toxicity, and the ability to reverse their gelation process. Typically, the sol-gel transition in CS hydrogels is triggered by changes in pH, ionic strength, and temperature. The characterization of CS hydrogels formed through different physical cross-linking mechanisms is presented in Table 2.
Table 2. Characterization of physically cross-linked chitosan-based hydrogels.
Table 2. Characterization of physically cross-linked chitosan-based hydrogels.
Physical Cross-LinkingCharacteristics of the HydrogelsRef.
Ionic interactionStable network structure
Enhanced mechanical strength and stability
Control over pore morphology and surface properties of the hydrogel
Low cytotoxicity
[75]
Hydrogen bonding interactionUnique shapes and mechanical properties, including elasticity and the ability to bear pressure-induced deformation
pH-sensitive, temperature-sensitive, and dual-responsiveness CS-based hydrogels
[76]
Hydrophobic interactionInduce structural modifications, including changes in porosity and surface area[77]
Electrostatic interactionDecreased degree of swelling
Increased viscoelasticity
Injectable hydrogel
[78]

3.2.1. Cross-Linking in Different pH

The basic preparation of a CS hydrogel with physical cross-links involves solubilizing the macromolecules in an acidic solution. However, both alkaline and acidic solvent systems are important for preparing physically cross-linked CS hydrogels [79].
In an acidic system, the gelation mechanism of CS is primarily driven by the deprotonation and entanglement of CS macromolecules, a process promoted by the diffusion of OH from the coagulation bath (Figure 2a).
In contrast, in an alkaline CS system (alkaline-urea aqueous solvent), a thermally induced gelation mechanism occurs, driven by the evolution of intermolecular hydrogen bond interactions. As the temperature rises, CS tends to form inter- and intramolecular hydrogen bonds among its own chains rather than interacting with OH ions. This leads to the aggregation of macromolecules due to their self-association tendency, with the degree of aggregation increasing as the protective effect of the solvent weakens. Consequently, the system transitions from a polymer solution to a three-dimensional network (Figure 2b) [80].

3.2.2. Ionotropic Gelation of Chitosan

As mentioned, ionically cross-linked CS hydrogels offer distinct advantages over covalently cross-linked hydrogels, particularly in medical and pharmaceutical applications. Due to the biocompatibility of ionic cross-linkers, these hydrogels are generally well-tolerated by the body, making them especially promising as DDSs. The use of ionic cross-linking allows for the development of CS hydrogels that are not only effective in delivering therapeutic agents but also safer and more adaptable to various biomedical applications [55,56].
Ionotropic gelation is a simple, fast, and cost-effective process performed under mild conditions, as it does not involve the use of organic solvents [81]. In counterionic gelation, CS forms gels in the presence of counterions that neutralize its positive charge, allowing for the formation of gel networks. This process is used in pharmacy to create various drug forms, such as microcapsules, NPs, and films, which can be utilized for controlled drug release [82]. The cross-linking agents are typically charged ionic entities with well-defined molecular weights that neutralize the positive charges of CS, leading to the formation of ionic bridges between CS molecules and the subsequent creation of a gel.
This technique was first reported by Calvo et al. in 1997 for the preparation of CS NPs [83]. They produced CS-based hydrogel capsules by adding a CS acid solution to a solution of counterions with stirring (Figure 3).
Numerous studies have explored various counterions for CS ionotropic gelation, including tripolyphosphate [84], hexametaphosphate [85], (Fe(CN)6)−4/(Fe(CN)6)−3 [86], alginate [87], hyaluronan [88,89], chondroitin [90], κ-carragenan [91], polyaldehydrocarbonic acid [92], sulfate (such as octyl sulfate, lauryl sulfate, hexadecyl sulfate, and cetylstearyl sulfate [32]), citrate [93], and glycerophosphate [94].

4. Smart Chitosan-Based Hydrogels for the Treatment of Colorectal Cancer

Smart hydrogels are a new family of polymers that are capable of altering their chemical and/or physical properties upon exposure to endogenous (pH, enzymes, biomolecules, oxidation-reduction) or exogenous stimuli (e.g., temperature, light, mechanical force, magnetic or electric field, or ultrasound) [95,96,97].
Smart hydrogels possess unique properties, as shown in Figure 4. They can load and protect both small hydrophilic molecules and macromolecules, offering higher stability for prolonged circulation in the bloodstream. These hydrogels can significantly improve drug-loading efficiency and bioavailability while reducing the side effects. Being a soft material, they have a higher likelihood of specific retention at the target site, with the capacity to flatten on the vascular surface and anchor at multiple points simultaneously [97].
In this review article, smart CS-based hydrogel systems have been divided into two categories: stimuli-responsive injectable hydrogels that undergo sol/gel transition in situ, and stimuli-responsive CS-based hydrogels that are able to release drugs upon various triggers for CRC therapy.

4.1. In Situ-Forming Chitosan-Based Hydrogels for Colorectal Cancer Therapy

In situ gel technology has emerged as a promising approach for localized and controlled drug delivery, particularly useful in postoperative adjuvant chemotherapy for cancer patients with residual tumors following ablation surgeries [98]. This method is also gaining traction in intraperitoneal chemotherapy, which has shown potential in treating peritoneal carcinomatosis in CRC by possibly reducing mortality rates [99]. However, the challenge remains in maintaining a high intraperitoneal-to-plasma drug concentration ratio, as it is critical for ensuring that more of the drug remains in the peritoneal cavity rather than being absorbed into the systemic circulation [100].
In situ gels, which can be injected in liquid form and solidify upon exposure to specific physiological conditions or stimuli, offer unique pharmacokinetic parameters such as controlled and prolonged drug release. This technology encapsulates drugs within a polymer network in vivo with high payload efficiency, releasing them gradually through diffusion [101]. Based on the type of cross-linking trigger, in situ gels are categorized into several types, including temperature modulation, solvent exchange, UV irradiation, ionic cross-linkage, and pH change [102]. Current research is increasingly focused on developing multi-responsive gel technology for DDSs to enhance pharmacokinetics [103].
Thermosensitive hydrogels, a type of in situ gel, are particularly promising for CRC therapy. These hydrogels can be injected directly into tumor sites, where they undergo a sol-gel transition at body temperature, acting as drug reservoirs that mimic the properties of soft tissues [104]. They biodegrade naturally over time, eliminating the need for surgical removal. This method, especially when applied intraperitoneally, provides a simpler, cost-effective, and time-saving alternative to open surgery. CS-based thermosensitive, injectable hydrogels have shown great potential in CRC therapy as carriers for the sustained delivery of drugs [105]. Additionally, they may reduce peritoneal adhesion formation, which is crucial because severe adhesions can interfere with the exposure of residual tumors to the drug, thereby diminishing the effectiveness of intraperitoneal chemotherapy [106].
Despite their potential, thermosensitive CS hydrogels face significant challenges. The inherent fragility and low mechanical strength of pure CS hydrogels are due to their high water content. The numerous hydrogen bonds among CS chains create a relatively loose three-dimensional network with larger pores, which allows drug molecules to diffuse easily, leading to a burst release and limiting the hydrogel’s broader applicability in in situ DDSs [107].
Furthermore, a major challenge in developing thermosensitive CS hydrogels has been their inability to remain in solution at physiological pH [108]. When CS solutions are neutralized with strong bases, CS tends to precipitate [109]. Moreover, CS in aqueous solution does not naturally exhibit thermosensitive properties.
The introduction of a weak base, β-glycerophosphate (β-GP), with a pKa close to that of CS (6.65 at 25 °C) [110], extends the solubility of CS to physiological pH, enabling homogeneous gelation when heating to 37 °C [111]. The gelation mechanism is attributed to the neutralization of the positively charged ammonium groups in CS, which reduces the static charge surrounding the polymer. This process is driven by the heat-induced transfer of protons from CS to β-GP, neutralizing CS and allowing attractive interchain forces to form a physical gel [112]. Additionally, the ionic interaction between CS chains and β-GP, the breaking of intrachain hydrogen bonds upon heating, enhanced hydrophobic interactions among chains after neutralization, and stronger dehydration of chains by the glycerol component of β-GP all contribute to facilitating these hydrophobic interactions (Figure 5) [108,113,114].
The time required for gelation is a critical factor in the use of CS-based hydrogels, with several factors, such as the concentration of CS and other components in the hydrogel, influencing this process. Increasing the CS/β-GP ratio accelerates the gelation process [94].
Additionally, β-GP elevates the pH to the physiological range of 7.0–7.4, preventing instant precipitation or gelation and facilitating controlled gel formation upon temperature increase [94]. The study by Taherian et al. demonstrates how commercial CS with three different molecular weights (110 kD; 166,7 kD; 250 kD) and high DD (91.6–95.6%) can be neutralized closed to physiological pH using β-GP. This process produces a liquid solution at room temperature that forms a hydrogel upon subsequent heating to body temperature. The study found that the strength of the gel network is determined by the molecular weight of the CS, with higher molecular weights resulting in stronger gels. Additionally, investigations into the retention of the hydrogel in various mediums revealed that water had the greatest affinity for maintaining the integrity of the gel network [115].
However, the application of CS/β-GP thermosensitive hydrogel has been limited by challenges such as slow gelation, weak mechanical resistance, and poor cytocompatibility. To enhance the physicochemical and biological properties of these hydrogels, sodium hydrogen carbonate (NaHCO3) can be used in combination with β-GP [116] or phosphate buffer [109] to produce high-strength, CS-based hydrogels. Notably, Assaad et al. found that the transition from sol to gel is influenced by pH sensitivity, while the gelation time is affected by temperature variations. However, as the concentration of the gelling agent increases, the gelation kinetics become less sensitive to temperature changes [109]. Moreover, some studies have developed 3D-printed thermosensitive CS hydrogels. Recently, Rahimnejad et al. prepared a CS thermosensitive hydrogel using a mix of NaHCO3 and β-GP through the FRESH bioprinting approach [117].
Yun et al. developed an injectable, thermosensitive CS-based system for 5-fluorouracil (5-FU) and cisplatin (DDP) against colorectal peritoneal carcinomatosis using β-GP (10% w/v). This novel hydrogel DDS was a fluid solution at low temperatures that transitioned into a non-flowing gel upon reaching body temperature. Intraperitoneal administration of the system inhibited tumor growth and metastasis to the liver and lung in an animal model (Table 3) [118].
Thermosensitive hydrogels are widely used in biomedical applications, often incorporating various synthetic polymers such as poly(N-isopropylacrylamide) (PNIPAM). The incorporation of natural polymers, such as CS, HA, or alginate, into PNIPAM can improve the biocompatibility of the thermosensitive hydrogel while retaining its thermo-responsive properties and enhancing its mechanical strength (Figure 6) [130]. The new system could improve the efficacy of intraperitoneal chemotherapy and reduce peritoneal adhesion formation [119].

