Next Article in Journal
Cold Exposure Exacerbates Cardiac Dysfunction in a Model of Heart Failure with Preserved Ejection Fraction in Male and Female C57Bl/6J Mice
Previous Article in Journal
Combined Genetic and Transcriptional Study Unveils the Role of DGAT1 Gene Mutations in Congenital Diarrhea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Strategies and Methods of Hydrogels for Antitumor Drug Delivery

1
Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Namiki 1-1, Tsukuba 305-0044, Ibaraki, Japan
2
Graduate School of Science and Technology, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan
3
Graduate School of Life and Environment Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Ibaraki, Japan
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(8), 1899; https://doi.org/10.3390/biomedicines13081899
Submission received: 27 June 2025 / Revised: 25 July 2025 / Accepted: 1 August 2025 / Published: 4 August 2025
(This article belongs to the Section Drug Discovery, Development and Delivery)

Abstract

Tumor treatments have substantially advanced through various approaches, including chemotherapy, radiotherapy, immunotherapy, and gene therapy. However, efficient treatment necessitates overcoming physiological barriers that impede the delivery of therapeutic agents to target sites. Drug delivery systems (DDSs) are a prominent research area, particularly in tumor therapy. This review provides a comprehensive overview of hydrogel-based DDSs for tumor treatment, focusing on the strategies and designs of DDSs based on the unique pathophysiological characteristics of tumors. The design and preparation of hydrogel systems for DDSs are summarized and highlighted. The challenges and opportunities for translating hydrogel-based DDSs into clinical applications are discussed.

1. Introduction

Drug delivery systems (DDSs) have attracted considerable attention, particularly in the context of antitumor treatments. A DDS is a formulation or device that enables the introduction of therapeutic substances into the body and improves treatment efficiency and safety by controlling the rate, timing, and location of drug release [1]. The sustained drug mechanism of DDSs is more conducive to eliminating tumor cells compared with that using free drug administration [2,3,4]. Furthermore, during antitumor therapy, the DDS improves local drug accumulation, which enhances therapeutic efficacy and reduces off-target toxicity to normal cells, thus alleviating safety issues [5,6].
Currently, a variety of carriers are used in DDSs for tumor therapy, including liposomes, nanoparticles (NPs), and hydrogels [7,8,9]. Among them, liposomes have achieved clinical success and several formulations have already been approved for use. For example, Doxil was the first liposomal product approved by the FDA for ovarian tumor treatment in 1995. More recently, Vyxeos is a new liposomal product that combines two currently used chemotherapies (daunorubicin and cytarabine) for leukemia treatment [10]. Other nanocarriers, such as dendrimers and inorganic NPs, have also been explored for DDS applications [11,12]. Despite the differences in their mechanisms and applications, most DDSs focus on three primary goals: achieving controlled drug release, targeting specific release sites, and maintaining the therapeutic potency of drugs. However, liposomes and NPs have limitations, including low drug entrapment efficiency, endosomal entrapment, and rapid clearance by the mononuclear phagocyte system, which ultimately diminishes their antitumor efficacy [13,14,15,16]. Therefore, alternative biomaterials capable of enabling controlled drug release are required to overcome these drawbacks.
Hydrogels, a class of water-swollen polymer networks composed of macromolecules crosslinked either physically or chemically, are promising candidates for DDSs. Hydrogels have received substantial attention as drug delivery devices in medical applications, especially in antitumor treatment, owing to their high water content, excellent biocompatibility, combinatorial optimization potential, and adaptable structure.
For example, hydrophilic drugs, such as vitamin C (Vit C), often face challenges due to rapid excretion, limiting their therapeutic effectiveness. To address this issue, Zhang et al. developed amphiphilic vitamin C self-assembled nanofiber hydrogels to achieve controlled release. By chemically modifying Vit C with a hydrophobic alkyl chain linked via an ester bond that is enzymatically degradable in vivo, hydrogels are formed through hydrogen bonding and hydrophobic interactions and can be used as Vit C depots. Approximately 75% of Vit C was released over 10 days, demonstrating a controllable release profile and improved antitumor effects [17]. In another example, alginate–pectin hydrogels were prepared using ionic crosslinking for the oral delivery of resveratrol (RES) to the intestines. Alginate hydrogels remained stable in acidic environments but degraded under alkaline conditions, whereas pectin enhanced the mechanical strength of the hydrogels. This approach protects RES from stomach degradation, enables targeted delivery to the intestine, and improves therapeutic outcomes [18].
Moreover, hydrogels combined with liposomes can markedly enhance drug delivery efficiency. Li et al. developed a multilevel DDS platform in which drugs were encapsulated in liposomes and subsequently coated with thiolate chitosan to form a liposomal hydrogel. Hydrophobic curcumin was loaded into liposomes to enhance its solubility, enabling localized drug delivery. This system demonstrated superior antitumor efficacy compared with that of free drugs or liposomal drugs alone, effectively inhibiting tumor recurrence and promoting tissue repair [19]. Hydrogels have been extensively studied as drug delivery devices for disease treatment.
Despite extensive research on hydrogel-based DDSs, most existing reviews have focused on their fundamental properties and general applications [20,21,22]. Few reviews have focused on the strategies and methods for designing hydrogels specifically for antitumor drug delivery. Hence, this review summarizes the strategies and preparation methods for hydrogels used in antitumor therapy, with an emphasis on common approaches for evaluating hydrogel-based DDSs both in vitro and in vivo (Figure 1).