4.2. Stimuli-Responsive Multi-Drug Chitosan-Based Hydrogels

Hydrogels have shown a potential for the delivery of orally administered drugs targeted to the colon based on pH-controlled release along the pH change in the human gastrointestinal tract [124]. The oral route of drug administration is the most preferred by patients due to its ease of use, non-invasiveness, and convenience for self-administration without the need for medical supervision. However, administering pH-sensitive drugs orally in CRC therapy presents substantial challenges. The human gastrointestinal tract is a complex system that introduces several physiological barriers to effective drug delivery [131]. The acidic environment of the stomach, with a pH typically ranging from 1 to 3, can accelerate the degradation of pH-sensitive drugs, significantly reducing their therapeutic effectiveness before they even reach the target site. Furthermore, the varying pH levels throughout different parts of the gastrointestinal tract, coupled with the presence of digestive enzymes, can further complicate the stability and absorption of these drugs [132].
To overcome these challenges, various strategies are being explored, including the use of advanced drug delivery systems such as pH-sensitive carriers. CS is well known for its pH sensitivity due to the presence of amino groups that can be protonated or deprotonated depending on the pH of the environment. However, CS-based hydrogels can lead to a burst release of the drug in the stomach due to their solubility in gastric contents. To address these limitations, pH-responsive hydrogels have been developed that release the drug after swelling at specific pH levels, thereby improving the oral bioavailability of the drug [133].
Ghobashy et al. achieved the polymerization of abundant amino groups (-NH2) on the backbone of CS with the carboxyl and sulfonic groups of two anionic polymers, acrylic acid (AA) and 2-acrylamido-2-methylpropane sulfonic acid (AMPS), through gamma irradiation cross-linking to obtain an amphiphilic, sterilized, and pH-sensitive hydrogel. The amphiphilic hydrogel was loaded with 5-FU, and the stimuli-responsive properties of the new system allowed for enhanced drug release at pH 7 (25.3% after 30 min; 96% after 7 h) compared to pH 1 (1.55% after 30 min) [125].
Zarbab and colleagues developed an effective biodegradable and biocompatible hydrogel (GG/PVA/CS) cross-linked with TEOS and incorporated with methotrexate (MTX), designed to be sensitive to pH variations. Their investigation of hydrogel variants with different polymer ratios (GG, CS, and PVA) and cross-linker concentrations revealed significant impacts on pore size, swelling behavior, and water-retention capacity. This hydrogel, obtained through a solution casting technique, shows potential for sustained drug release, with 96% methotrexate released within 7 h, enabling localized delivery of MTX to target colon cancer [126].
Recently, much research has focused on the synergistic effect of multiple drugs (such as two cytostatic, or the combination of chemotherapy and pain relief) in colorectal cancer therapy, as well as the development of multi-drug-loaded biomaterials [122,134].
Liang et al. developed MoS2 nanoflower-doped chitosan/oxidized dextran (CS/OD) hydrogels for the sequential delivery of 5-FU and methotrexate (MTX). These dual-loaded drug delivery systems release cytostatic drugs via a dual-stimuli process: near-infrared (NIR) irradiation triggers the release of 5-FU, while pH stimuli control the release of MTX. The unique, sequential delivery of these two chemotherapeutics from the hydrogels is closely related to the electrostatic attraction and the –N=CH– bonds between CS and OD. Due to the synergistic effect of the multi-drug chitosan-based hydrogels, cell viability drops to less than 10% when treated with 512 μg/mL. Moreover, cell viability further decreases to 4.9% after 30 min of NIR irradiation, which can be attributed to the effect of the synergistic, stimuli-responsive therapy [123].
Photothermal therapy (PTT) offers an alternative route for cancer treatment. This kind of therapy relies on photothermal agents with photothermal conversion capability to eliminate tumors at high temperatures, providing advantages of high precision and low toxicity [135]. However, it suffers from a lack of accumulation of photothermal agents, low photothermal conversion efficiency, and incomplete tumor elimination, leading to a very low therapeutic effect [136]. To overcome these difficulties, combined localized and dual-stimuli PTT and chemotherapy were developed. The addition of drug molecules and photothermal materials has realized multifunctional in vivo tumor PTT and chemotherapy, ensuring their in vivo biosafety [120].
Wang et al. developed an enzyme-responsive, CS-based hydrogel designed to efficiently encapsulate the hydrophobic drug imatinib (IMT) and sodium deoxycholate (a permeation enhancer). This hydrogel is intended for oral administration, where it triggers release in response to intestinal enzymes and opens epithelial tight junctions, thereby enhancing the treatment of CRC [127]. Oral administration provides an effective, non-invasive approach for the treatment of CRC.

4.3. Nanocomposite Hydrogels

Currently, many methods have been developed to enhance the mechanical strength and thermal stability of hydrogels, with nanocomposite hydrogels emerging as a particularly promising approach. By integrating hydrogels with nanocarriers, nanocomposites with superior properties and tailored functionality can be obtained [137]. Nanocarriers can significantly enhance drug delivery and minimize side effects, particularly for poorly water-soluble drugs. By reducing burst drug release effects and modulating drug release profiles, nanocarriers have become promising candidates for anti-cancer DDSs. However, a major challenge in vivo is the lack of colloidal stability of these carriers, leading to their rapid elimination from the bloodstream before reaching the target site [138]. To address this issue, many researchers are focusing on developing nanocomposite systems. A composite is a structural material made by combining two or more constituents on a macroscopic scale while preserving their individual properties. Nanocomposite systems based on CS can adsorb onto the surface of metal oxide NPs and stabilize their dispersion through electrostatic repulsion [139]. Additionally, this surface modification can enhance the physical, chemical, and biological properties of the NPs [140].
The co-administration of CS with drugs has been shown to enhance both transcellular and paracellular transport of drugs across the mucosal epithelium. CS, due to its positive charge, interacts directly with cell membranes, binding to them and decreasing the trans-epithelial electrical resistance of cell monolayers. This interaction increases paracellular permeability, facilitating drug transport across the epithelial barrier [141,142]. Research has demonstrated that CS solutions can increase transcellular and paracellular permeability in a reversible, dose-dependent manner. This effect is influenced by the molecular weight and degree of deacetylation of the CS, as well as by the pH of the surrounding environment [143].
In DDSs, CS-based NPs and CS-coated microspheres facilitate drug transport partly through endocytosis and transcytosis. Additionally, CS NPs have been shown to decrease transepithelial electrical resistance and increase the permeability of molecules like FITC-dextran, further indicating their impact on paracellular transport as well [144,145].
Recently, Sun et al. developed core–shell ZnO/CMC/CS bio-nanocomposite hydrogel beads using an electrostatic self-assembly method. These beads serve as a pH-sensitive carrier for 5-FU (Table 4). The ionic interactions between the carboxylate anions of carboxymethyl cellulose (CMC) and the cationic amine groups of CS enhance the hydrogel’s sensitivity to gastrointestinal conditions. Additionally, the incorporation of ZnO nanoparticles contributes to controlled drug release profiles, imparts antibacterial properties, and provides remarkable mechanical strength to the hydrogel system [146].
Nowadays, combinatorial therapy is gaining attention in tumor treatment due to its low side effects and the synergistic effect of enhanced cancer cell inhibition. Dhanavel and colleagues reported on CS/graphene nanocomposites, which are based on graphene nanosheets, as DDS. These nanocomposites are particularly effective because the large surface area and π-conjugated structure of graphene allow for the encapsulation of more than one therapeutic agent [150].

5. Clinical Challenges and Limitations

While CS is widely studied for its biomedical applications, including DDSs and tissue engineering, the use of CS-based hydrogels in CRC treatment remains in the early, preclinical stages. There are significant challenges to overcome in translating CS-based hydrogels into clinical practice, particularly concerning regulatory approval. Additionally, manufacturing CS-based hydrogels with consistent quality and in sufficient quantities for clinical use poses difficulties, which limits large-scale applications. The U.S. Food and Drug Administration approved CS as a GRAS (Generally Recognized as Safe) biomaterial in 2003 for use in wound hemostatic dressing due to its biocompatibility, biodegradability, and hemostatic properties. However, CS has not yet been approved for pharmaceutical use, primarily due to concerns over its source, purity, and potential immunogenicity [154,155].
In cancer therapy, ClinicalTrials.gov (accessed on 16 September 2024) reports only a few clinical trials involving CS, including studies on breast neoplasm (NCT03202446), breast cancer (NCT02967146), cancer pain (NCT02591017), prostate cancer (NCT03712371), lung cancer (NCT04218188), and advanced solid tumors (NCT03993678). Despite this, many preclinical studies have demonstrated the potential of CS-based smart hydrogels in CRC. Table 5 summarizes the current advantages and limitations of using smart CS-based hydrogels in clinical practice.

6. Future Perspective

CS-based hydrogels present a promising future in DDSs for CRC therapy due to their unique physicochemical and biological properties, which can be modulated through synthesis. Future research could focus on more precise therapies by functionalizing CS-based hydrogels with targeting ligands or antibodies specific to CRC cells, thereby improving efficacy and reducing off-target effects. Moreover, the development of new cross-linkers and methods for synthesizing CS-based hydrogels could enable the production of hydrogels with enhanced mechanical properties and biocompatibility.
The development of multi-stimuli-responsive smart hydrogels capable of releasing drugs in response to various triggers, such as pH changes, temperature, or enzymatic activity associated with CRC, could enable controlled and site-specific drug release, further optimizing treatment outcomes. Moreover, synergistic approaches that combine various chemotherapeutics and immunotherapeutics hold significant promise for enhancing treatment efficacy.
Advances in personalized medicine could lead to the development of customized CS-based hydrogels tailored to individual patient profiles, including specific tumor characteristics and patient-specific drug sensitivities. These hydrogels could also be utilized in 3D bioprinting to develop advanced in vitro models for anticancer drug screening and personalized medicine.
Advanced formulations may focus on reducing peritoneal adhesion formation or promoting tissue regeneration following CRC surgery, thereby improving surgical outcomes.
In summary, the future of CS-based hydrogels in CRC therapy holds significant promise. Continued research and technological advancements are expected to enhance their functionality, effectiveness, and safety, potentially revolutionizing the approach to CRC treatment and improving patient outcomes.

Author Contributions

Conceptualization, U.P.; formal analysis, U.P.; investigation, U.P. and K.O.; writing—original draft preparation, U.P. and K.O.; writing—review and editing, U.P.; visualization, U.P.; supervision, U.P.; project administration, U.P.; funding acquisition, U.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in whole or in part by National Science Centre, Poland [2022/47/D/NZ7/01403]. For the purpose of Open Access, the author has applied a CC-BY public copyright license to any Author Accepted Manuscript (AAM) version arising from this submission.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

5-ASA5-aminosalicylic acid
5-FU5-fluorouracil
AAAcrylic acid
AMPS2-acrylamido-2-methylpropane sulfonic acid
ASAAcetylsalicylic acid
CMCCarboxymethyl cellulose
CMCSCarboxymethyl chitosan
CRCColorectal cancer
CSChitosan
CSDAGG Chitosan-dialdehyde guar gum hydrogels
CURCurcumin
DDDegree of deacetylation
DDP Cisplatin
DDSsDrug delivery systems
DOXDoxorubicin
EVitamin E
FTIR Fourier Transform Infrared Spectroscopy
GGGuar gum
GRAS Generally Recognized as Safe
HAHyaluronic acid
HACPNPoly(N-isopropylacrylamide)-based hydrogel
IMT Imatinib
MTXMethotrexate
NMR Nuclear Magnetic Resonance
NPsNanoparticles
OD Oxidized dextran
ONBOrtho-nitro benzyl
PAAPoly (acrylic acid)
PEG Poly(ethylene glycol)
PNIPAM Poly N-isopropyl acrylamide
PTTPhotothermal therapy
PVAPoly(vinyl alcohol)
SCF Simulated colonic fluid
SGFSimulated gastric fluid
SIFSimulated intestinal fluid
TATannic acid
TJsTight junctions
TzsTetrazines
β-GPβ-glycerophosphate