2. Design of Hydrogel-Based DDSs

Hydrogel materials are broadly categorized as naturally derived, semisynthetic, and synthetic polymers [23]. Naturally derived polymers such as collagen, gelatin, and fibrin exhibit advantageous properties, including biodegradability, low cytotoxicity, and a minimal immune response in vivo. Additionally, many naturally derived polymers facilitate cellular adhesion, making them suitable for applications in tissue engineering and localized DDSs [24,25,26]. Commonly used synthetic polymers exhibit good mechanical features, plasticity, and variable properties for on-demand applications [27,28]. Following this development [29], polymers frequently used to prepare hydrogel-based DDSs are summarized in Table 1.
Table 1. Polymers and preparation methods for hydrogel-based DDSs after 2019.
Table 1. Polymers and preparation methods for hydrogel-based DDSs after 2019.
PolymersLoading DrugsPreparing MethodDegradation and StabilitySterilization MethodIn Vitro/In Vivo AnalysisBiocompatibilityReference
Naturally derived polymersGelatin and alginateDoxorubicin (DOX)Chemically crosslinking by “Shift-Base” condensation reaction/No description while Schiff base has part of antibacterial effectIn vitroGood biocompatibility (less than 10% decrease in cell viability of no-drug-loading hydrogel)[30]
Bisphosphonate-functionalized hyaluronic acidDOXChemical interaction of bisphosphonate–zinc60% mass decrease in drug-loading hydrogel after 316 h in medium/In vitro and in vivoGood biocompatibility (less than 5% decrease in cell viability of no-drug-loading hydrogel)[31]
Alginate sodium carboxymethyl celluloseMethotrexate (MTX) and aspirin (AP)Physical crosslinking42% mass decrease after 8 days in PBS/In vitroGood biocompatibility (less than 5% decrease in cell viability of no-drug-loading hydrogel)[32]
Alginate and magnetic hydroxyapatite5-fluorouracil (5-FU)Physical crosslinkingIncreasing light and temperature reduces the storage stabilityFiltrationIn vitroGood biocompatibility (less than 15% decrease in cell viability of no-drug-loading hydrogel)[33]
Synthetic polymersPoly (D, L-lactide-co-glycolide)-poly(ethylene-glycol)-poly (D, L-lactide-co-glycolide), beta-cyclodextrinDOX and curcuminPhysical crosslinking13.3% mass decrease after 24 h in PBS/In vitro and in vivoGood biocompatibility (less than 10% decrease in cell viability of no-drug-loading hydrogel)[34]
Methacrylic acid (MAA)Camptothecin (CPT)Chemically crosslinking through disulfide linkage between CPT and MAAStable in normal physiological environment; quickly shrinks to a smaller volume in low pH environment of tumor tissue or tumor cells/In vitro and in vivoGood biocompatibility (less than 20% decrease in cell viability of no-drug-loading hydrogel)[35]
Polyethylene glycol (PEG)DOX and curcuminChemically crosslinkingInstability/In vitroGood biocompatibility (less than 10% decrease in cell viability of no-drug-loading hydrogel)[36]
Polyacrylamide (PAM) and carbon nanotubeDOXPhysically crosslinking by hydrogen bondStable in PBS after 80 h/In vitroGood biocompatibility (less than 20% decrease in cell viability of no-drug-loading hydrogel)[37]
Combination and/or modified polymersChitosan, polypyrroleDOXChemically crosslinking by “Shift-Base” condensation reactionStable in PBS (both pH 7.4 and 6.5) after 7 days/In vitro and in vivoGood biocompatibility (less than 3% decrease in cell viability of no-drug-loading hydrogel)[38]
PEG-modified bovine serum albuminPaclitaxel (PTX)Physical crosslinking of PEG-BSA46% mass loss in PBS after 50 days. Degraded completely within 200 days in vivo/In vitro and in vivoNo toxicity of hydrogel[39]
Carboxymethyl arabinoxylan5-FUPhysically crosslinking5 to 15% mass decrease after 7 days in PBS, 37 °CThe nanocomposite hydrogel has antibacterial activityIn vitroDegradation leads to cell death[40]
Carboxymethyl chitosan and polyvinyl alcoholOxaliplatinChemically crosslinking by free-radical polymerization//In vitro and in vivoGood biocompatibility by acute oral toxicity assay[41]
Ultrasmall peptideDOXDimerization of the PyKC peptideHigh stability after 18 days in vivo/In vitro and in vivoGood biocompatibility (less than 5% decrease in cell viability of no-drug-loading hydrogel)[42]
Thiol-modified hyaluronic acid (HASH) and vinyl sulfone-modified β-cyclodextrinDOXChemically crosslinking of a “click reaction” between thiol and vinyl sulfone groupsKeep stable after 15 days in PBS while degraded after 5 days in enzymatic conditions/In vitroGood biocompatibility[43]
N-isopropylacrylamide (NIPAAm) and maleic anhydride (MA) copolymer (poly (NIPAAm-co-MA)) chitosanMTX and curcuminPhysical crosslinkingOver 80% weight loss after 28 days in PBS/In vitroGood biocompatibility (less than 10% decrease in cell viability of no-drug-loading hydrogel)[44]
Although simple polymer hydrogels have been successfully used in DDSs, hybrid hydrogels composed of combinations of different polymers provide enhanced versatility [45]. Regardless of the type of polymer used to prepare the hydrogels, environment-responsive functionalities and cargo-specific designs are the two key considerations that dictate polymer selection and modification to achieve efficient and safe drug delivery in clinical applications.