References

  1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA A Cancer J. Clin. 2024, 74, 12–29. [Google Scholar] [CrossRef] [PubMed]
  2. Akinkuotu, A.C.; Maduekwe, U.N.; Hayes-Jordan, A. Surgical outcomes and survival rates of colon cancer in children and young adults. Am. J. Surg. 2021, 221, 718–724. [Google Scholar] [CrossRef] [PubMed]
  3. Nguyen, H.T.; Duong, H.Q. The molecular characteristics of colorectal cancer: Implications for diagnosis and therapy. Oncol. Lett. 2018, 16, 9–18. [Google Scholar] [CrossRef] [PubMed]
  4. Van Der Flier, L.G.; Clevers, H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu. Rev. Physiol. 2009, 71, 241–260. [Google Scholar] [CrossRef]
  5. Ahmad, R.; Singh, J.K.; Wunnava, A.; Al-Obeed, O.; Abdulla, M.; Srivastava, S.K. Emerging trends in colorectal cancer: Dysregulated signaling pathways. Int. J. Mol. Med. 2021, 47, 14. [Google Scholar] [CrossRef]
  6. Burnett-Hartman, A.N.; Newcomb, P.A.; Potter, J.D. Infectious agents and colorectal cancer: A review of Helicobacter pylori, Streptococcus bovis, JC virus, and human papillomavirus. Cancer Epidemiol. Biomark. Prev. 2008, 17, 2970–2979. [Google Scholar] [CrossRef]
  7. Wong, S.H.; Yu, J. Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 690–704. [Google Scholar] [CrossRef]
  8. Hussain, M.; Waqas, O.; Hassan, U.; Loya, A.; Akhtar, N.; Mushtaq, S.; Yusuf, M.A.; Syed, A.A. Right-sided and left-sided colon cancers are two distinct disease entities: An analysis of 200 cases in Pakistan. Asian Pac. J. Cancer Prev. 2016, 17, 2545–2548. [Google Scholar]
  9. Sears, C.L.; Garrett, W.S. Microbes, microbiota, and colon cancer. Cell Host Microbe 2014, 15, 317–328. [Google Scholar] [CrossRef]
  10. Zhong, M.; Xiong, Y.; Ye, Z.; Zhao, J.; Zhong, L.; Liu, Y.; Zhu, Y.; Tian, L.; Qiu, X.; Hong, X. Microbial community profiling distinguishes left-sided and right-sided colon cancer. Front. Cell. Infect. Microbiol. 2020, 10, 498502. [Google Scholar] [CrossRef]
  11. Roeder, F.; Meldolesi, E.; Gerum, S.; Valentini, V.; Rödel, C. Recent advances in (chemo-) radiation therapy for rectal cancer: A comprehensive review. Radiat. Oncol. 2020, 15, 262. [Google Scholar] [CrossRef] [PubMed]
  12. Chibaudel, B.; Tournigand, C.; Bonnetain, F.; Richa, H.; Benetkiewicz, M.; André, T.; de Gramont, A. Therapeutic strategy in unresectable metastatic colorectal cancer: An updated review. Ther. Adv. Med. Oncol. 2015, 7, 153–169. [Google Scholar] [CrossRef] [PubMed]
  13. Mi, Y.; Zhao, J.; Feng, S.-S. Targeted co-delivery of docetaxel, cisplatin and herceptin by vitamin E TPGS-cisplatin prodrug nanoparticles for multimodality treatment of cancer. J. Control. Release 2013, 169, 185–192. [Google Scholar] [CrossRef]
  14. Dai, L.; Liu, J.; Luo, Z.; Li, M.; Cai, K. Tumor therapy: Targeted drug delivery systems. J. Mater. Chem. B 2016, 4, 6758–6772. [Google Scholar] [CrossRef]
  15. Pacheco, C.; Baiao, A.; Ding, T.; Cui, W.; Sarmento, B. Recent advances in long-acting drug delivery systems for anticancer drug. Adv. Drug Deliv. Rev. 2023, 194, 114724. [Google Scholar] [CrossRef]
  16. Kesharwani, P.; Bisht, A.; Alexander, A.; Dave, V.; Sharma, S. Biomedical applications of hydrogels in drug delivery system: An update. J. Drug Deliv. Sci. Technol. 2021, 66, 102914. [Google Scholar] [CrossRef]
  17. Ho, T.-C.; Chang, C.-C.; Chan, H.-P.; Chung, T.-W.; Shu, C.-W.; Chuang, K.-P.; Duh, T.-H.; Yang, M.-H.; Tyan, Y.-C. Hydrogels: Properties and applications in biomedicine. Molecules 2022, 27, 2902. [Google Scholar] [CrossRef]
  18. Wu, Q.; Hu, Y.; Yu, B.; Hu, H.; Xu, F.-J. Polysaccharide-based tumor microenvironment-responsive drug delivery systems for cancer therapy. J. Control. Release 2023, 362, 19–43. [Google Scholar] [CrossRef]
  19. Kurakula, M.; Gorityala, S.; Moharir, K. Recent trends in design and evaluation of chitosan-based colon targeted drug delivery systems: Update 2020. J. Drug Deliv. Sci. Technol. 2021, 64, 102579. [Google Scholar] [CrossRef]
  20. Sanford, P.; Hutchings, G. Chitosan-A Natural, Cationic Biopolymer: Commercial Application in Industrial Polysaccharides; Elsevier: Amsterdam, The Netherlands, 1987. [Google Scholar]
  21. Croisier, F.; Jérôme, C. Chitosan-based biomaterials for tissue engineering. Eur. Polym. J. 2013, 49, 780–792. [Google Scholar] [CrossRef]
  22. Younes, I.; Rinaudo, M. Chitin and chitosan preparation from marine sources. Structure, properties and applications. Mar. Drugs 2015, 13, 1133–1174. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, J.; Zhuang, S. Chitosan-based materials: Preparation, modification and application. J. Clean. Prod. 2022, 355, 131825. [Google Scholar] [CrossRef]
  24. Aranaz, I.; Alcántara, A.R.; Civera, M.C.; Arias, C.; Elorza, B.; Heras Caballero, A.; Acosta, N. Chitosan: An overview of its properties and applications. Polymers 2021, 13, 3256. [Google Scholar] [CrossRef] [PubMed]
  25. Huq, T.; Khan, A.; Brown, D.; Dhayagude, N.; He, Z.; Ni, Y. Sources, production and commercial applications of fungal chitosan: A review. J. Bioresour. Bioprod. 2022, 7, 85–98. [Google Scholar] [CrossRef]
  26. Abourehab, M.A.; Pramanik, S.; Abdelgawad, M.A.; Abualsoud, B.M.; Kadi, A.; Ansari, M.J.; Deepak, A. Recent advances of chitosan formulations in biomedical applications. Int. J. Mol. Sci. 2022, 23, 10975. [Google Scholar] [CrossRef]
  27. Desai, R.; Pachpore, R.; Patil, A.; Jain, R.; Dandekar, P. Review of the structure of chitosan in the context of other sugar-based polymers. In Chitosan for Biomaterials III: Structure-Property Relationships; Springer: Cham, Switzerland, 2021; pp. 23–74. [Google Scholar]
  28. Feng, C.; Li, J.; Kong, M.; Liu, Y.; Cheng, X.J.; Li, Y.; Park, H.J.; Chen, X.G. Surface charge effect on mucoadhesion of chitosan based nanogels for local anti-colorectal cancer drug delivery. Colloids Surf. B Biointerfaces 2015, 128, 439–447. [Google Scholar] [CrossRef]
  29. Smith, J.; Wood, E.; Dornish, M. Effect of chitosan on epithelial cell tight junctions. Pharm. Res. 2004, 21, 43–49. [Google Scholar] [CrossRef]
  30. Tallian, C.; Tegl, G.; Quadlbauer, L.; Vielnascher, R.; Weinberger, S.; Cremers, R.; Pellis, A.; Salari, J.W.; Guebitz, G.M. Lysozyme-responsive spray-dried chitosan particles for early detection of wound infection. ACS Appl. Bio Mater. 2019, 2, 1331–1339. [Google Scholar] [CrossRef]
  31. Ilium, L. Chitosan and its use as a pharmaceutical excipient. Pharm. Res. 1998, 15, 1326–1331. [Google Scholar] [CrossRef]
  32. Singla, A.; Chawla, M. Chitosan: Some pharmaceutical and biological aspects-an update. J. Pharm. Pharmacol. 2001, 53, 1047–1067. [Google Scholar] [CrossRef]
  33. Kas, H.S. Chitosan: Properties, preparations and application to microparticulate systems. J. Microencapsul. 1997, 14, 689–711. [Google Scholar] [CrossRef] [PubMed]
  34. Shanmuganathan, R.; Edison, T.N.J.I.; LewisOscar, F.; Kumar, P.; Shanmugam, S.; Pugazhendhi, A. Chitosan nanopolymers: An overview of drug delivery against cancer. Int. J. Biol. Macromol. 2019, 130, 727–736. [Google Scholar] [CrossRef] [PubMed]
  35. Sánchez-Machado, D.I.; López-Cervantes, J.; Escárcega-Galaz, A.A.; Campas-Baypoli, O.N.; Martínez-Ibarra, D.M.; Rascón-León, S. Measurement of the degree of deacetylation in chitosan films by FTIR, 1H NMR and UV spectrophotometry. MethodsX 2024, 12, 102583. [Google Scholar] [CrossRef]
  36. Devlieghere, F.; Vermeulen, A.; Debevere, J. Chitosan: Antimicrobial activity, interactions with food components and applicability as a coating on fruit and vegetables. Food Microbiol. 2004, 21, 703–714. [Google Scholar] [CrossRef]
  37. Roller, S.; Covill, N. The antifungal properties of chitosan in laboratory media and apple juice. Int. J. Food Microbiol. 1999, 47, 67–77. [Google Scholar] [CrossRef]
  38. Papineau, A.M.; Hoover, D.G.; Knorr, D.; Farkas, D.F. Antimicrobial effect of water-soluble chitosans with high hydrostatic pressure. Food Biotechnol. 1991, 5, 45–57. [Google Scholar] [CrossRef]
  39. Kumar, M.R.; Muzzarelli, R.A.; Muzzarelli, C.; Sashiwa, H.; Domb, A. Chitosan chemistry and pharmaceutical perspectives. Chem. Rev. 2004, 104, 6017–6084. [Google Scholar] [CrossRef] [PubMed]
  40. Sogias, I.A.; Khutoryanskiy, V.V.; Williams, A.C. Exploring the factors affecting the solubility of chitosan in water. Macromol. Chem. Phys. 2010, 211, 426–433. [Google Scholar] [CrossRef]
  41. Errington, N.; Harding, S.; Vårum, K.; Illum, L. Hydrodynamic characterization of chitosans varying in degree of acetylation. Int. J. Biol. Macromol. 1993, 15, 113–117. [Google Scholar] [CrossRef]
  42. Qin, C.; Li, H.; Xiao, Q.; Liu, Y.; Zhu, J.; Du, Y. Water-solubility of chitosan and its antimicrobial activity. Carbohydr. Polym. 2006, 63, 367–374. [Google Scholar] [CrossRef]
  43. Hejazi, R.; Amiji, M. Chitosan-based gastrointestinal delivery systems. J. Control. Release 2003, 89, 151–165. [Google Scholar] [CrossRef] [PubMed]
  44. Hu, C.-s.; Chiang, C.-h.; Hong, P.-d.; Yeh, M.-k. Influence of charge on FITC-BSA-loaded chondroitin sulfate-chitosan nanoparticles upon cell uptake in human Caco-2 cell monolayers. Int. J. Nanomed. 2012, 7, 4861–4872. [Google Scholar]
  45. Kotze, A.; Luessen, H.; De Boer, A.; Verhoef, J.; Junginger, H. Chitosan for enhanced intestinal permeability: Prospects for derivatives soluble in neutral and basic environments. Eur. J. Pharm. Sci. 1999, 7, 145–151. [Google Scholar] [CrossRef]
  46. Kotzé, A.F.; Thanou, M.M.; Lueßen, H.L.; Verhoef, J.C.; Junginger, H.E. Effect of the degree of quaternization of N-trimethyl chitosan chloride on the permeability of intestinal epithelial cells (Caco-2). Eur. J. Pharm. Biopharm. 1999, 47, 269–274. [Google Scholar] [CrossRef]
  47. Schipper, N.G.; Olsson, S.; Hoogstraate, J.A.; deBoer, A.G.; Vårum, K.M.; Artursson, P. Chitosans as absorption enhancers for poorly absorbable drugs 2: Mechanism of absorption enhancement. Pharm. Res. 1997, 14, 923–929. [Google Scholar] [CrossRef]
  48. Rosenthal, R.; Günzel, D.; Finger, C.; Krug, S.M.; Richter, J.F.; Schulzke, J.-D.; Fromm, M.; Amasheh, S. The effect of chitosan on transcellular and paracellular mechanisms in the intestinal epithelial barrier. Biomaterials 2012, 33, 2791–2800. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, Y.; Ling, Z.; Wang, X.; Zong, S.; Yang, J.; Zhang, Q.; Zhang, J.; Li, X. The beneficial mechanism of chitosan and chitooligosaccharides in the intestine on different health status. J. Funct. Foods 2022, 97, 105232. [Google Scholar] [CrossRef]
  50. Wu, M.; Li, J.; An, Y.; Li, P.; Xiong, W.; Li, J.; Yan, D.; Wang, M.; Zhong, G. Chitooligosaccharides Prevents the Development of Colitis-Associated Colorectal Cancer by Modulating the Intestinal Microbiota and Mycobiota. Front. Microbiol. 2019, 10, 2101. [Google Scholar] [CrossRef]
  51. Calinescu, C.; Mateescu, M.A. Carboxymethyl high amylose starch: Chitosan self-stabilized matrix for probiotic colon delivery. Eur. J. Pharm. Biopharm. 2008, 70, 582–589. [Google Scholar] [CrossRef]
  52. Bhumkar, D.R.; Pokharkar, V.B. Studies on effect of pH on cross-linking of chitosan with sodium tripolyphosphate: A technical note. Aaps Pharmscitech 2006, 7, E138–E143. [Google Scholar] [CrossRef]
  53. Hong, F.; Qiu, P.; Wang, Y.; Ren, P.; Liu, J.; Zhao, J.; Gou, D. Chitosan-based hydrogels: From preparation to applications, a review. Food Chem. X 2023, 21, 101095. [Google Scholar] [CrossRef] [PubMed]
  54. Dragan, E.S.; Dinu, M.V. Advances in porous chitosan-based composite hydrogels: Synthesis and applications. React. Funct. Polym. 2020, 146, 104372. [Google Scholar] [CrossRef]
  55. Berger, J.; Reist, M.; Mayer, J.; Felt, O.; Gurny, R. Structure and interactions in chitosan hydrogels formed by complexation or aggregation for biomedical applications. Eur. J. Pharm. Biopharm. 2004, 57, 35–52. [Google Scholar] [CrossRef]
  56. Jin, S.; Liu, M.; Chen, S.; Gao, C. A drug-loaded gel based on polyelectrolyte complexes of poly (acrylic acid) with poly (vinylpyrrolidone) and chitosan. Mater. Chem. Phys. 2010, 123, 463–470. [Google Scholar] [CrossRef]
  57. Tian, B.; Hua, S.; Tian, Y.; Liu, J. Chemical and physical chitosan hydrogels as prospective carriers for drug delivery: A review. J. Mater. Chem. B 2020, 8, 10050–10064. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Guan, Y.; Zhou, S. Single component chitosan hydrogel microcapsule from a layer-by-layer approach. Biomacromolecules 2005, 6, 2365–2369. [Google Scholar] [CrossRef]
  59. McClements, D.J. Designing biopolymer microgels to encapsulate, protect and deliver bioactive components: Physicochemical aspects. Adv. Colloid Interface Sci. 2017, 240, 31–59. [Google Scholar] [CrossRef]
  60. Delmar, K.; Bianco-Peled, H. The dramatic effect of small pH changes on the properties of chitosan hydrogels crosslinked with genipin. Carbohydr. Polym. 2015, 127, 28–37. [Google Scholar] [CrossRef]
  61. Silva, S.S.; Motta, A.; Rodrigues, M.T.; Pinheiro, A.F.; Gomes, M.E.; Mano, J.F.; Reis, R.L.; Migliaresi, C. Novel genipin-cross-linked chitosan/silk fibroin sponges for cartilage engineering strategies. Biomacromolecules 2008, 9, 2764–2774. [Google Scholar] [CrossRef]
  62. Sharmin, N.; Rosnes, J.T.; Prabhu, L.; Böcker, U.; Sivertsvik, M. Effect of citric acid cross linking on the mechanical, rheological and barrier properties of chitosan. Molecules 2022, 27, 5118. [Google Scholar] [CrossRef]
  63. Zhang, N.; Zhang, H.; Li, R.; Xing, Y. Preparation and adsorption properties of citrate-crosslinked chitosan salt microspheres by microwave assisted method. Int. J. Biol. Macromol. 2020, 152, 1146–1156. [Google Scholar] [CrossRef] [PubMed]
  64. Nguyen, N.-T.; Liu, J.-H. Fabrication and characterization of poly (vinyl alcohol)/chitosan hydrogel thin films via UV irradiation. Eur. Polym. J. 2013, 49, 4201–4211. [Google Scholar] [CrossRef]
  65. Wang, Y.; Zhang, S.; Wang, J. Photo-crosslinkable hydrogel and its biological applications. Chin. Chem. Lett. 2021, 32, 1603–1614. [Google Scholar] [CrossRef]
  66. Hyun, H.; Park, M.H.; Lim, W.; Kim, S.Y.; Jo, D.; Jung, J.S.; Jo, G.; Um, S.; Lee, D.-W.; Yang, D.H. Injectable visible light-cured glycol chitosan hydrogels with controlled release of anticancer drugs for local cancer therapy in vivo: A feasible study. Artif. Cells Nanomed. Biotechnol. 2018, 46, 874–882. [Google Scholar] [CrossRef] [PubMed]
  67. Tatarusanu, S.-M.; Sava, A.; Profire, B.-S.; Pinteala, T.; Jitareanu, A.; Iacob, A.-T.; Lupascu, F.; Simionescu, N.; Rosca, I.; Profire, L. New Smart Bioactive and Biomimetic Chitosan-Based Hydrogels for Wounds Care Management. Pharmaceutics 2023, 15, 975. [Google Scholar] [CrossRef]
  68. Guaresti, O.; García–Astrain, C.; Aguirresarobe, R.; Eceiza, A.; Gabilondo, N. Synthesis of stimuli–responsive chitosan–based hydrogels by Diels–Alder cross–linkingclick reaction as potential carriers for drug administration. Carbohydr. Polym. 2018, 183, 278–286. [Google Scholar] [CrossRef]
  69. Guaresti, O.; Basasoro, S.; González, K.; Eceiza, A.; Gabilondo, N. In situ cross–linked chitosan hydrogels via Michael addition reaction based on water–soluble thiol–maleimide precursors. Eur. Polym. J. 2019, 119, 376–384. [Google Scholar] [CrossRef]
  70. Li, R.; Liu, Q.; Wu, H.; Wang, K.; Li, L.; Zhou, C.; Ao, N. Preparation and characterization of in-situ formable liposome/chitosan composite hydrogels. Mater. Lett. 2018, 220, 289–292. [Google Scholar] [CrossRef]
  71. Tsuda, Y.; Ishihara, M.; Amako, M.; Arino, H.; Hattori, H.; Kanatani, Y.; Yura, H.; Nemoto, K. Photocrosslinkable chitosan hydrogel can prevent bone formation in both rat skull and fibula bone defects. Artif. Organs 2009, 33, 74–77. [Google Scholar] [CrossRef]
  72. Pei, M.; Mao, J.; Xu, W.; Zhou, Y.; Xiao, P. Photocrosslinkable chitosan hydrogels and their biomedical applications. J. Polym. Sci. Part A Polym. Chem. 2019, 57, 1862–1871. [Google Scholar] [CrossRef]
  73. Cho, I.S.; Cho, M.O.; Li, Z.; Nurunnabi, M.; Park, S.Y.; Kang, S.-W.; Huh, K.M. Synthesis and characterization of a new photo-crosslinkable glycol chitosan thermogel for biomedical applications. Carbohydr. Polym. 2016, 144, 59–67. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, Q.; Lv, Y.; Liu, M.; Wang, X.; Mi, Y.; Wang, Q. Nanoinitiator for enzymatic anaerobic polymerization and graft enhancement of gelatin–PAAM hydrogel. J. Mater. Chem. B 2018, 6, 1402–1409. [Google Scholar] [CrossRef] [PubMed]
  75. Ravishankar, K.; Dhamodharan, R. Advances in chitosan-based hydrogels: Evolution from covalently crosslinked systems to ionotropically crosslinked superabsorbents. React. Funct. Polym. 2020, 149, 104517. [Google Scholar] [CrossRef]
  76. Tian, B.; Liu, J. Smart stimuli-responsive chitosan hydrogel for drug delivery: A review. Int. J. Biol. Macromol. 2023, 235, 123902. [Google Scholar] [CrossRef]
  77. He, Q.; Huang, D.; Yang, J.; Huang, Y.; Wang, S. Dual cross-link networks to preserve physical interactions induced by soaking methods: Developing a strong and biocompatible protein-based hydrogel. ACS Appl. Bio Mater. 2019, 2, 3352–3361. [Google Scholar] [CrossRef]
  78. Papagiannopoulos, A.; Nikolakis, S.-P.; Pamvouxoglou, A.; Koutsopoulou, E. Physicochemical properties of electrostatically crosslinked carrageenan/chitosan hydrogels and carrageenan/chitosan/Laponite nanocomposite hydrogels. Int. J. Biol. Macromol. 2023, 225, 565–573. [Google Scholar] [CrossRef] [PubMed]
  79. Nie, J.; Wang, Z.; Hu, Q. Difference between chitosan hydrogels via alkaline and acidic solvent systems. Sci. Rep. 2016, 6, 36053. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, Z.; Nie, J.; Qin, W.; Hu, Q.; Tang, B.Z. Gelation process visualized by aggregation-induced emission fluorogens. Nat. Commun. 2016, 7, 12033. [Google Scholar] [CrossRef]
  81. Reig-Vano, B.; Tylkowski, B.; Montané, X.; Giamberini, M. Alginate-based hydrogels for cancer therapy and research. Int. J. Biol. Macromol. 2021, 170, 424–436. [Google Scholar] [CrossRef]
  82. Fernández-Quiroz, D.; Loya-Duarte, J.; Silva-Campa, E.; Argüelles-Monal, W.; Sarabia-Sainz, A.í.; Lucero-Acuña, A.; del Castillo-Castro, T.; San Román, J.; Lizardi-Mendoza, J.; Burgara-Estrella, A.J. Temperature stimuli-responsive nanoparticles from chitosan-graft-poly (N-vinylcaprolactam) as a drug delivery system. J. Appl. Polym. Sci. 2019, 136, 47831. [Google Scholar] [CrossRef]
  83. Calvo, P.; Remunan-Lopez, C.; Vila-Jato, J.L.; Alonso, M. Novel hydrophilic chitosan-polyethylene oxide nanoparticles as protein carriers. J. Appl. Polym. Sci. 1997, 63, 125–132. [Google Scholar] [CrossRef]
  84. Bugnicourt, L.; Ladavière, C. Interests of chitosan nanoparticles ionically cross-linked with tripolyphosphate for biomedical applications. Prog. Polym. Sci. 2016, 60, 1–17. [Google Scholar] [CrossRef]
  85. Angelova, N.; Hunkeler, D. Permeability and stability of chitosan-based capsules: Effect of preparation. Int. J. Pharm. 2002, 242, 229–232. [Google Scholar] [CrossRef]
  86. Peniche, H.; Peniche, C. Chitosan nanoparticles: A contribution to nanomedicine. Polym. Int. 2011, 60, 883–889. [Google Scholar] [CrossRef]
  87. Wang, Q.-S.; Wang, G.-F.; Zhou, J.; Gao, L.-N.; Cui, Y.-L. Colon targeted oral drug delivery system based on alginate-chitosan microspheres loaded with icariin in the treatment of ulcerative colitis. Int. J. Pharm. 2016, 515, 176–185. [Google Scholar] [CrossRef]
  88. Balima, M.; Morfin, I.; Sudre, G.; Montembault, A. Stretchable hydrogels of chitosan/hyaluronic acid induced by polyelectrolyte complexation around neutral pH. Carbohydr. Polym. 2024, 339, 122265. [Google Scholar] [CrossRef] [PubMed]
  89. Oyarzun-Ampuero, F.; Brea, J.; Loza, M.; Torres, D.; Alonso, M. Chitosan–hyaluronic acid nanoparticles loaded with heparin for the treatment of asthma. Int. J. Pharm. 2009, 381, 122–129. [Google Scholar] [CrossRef]
  90. Denuziere, A.; Ferrier, D.; Domard, A. Chitosan-chondroitin sulfate and chitosan-hyaluronate polyelectrolyte complexes. Physico-chemical aspects. Carbohydr. Polym. 1996, 29, 317–323. [Google Scholar] [CrossRef]
  91. Pourjavadi, A.; Doroudian, M.; Ahadpour, A.; Azari, S. Injectable chitosan/κ-carrageenan hydrogel designed with au nanoparticles: A conductive scaffold for tissue engineering demands. Int. J. Biol. Macromol. 2019, 126, 310–317. [Google Scholar] [CrossRef]
  92. Sinha, V.; Singla, A.K.; Wadhawan, S.; Kaushik, R.; Kumria, R.; Bansal, K.; Dhawan, S. Chitosan microspheres as a potential carrier for drugs. Int. J. Pharm. 2004, 274, 1–33. [Google Scholar] [CrossRef]
  93. Bezerra, M.C.; Duarte, G.A.; Talabi, S.I.; Lucas, A.A. Microstructure and properties of thermomechanically processed chitosan citrate-based materials. Carbohydr. Polym. 2022, 278, 118984. [Google Scholar] [CrossRef] [PubMed]
  94. Zhou, H.Y.; Jiang, L.J.; Cao, P.P.; Li, J.B.; Chen, X.G. Glycerophosphate-based chitosan thermosensitive hydrogels and their biomedical applications. Carbohydr. Polym. 2015, 117, 524–536. [Google Scholar] [CrossRef] [PubMed]
  95. Yuan, Z.; Ding, J.; Zhang, Y.; Huang, B.; Song, Z.; Meng, X.; Ma, X.; Gong, X.; Huang, Z.; Ma, S. Components, mechanisms and applications of stimuli-responsive polymer gels. Eur. Polym. J. 2022, 177, 111473. [Google Scholar] [CrossRef]
  96. Wei, M.; Gao, Y.; Li, X.; Serpe, M.J. Stimuli-responsive polymers and their applications. Polym. Chem. 2017, 8, 127–143. [Google Scholar] [CrossRef]
  97. Kumar, N.; Singh, S.; Sharma, P.; Kumar, B.; Kumar, A. Single-, Dual-, and Multi-Stimuli-Responsive Nanogels for Biomedical Applications. Gels 2024, 10, 61. [Google Scholar] [CrossRef]
  98. Elias, D.; Blot, F.; El Otmany, A.; Antoun, S.; Lasser, P.; Boige, V.; Rougier, P.; Ducreux, M. Curative treatment of peritoneal carcinomatosis arising from colorectal cancer by complete resection and intraperitoneal chemotherapy. Cancer 2001, 92, 71–76. [Google Scholar] [CrossRef]
  99. Verwaal, V.J.; Bruin, S.; Boot, H.; Van Slooten, G.; Van Tinteren, H. 8-year follow-up of randomized trial: Cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann. Surg. Oncol. 2008, 15, 2426–2432. [Google Scholar] [CrossRef]
  100. Levine, E.A.; Stewart IV, J.H.; Shen, P.; Russell, G.B.; Loggie, B.L.; Votanopoulos, K.I. Intraperitoneal chemotherapy for peritoneal surface malignancy: Experience with 1,000 patients. J. Am. Coll. Surg. 2014, 218, 573–585. [Google Scholar] [CrossRef]
  101. Garg, A.; Agrawal, R.; Chauhan, C.S.; Deshmukh, R. In-situ gel: A smart carrier for drug delivery. Int. J. Pharm. 2024, 652, 123819. [Google Scholar] [CrossRef]
  102. Ruel-Gariepy, E.; Leroux, J.-C. In situ-forming hydrogels—Review of temperature-sensitive systems. Eur. J. Pharm. Biopharm. 2004, 58, 409–426. [Google Scholar] [CrossRef]
  103. Gao, Y.; Deng, A.; Wu, X.; Sun, C.; Qi, C. Injectable multi-responsive hydrogels cross-linked by responsive macromolecular micelles. React. Funct. Polym. 2021, 161, 104866. [Google Scholar] [CrossRef]
  104. Qiu, Y.; Park, K. Environment-sensitive hydrogels for drug delivery. Adv. Drug Deliv. Rev. 2001, 53, 321–339. [Google Scholar] [CrossRef] [PubMed]
  105. Ruel-Gariepy, E.; Chenite, A.; Chaput, C.; Guirguis, S.; Leroux, J.-C. Characterization of thermosensitive chitosan gels for the sustained delivery of drugs. Int. J. Pharm. 2000, 203, 89–98. [Google Scholar] [CrossRef] [PubMed]
  106. Kehoe, S.M.; Williams, N.L.; Yakubu, R.; Levine, D.A.; Chi, D.S.; Sabbatini, P.J.; Aghajanian, C.A.; Barakat, R.R.; Abu-Rustum, N.R. Incidence of intestinal obstruction following intraperitoneal chemotherapy for ovarian tubal and peritoneal malignancies. Gynecol. Oncol. 2009, 113, 228–232. [Google Scholar] [CrossRef]
  107. Zan, J.; Chen, H.; Jiang, G.; Lin, Y.; Ding, F. Preparation and properties of crosslinked chitosan thermosensitive hydrogel for injectable drug delivery systems. J. Appl. Polym. Sci. 2006, 101, 1892–1898. [Google Scholar] [CrossRef]
  108. Chenite, A.; Buschmann, M.; Wang, D.; Chaput, C.; Kandani, N. Rheological characterisation of thermogelling chitosan/glycerol-phosphate solutions. Carbohydr. Polym. 2001, 46, 39–47. [Google Scholar] [CrossRef]
  109. Assaad, E.; Maire, M.; Lerouge, S. Injectable thermosensitive chitosan hydrogels with controlled gelation kinetics and enhanced mechanical resistance. Carbohydr. Polym. 2015, 130, 87–96. [Google Scholar] [CrossRef]
  110. Goldberg, R.N.; Kishore, N.; Lennen, R.M. Thermodynamic quantities for the ionization reactions of buffers. J. Phys. Chem. Ref. Data 2002, 31, 231–370. [Google Scholar] [CrossRef]
  111. Chenite, A.; Chaput, C.; Wang, D.; Combes, C.; Buschmann, M.D.; Hoemann, C.; Leroux, J.; Atkinson, B.; Binette, F.; Selmani, A. Novel injectable neutral solutions of chitosan form biodegradable gels in situ. Biomaterials 2000, 21, 2155–2161. [Google Scholar] [CrossRef] [PubMed]
  112. Lavertu, M.; Filion, D.; Buschmann, M.D. Heat-induced transfer of protons from chitosan to glycerol phosphate produces chitosan precipitation and gelation. Biomacromolecules 2008, 9, 640–650. [Google Scholar] [CrossRef]
  113. Cho, J.; Heuzey, M.-C.; Bégin, A.; Carreau, P.J. Physical gelation of chitosan in the presence of β-glycerophosphate: The effect of temperature. Biomacromolecules 2005, 6, 3267–3275. [Google Scholar] [CrossRef] [PubMed]
  114. Rezakhani, L.; Alizadeh, M.; Alizadeh, A. A three dimensional in vivo model of breast cancer using a thermosensitive chitosan-based hydrogel and 4 T1 cell line in Balb/c. J. Biomed. Mater. Res. Part A 2021, 109, 1275–1285. [Google Scholar] [CrossRef] [PubMed]
  115. Taherian, A.R.; Lacasse, P.; Bisakowski, B.; Pelletier, M.; Lanctôt, S.; Fustier, P. Rheological and thermogelling properties of commercials chitosan/β-glycerophosphate: Retention of hydrogel in water, milk and UF-milk. Food Hydrocoll. 2017, 63, 635–645. [Google Scholar] [CrossRef]
  116. Deng, A.; Kang, X.; Zhang, J.; Yang, Y.; Yang, S. Enhanced gelation of chitosan/β-sodium glycerophosphate thermosensitive hydrogel with sodium bicarbonate and biocompatibility evaluated. Mater. Sci. Eng. C 2017, 78, 1147–1154. [Google Scholar] [CrossRef]
  117. Rahimnejad, M.; Adoungotchodo, A.; Demarquette, N.R.; Lerouge, S. FRESH bioprinting of biodegradable chitosan thermosensitive hydrogels. Bioprinting 2022, 27, e00209. [Google Scholar] [CrossRef]
  118. Yun, Q.; Wang, S.S.; Xu, S.; Yang, J.P.; Fan, J.; Yang, L.L.; Chen, Y.; Fu, S.Z.; Wu, J.B. Use of 5-Fluorouracil Loaded Micelles and Cisplatin in Thermosensitive Chitosan Hydrogel as an Efficient Therapy against Colorectal Peritoneal Carcinomatosis. Macromol. Biosci. 2017, 17, 1600262. [Google Scholar] [CrossRef]
  119. Chen, C.-H.; Kuo, C.-Y.; Chen, S.-H.; Mao, S.-H.; Chang, C.-Y.; Shalumon, K.; Chen, J.-P. Thermosensitive injectable hydrogel for simultaneous intraperitoneal delivery of doxorubicin and prevention of peritoneal adhesion. Int. J. Mol. Sci. 2018, 19, 1373. [Google Scholar] [CrossRef]
  120. Zheng, Y.; Wang, W.; Zhao, J.; Wu, C.; Ye, C.; Huang, M.; Wang, S. Preparation of injectable temperature-sensitive chitosan-based hydrogel for combined hyperthermia and chemotherapy of colon cancer. Carbohydr. Polym. 2019, 222, 115039. [Google Scholar] [CrossRef]
  121. Songkroh, T.; Xie, H.; Yu, W.; Liu, X.; Sun, G.; Xu, X.; Ma, X. Injectable in situ forming chitosan-based hydrogels for curcumin delivery. Macromol. Res. 2015, 23, 53–59. [Google Scholar] [CrossRef]
  122. 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]
  123. Liang, Z.; Gao, J.; Yin, Z.-Z.; Li, J.; Cai, W.; Kong, Y. A sequential delivery system based on MoS2 nanoflower doped chitosan/oxidized dextran hydrogels for colon cancer treatment. Int. J. Biol. Macromol. 2023, 233, 123616. [Google Scholar] [CrossRef] [PubMed]
  124. Khan, S.; Anwar, N. Highly porous pH-responsive carboxymethyl chitosan-grafted-poly (acrylic acid) based smart hydrogels for 5-fluorouracil controlled delivery and colon targeting. Int. J. Polym. Sci. 2019, 2019, 6579239. [Google Scholar] [CrossRef]
  125. Ghobashy, M.M.; Elbarbary, A.M.; Hegazy, D.E. Gamma radiation synthesis of a novel amphiphilic terpolymer hydrogel pH-responsive based chitosan for colon cancer drug delivery. Carbohydr. Polym. 2021, 263, 117975. [Google Scholar] [CrossRef]
  126. Zarbab, A.; Sajjad, A.; Rasul, A.; Jabeen, F.; Iqbal, M.J. Synthesis and characterization of Guar gum based biopolymeric hydrogels as carrier materials for controlled delivery of methotrexate to treat colon cancer. Saudi J. Biol. Sci. 2023, 30, 103731. [Google Scholar] [CrossRef]
  127. Wang, C.-Y.; Sun, M.; Fan, Z.; Du, J.-Z. Intestine enzyme-responsive polysaccharide-based hydrogel to open epithelial tight junctions for oral delivery of imatinib against colon cancer. Chin. J. Polym. Sci. 2022, 40, 1154–1164. [Google Scholar] [CrossRef]
  128. Hoang, H.T.; Jo, S.-H.; Phan, Q.-T.; Park, H.; Park, S.-H.; Oh, C.-W.; Lim, K.T. Dual pH-/thermo-responsive chitosan-based hydrogels prepared using” click” chemistry for colon-targeted drug delivery applications. Carbohydr. Polym. 2021, 260, 117812. [Google Scholar] [CrossRef]
  129. Nisar, S.; Pandit, A.H.; Wang, L.-F.; Rattan, S. Strategy to design a smart photocleavable and pH sensitive chitosan based hydrogel through a novel crosslinker: A potential vehicle for controlled drug delivery. RSC Adv. 2020, 10, 14694–14704. [Google Scholar] [CrossRef]
  130. Chen, J.-P.; Cheng, T.-H. Preparation and evaluation of thermo-reversible copolymer hydrogels containing chitosan and hyaluronic acid as injectable cell carriers. Polymer 2009, 50, 107–116. [Google Scholar] [CrossRef]
  131. Hua, S. Advances in oral drug delivery for regional targeting in the gastrointestinal tract-influence of physiological, pathophysiological and pharmaceutical factors. Front. Pharmacol. 2020, 11, 524. [Google Scholar] [CrossRef]
  132. Ding, H.; Tan, P.; Fu, S.; Tian, X.; Zhang, H.; Ma, X.; Gu, Z.; Luo, K. Preparation and application of pH-responsive drug delivery systems. J. Control. Release 2022, 348, 206–238. [Google Scholar] [CrossRef]
  133. Tan, J.P.; Goh, C.H.; Tam, K.C. Comparative drug release studies of two cationic drugs from pH-responsive nanogels. Eur. J. Pharm. Sci. 2007, 32, 340–348. [Google Scholar] [CrossRef] [PubMed]
  134. Herrmann, R.; Kunz, W.; Osswald, H.; Ritter, M.; Port, R. The effect of methotrexate pretreatment on 5-fluorouracil kinetics in sarcoma 180 in vivo. Eur. J. Cancer Clin. Oncol. 1985, 21, 753–758. [Google Scholar] [CrossRef] [PubMed]
  135. Duan, S.; Hu, Y.; Zhao, Y.; Tang, K.; Zhang, Z.; Liu, Z.; Wang, Y.; Guo, H.; Miao, Y.; Du, H. Nanomaterials for photothermal cancer therapy. RSC Adv. 2023, 13, 14443–14460. [Google Scholar] [CrossRef] [PubMed]
  136. Yu, S.; Xia, G.; Yang, N.; Yuan, L.; Li, J.; Wang, Q.; Li, D.; Ding, L.; Fan, Z.; Li, J. Noble Metal Nanoparticle-Based Photothermal Therapy: Development and Application in Effective Cancer Therapy. Int. J. Mol. Sci. 2024, 25, 5632. [Google Scholar] [CrossRef]
  137. Gaharwar, A.K.; Peppas, N.A.; Khademhosseini, A. Nanocomposite hydrogels for biomedical applications. Biotechnol. Bioeng. 2014, 111, 441–453. [Google Scholar] [CrossRef]
  138. Cheng, X.; Xie, Q.; Sun, Y. Advances in nanomaterial-based targeted drug delivery systems. Front. Bioeng. Biotechnol. 2023, 11, 1177151. [Google Scholar] [CrossRef]
  139. Zhu, A.; Yuan, L.; Liao, T. Suspension of Fe3O4 nanoparticles stabilized by chitosan and o-carboxymethylchitosan. Int. J. Pharm. 2008, 350, 361–368. [Google Scholar] [CrossRef]
  140. Zhu, N.; Ji, H.; Yu, P.; Niu, J.; Farooq, M.; Akram, M.W.; Udego, I.; Li, H.; Niu, X. Surface modification of magnetic iron oxide nanoparticles. Nanomaterials 2018, 8, 810. [Google Scholar] [CrossRef]
  141. Thanou, M.; Verhoef, J.; Junginger, H. Oral drug absorption enhancement by chitosan and its derivatives. Adv. Drug Deliv. Rev. 2001, 52, 117–126. [Google Scholar] [CrossRef]
  142. Fang, N.; Chan, V.; Mao, H.-Q.; Leong, K.W. Interactions of phospholipid bilayer with chitosan: Effect of molecular weight and pH. Biomacromolecules 2001, 2, 1161–1168. [Google Scholar] [CrossRef]
  143. Bowman, K.; Leong, K.W. Chitosan nanoparticles for oral drug and gene delivery. Int. J. Nanomed. 2006, 1, 117–128. [Google Scholar] [CrossRef] [PubMed]
  144. Vllasaliu, D.; Exposito-Harris, R.; Heras, A.; Casettari, L.; Garnett, M.; Illum, L.; Stolnik, S. Tight junction modulation by chitosan nanoparticles: Comparison with chitosan solution. Int. J. Pharm. 2010, 400, 183–193. [Google Scholar] [CrossRef] [PubMed]
  145. Kudsiova, L.; Lawrence, M. A comparison of the effect of chitosan and chitosan-coated vesicles on monolayer integrity and permeability across Caco-2 and 16HBE14o-cells. J. Pharm. Sci. 2008, 97, 3998–4010. [Google Scholar] [CrossRef] [PubMed]
  146. Sun, X.; Liu, C.; Omer, A.; Lu, W.; Zhang, S.; Jiang, X.; Wu, H.; Yu, D.; Ouyang, X.-k. pH-sensitive ZnO/carboxymethyl cellulose/chitosan bio-nanocomposite beads for colon-specific release of 5-fluorouracil. Int. J. Biol. Macromol. 2019, 128, 468–479. [Google Scholar] [CrossRef] [PubMed]
  147. Dalei, G.; Jena, D.; Das, B.R.; Das, S. Bio-valorization of Tagetes floral waste extract in fabrication of self-healing Schiff-base nanocomposite hydrogels for colon cancer remedy. Environ. Sci. Pollut. Res. 2024, 31, 4330–4347. [Google Scholar] [CrossRef]
  148. Amini-Fazl, M.S.; Mohammadi, R.; Kheiri, K. 5-Fluorouracil loaded chitosan/polyacrylic acid/Fe3O4 magnetic nanocomposite hydrogel as a potential anticancer drug delivery system. Int. J. Biol. Macromol. 2019, 132, 506–513. [Google Scholar] [CrossRef]
  149. El-Maadawy, M.W.; Mohamed, R.R.; Hanna, D.H. Preparation of carrageenan/chitosan-based (N, N, N-trimeth (yl chitosan chloride) silver nanocomposites as pH sensitive carrier for effective controlled curcumin delivery in cancer cells. OpenNano 2022, 7, 100050. [Google Scholar] [CrossRef]
  150. Dhanavel, S.; Revathy, T.; Sivaranjani, T.; Sivakumar, K.; Palani, P.; Narayanan, V.; Stephen, A. 5-Fluorouracil and curcumin co-encapsulated chitosan/reduced graphene oxide nanocomposites against human colon cancer cell lines. Polym. Bull. 2020, 77, 213–233. [Google Scholar] [CrossRef]
  151. Sabra, R.; Billa, N.; Roberts, C.J. An augmented delivery of the anticancer agent, curcumin, to the colon. React. Funct. Polym. 2018, 123, 54–60. [Google Scholar] [CrossRef]
  152. Nag, S.; Das Saha, K. Chitosan-decorated PLGA-NPs loaded with tannic acid/vitamin E mitigate colon cancer via the NF-κB/β-Cat/EMT pathway. ACS Omega 2021, 6, 28752–28769. [Google Scholar] [CrossRef]
  153. Alsadooni, J.F.K.; Haghi, M.; Barzegar, A.; Feizi, M.A.H. The effect of chitosan hydrogel containing gold nanoparticle complex with paclitaxel on colon cancer cell line. Int. J. Biol. Macromol. 2023, 247, 125612. [Google Scholar] [CrossRef] [PubMed]
  154. Kean, T.; Thanou, M. Biodegradation, biodistribution and toxicity of chitosan. Adv. Drug Deliv. Rev. 2010, 62, 3–11. [Google Scholar] [CrossRef] [PubMed]
  155. Kantak, M.N.; Bharate, S.S. Analysis of clinical trials on biomaterial and therapeutic applications of chitosan: A review. Carbohydr. Polym. 2022, 278, 118999. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of chitin and chitosan. This figure is adapted from [22] with permission under CC BY 4.0 license.
Figure 1. Chemical structure of chitin and chitosan. This figure is adapted from [22] with permission under CC BY 4.0 license.
Pharmaceuticals 17 01260 g001
Figure 2. The gelation process of CS hydrogel (a) via acidic solvent; (b) via alkaline solvent. This figure is adapted from [79] with permission under CC BY 4.0 license.
Figure 2. The gelation process of CS hydrogel (a) via acidic solvent; (b) via alkaline solvent. This figure is adapted from [79] with permission under CC BY 4.0 license.
Pharmaceuticals 17 01260 g002
Figure 3. Preparation of chitosan nanoparticles by ionic gelation method.
Figure 3. Preparation of chitosan nanoparticles by ionic gelation method.
Pharmaceuticals 17 01260 g003
Figure 4. Unique properties of smart hydrogels.
Figure 4. Unique properties of smart hydrogels.
Pharmaceuticals 17 01260 g004
Figure 5. In situ gelation mechanism between chitosan and β-glycerophosphate.
Figure 5. In situ gelation mechanism between chitosan and β-glycerophosphate.
Pharmaceuticals 17 01260 g005
Figure 6. Phase transition behavior of HACPN-DOX and HACPN solutions at 25 and 37 °C. This figure is adapted from [119] with permission under CC BY 4.0 license.
Figure 6. Phase transition behavior of HACPN-DOX and HACPN solutions at 25 and 37 °C. This figure is adapted from [119] with permission under CC BY 4.0 license.
Pharmaceuticals 17 01260 g006
Table 3. Chitosan-based stimuli-responsive hydrogels for colorectal cancer treatment.
Table 3. Chitosan-based stimuli-responsive hydrogels for colorectal cancer treatment.
Hydrogel SystemTherapeutic AgentRoute of AdministrationIn Vitro StudiesIn Vivo StudiesRef.
Drug Release
Study
CytotoxicityCell LineAnimal ModelPDCell Line
CS/GP5-FU, DDPintraperitoneal injectionSustained manner over an extended period, cumulative release rate of DDP is higher than that of 5-FU.IC50 = 3.43 μg mL−1 (5-FU micelles after 48 h);
IC50 = 6.48 μg mL−1 (5-FU after 48 h).
CT26CRPC mouse model
BALB/c female mice
CS hydrogel drug suppressed the growth of implanted tumor (10.33 ± 2.66, 0.49 ± 0.11 g) compared with NS group (53.83 ± 9.99, 2.31 ± 0.38 g, p < 0.001) and impaired tumor metastasis, as well as prolonged survival time of the tumor-bearing mice.CT26[118]
HACPN
(hyaluronic acid-g-chitosan-g-PNIPAM)
DOXintraperitoneal injectionAt pH 7.4, 40% of the drug was released within 8 h (burst release),sustained release of DOX was observed thereafter with 80% of the drug released in 12 days.Cell viability in the HACPN-DOX group was significantly reduced, reaching 48% and 2% of the control group after 24 and 48 h, respectively.CT26BALB/c miceSuppressed tumor growth and inhibited tumor angiogenesis.CT26[119]
CS/MoS2/Bi2S3-PEGDOXinjection4.5% (at 37 °C and pH = 7.4), 20.3% (at 37 °C and pH = 5.4) and 16.5% (at 50 °C and pH = 5.4)L929 cells viability treated with CS/MoS2/Bi2S3-PEG/DOX: 47.7% (higher compared to DOX alone 36.53%). HT29 cells viability treated with CS/MoS2/Bi2S3-PEG/DOX decreased to 57.12% ± 3.87%
When irradiated with 1064 nm laser, CS/MoS2/Bi2S3-PEG/DOX, CS/MoS2/Bi2S3-PEG +NIR and
CS/MoS2/Bi2S3-PEG/DOX+NIR cultured HT29 tumor cell viability declined to 55.13% ± 3.77%, 36.03 ± 3.29%, and 12.02% ± 0.41%, respectively
confirming the combined tumor therapy efficiency.
HT29
L929
HT29 xenografted
tumor bearing mice
The volume of the
tumor was significantly reduced or even disappeared under the function of combined photothermal and chemotherapy after 14 days’ feeding.
HT29[120]
CS/genipin/sodium saltsCURinjectionThe initial burst releases at the first 24 h were observed in all gel samples and followed by the sustained
release of c.a. 1.0% to 1.8% over day 2 to 7.
Cell viability in 3T3 mouse fibroblast cell lines was above 80%, indicating high cell survival and minimal cytotoxicity under the experimental conditions.3T3Sprague-Dawley rat--[121]
CSDAGGCUR, ASAoralA minute amount of CUR and ASA was released during the initial 2 h in the SGF (pH 1.2). In the SIF (pH 7.4), the release of ASA and CUR was 50% and 25%, respectively. In colonic fluid (pH 6.5), the cumulative release of ASA and CUR was approximately 90% and 42%, respectively, at 24 h.At an equivalent drug concentration, the dual drug-loaded hydrogel exhibited higher cellular cytotoxicity compared to the other samples (pristine CUR, CUR-loaded hydrogel, and ASA-loaded hydrogel).HT29-- [122]
CS/OD/MTX/TFPM5-FU, MTXoralCumulative release of MTX was determined to be 10.99%, 27.53%, 21.47% and 86.51% at pH 1.2, 5.0, 6.8 and 7.4, respectively. The cumulative release of 5-FU at pH 7.4 (maximum of release of MTX) remarkably increased to 89.78% under NIR irradiation.Cell viability < 10% after treatment with 512 μg/mL CS/OD/MTX/TFPM.
The cell viability decreased to 4.9% under NIR irradiation for 30 min.
HT-29---[123]
CMCS/AA5-FUoralat pH 1.2: 21.37–27.76%; at pH 6.5: 61.79–77.69%; at pH 7.5, 77.08–88.89% of the drug was released within 12 h5-FU had dose-dependent cytotoxic potential and the % cell viability decreased with increasing dose per well; 5-FU retained its cytotoxic potential after loading into the hydrogel matrix;
no detectable cytotoxicity on Vero cells.
HeLa,
Vero cells
---[124]
CS/AA/AMPS5-FUoralrelease of 5-FU after 30 min at pH 1 and 7 was 1.55% and 25.3%, respectively;
96% after 7 h at pH 7.
-----[125]
GG/PVA/CSMTXinjection50% drug release was observed in the first 5 h and a sustained drug release of 96% in 7.25 h.IC50 = 11.7 µg/mL at GG/PVA/CS +MTX concentration of 2.34 µg/200 mLHCT-116---[126]
MA-CMCSIMToralAfter 48 h in PBS (pH 7.4), the accumulated percentage of drug release for hydrogel was 55.8%.CS-based hydrogel was non-cytotoxic and had a good biocompatibility against normal and cancer cells; IMT-loaded hydrogel displayed a dose-dependent cytotoxicity, and cell viabilities declined with the increase of drug concentration.LS174T, L02Balb/c
female mice
Significantly enhanced in vivo tumor inhibition (six-fold higher compared to IMT) was achieved after oral administration with IMT-loaded hydrogel.LS174T[127]
PAA/CSNb/bisTz-PNIPAM5-ASAoralThe cumulative drug release was 8.5% at pH 2.2 and reached 92% at pH 7.4 within 48 h. Additionally, the cumulative drug release from the hydrogels at 25 °C was lower compared to that at 37 °C.cell viability exceeded 70%HFF-1---[128]
ONB–CSDOXoralHydrogel exhibited higher drug release at pH 5.7 (71.75%) than at pH 7.4 (30.82%) after 24 h.-----[129]
Table 4. Chitosan-based nanocomposite drug delivery systems for colorectal cancer treatment.
Table 4. Chitosan-based nanocomposite drug delivery systems for colorectal cancer treatment.
Nanocomposite DDSsTherapeutic AgentRoute of AdministrationIn Vitro StudiesRef.
Drug Release
Study
Cytotoxicity
(If Available)
Cell Line
core–shell ZnO/CMC/CS5-FU The drug accumulated release rate was <20 from ZnO/CMC/CS beads within 2 h at SGF (pH 1.2). The cumulative release reached 80% after the next 3 h at SIF (pH 6.8); at SCF (pH 7.4) for a further 3 h the release rate was still rising due to the more hydrophilic system, leading the whole state to collapse drastically.--[146]
CsDAP@ZnO5-FUoralNegligible amount of 5-FU was released during the initial 2 h in SGF (pH 1.2). The release was considerably expedited from 2 to 7 h in the SIF (pH 7.4) from both the hydrogels and gradually increased in SCF (pH 6.5).CsDAP@ZnO nanocomposite hydrogel demonstrated greater toxicity on the colon cancer cells with respect to Sap hydrogel at an equivalent concentration.HT-29[147]
CS/PAA/Fe3O45-FUcolon and rectalAt pH 7.4 in 37 °C the release rate of 5-FU from hydrogel was decreased with the increase of cross-linker and Fe3O4 NPs. Release kinetics from nanohydrogel conformed to the Weibull model.--[148]
CAR/TMC-AgCURoralSustained drug release reached 98.9% ± 0.9 within 24 h in pH 7.4.High cytotoxic effect with apoptotic induction against Caco-2 cells through G2/M cell cycle arrestCaco-2[149]
CS/rGO5-FU
CUR
-pH 5.0
In 72 h, 90% of the release was attained in 5-FU-loaded systems showing higher release over CUR-loaded composites.
IC50 = 23.8 μg/mL for dual-drug-loaded nanocomposite;
IC50 = 37.61 μg/mL for 5-FU loaded nanocomposite, IC50 = 48.12 μg/mL for CUR-loaded nanocomposite.
The cell viability at 40 µg/mL for the NIH 3T3 mouse embryonic fibroblast cells was found to be 80.3%.
HT-29
NIH 3T3
[150]
MCPCCURoralAt pH 1.2 18% CU was released during 2h, and up to 68% release in caecal medium over 24 h.- [151]
CS-PLGA NPsTA/Eintraperitoneally-CS-PLGA NPs
significantly inhibited tumor number and tumor volume and normalized colon histology in the colon cancer.
-[152]
CS hydrogel-coated Au NPsPTX -CS hydrogel-coated Au NPs were able to increase the expression of pro-apoptotic BAX and BAD and decrease the expression of anti-apoptotic BCL2 more than PTX alone.LS174T[153]
Table 5. Advantages and limitations of using smart chitosan-based hydrogels.
Table 5. Advantages and limitations of using smart chitosan-based hydrogels.
CS-Based Hydrogel SystemAdvantagesDisadvantages
In situ gelsLocalized therapy
Controlled drug delivery
Postoperative adjuvant chemotherapy
Endoscopic mucosal resection technique for accurate removal of polyps and early-stage tumors
Intraperitoneal chemotherapy
Unique pharmacokinetics parameters
High payload efficiency
Eliminating the need for surgical removal
Reduced peritoneal adhesion formation
Antibacterial activity (efflux pump inhibition)
High intraperitoneal-to-plasma drug concentration ratio
Low mechanical strength
Slow gelation time
Burst release
Inability of CS to remain in solution at physiological pH
Risk of obstructing the endoscopic needle during injection
Large-scale production challenges
Potential immunogenicity
Stimuli-responsive multi-drug hydrogelsPorous structures increase drug loading
Responses to colon selectivity (pH-, enzymatic-, temperature-, redox, pressure, and mechanical stimuli)
Bio-adhesiveness
Enhanced drug release
Antibacterial activity
Targeting photodynamic and PTT therapy
Burst release in stomach when applied oral
Low mechanical strength
Large-scale production challenges
Tendency to coagulate with protein at high pH
Complex drug release control
Potential immunogenicity
Biodegradation rate challenges
May cause localized tissue damage
Nanocomposite hydrogelsMechanical strength
Thermal stability
Enhance drug delivery
Minimize side effects
Extend drug lifetime in the bloodstream
Protection against acidic and enzymatic degradation in the gastrointestinal tract
Reduce burst release
Permeation enhancement
Controlled drug release
Improve drug-loading efficiency
Stabilize NPs
CS-based NPs and CS-coated microspheres facilitate drug transport partly through endocytosis and transcytosis
Antibacterial activity
Complex manufacturing process
Large-scale production challenges
Batch-to-batch variability
Rapid biodegradation
Potential immunogenicity
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

Piotrowska, U.; Orzechowska, K. Advances in Chitosan-Based Smart Hydrogels for Colorectal Cancer Treatment. Pharmaceuticals 2024, 17, 1260. https://doi.org/10.3390/ph17101260

AMA Style

Piotrowska U, Orzechowska K. Advances in Chitosan-Based Smart Hydrogels for Colorectal Cancer Treatment. Pharmaceuticals. 2024; 17(10):1260. https://doi.org/10.3390/ph17101260

Chicago/Turabian Style

Piotrowska, Urszula, and Klaudia Orzechowska. 2024. "Advances in Chitosan-Based Smart Hydrogels for Colorectal Cancer Treatment" Pharmaceuticals 17, no. 10: 1260. https://doi.org/10.3390/ph17101260

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