2.1. Stimulus-Responsive Drug Delivery Hydrogels

The tumor microenvironment (TME) significantly differs from normal tissues and is characterized by unique features, such as low pH, hypoxia, thermal sensitivity, and even different stiffness or viscosity of the extracellular matrix (ECM) [46]. These distinct properties of the TME not only influence the therapeutic response and clinical outcome [47] but also provide opportunities to design materials that specifically target the TME and physiological conditions, thereby improving safety and therapeutic efficacy [48]. Accordingly, stimuli-responsive materials have been extensively studied for DDSs.
Using the acidic pH of the TME to develop pH-responsive hydrogels is one of the most widely studied strategies (Table 2). The extracellular pH in tumor sites typically ranges from 6.5 to 7.0, caused by the high metabolic rate of tumors [49], whereas the pH value of normal tissues maintains a pH of approximately 7.4 [50]. pH-sensitive hydrogels exploit this difference to enhance drug release, specifically at tumor sites, thereby improving localized drug concentration and minimizing systemic toxicity. For instance, Qu et al. developed a hydrogel for the delivery of doxorubicin (DOX), a commonly used first-line antitumor agent. The hydrogel was synthesized using N-carboxyethyl chitosan (CEC) via the Michael reaction in an aqueous solution, combined with dibenzaldehyde-terminated poly (ethylene glycol) (PEGDA). Under acidic conditions, the protonated and positively charged amino groups of chitosan weaken the Schiff base bonds between -NH2 and -CHO, accelerating hydrogel degradation and enabling faster drug release in an acidic environment than in a neutral environment [51]. Qing et al. designed a hydrogel using dopamine, carboxymethyl cellulose (CMC), and hydroxyethyl cellulose to deliver the antibacterial drug, ciprofloxacin (CIP). In this system, the hydrolysis of the amide bond between CMC and CIP is the primary mechanism controlling drug release and is highly sensitive to pH changes [52]. Mukherjee et al. demonstrated that acidic environments not only increased the swelling ratio of the hydrogel but also facilitated the breakage and degradation of ester bonds within the polymer matrix, thus enhancing drug release [53].
The design of pH-responsive hydrogels typically aims to modulate the drug release profile by accelerating hydrogel degradation or weakening the crosslinking bonds of the hydrogel under acidic conditions (Table 2). By targeting the mildly acidic pH of the TME, these hydrogels can effectively concentrate drugs at tumor sites and help reduce systemic side effects. However, the sensitivity of some hydrogels to pH changes is a critical challenge. Some of the drug release curves exhibited significant differences with physiological conditions (pH around 7.4) only when the pH value decreased to 4.0, or even lower [52,53,54,55], which is much lower than the typical pH range of the TME (pH around 6.5 to 7.0). Such limitations may hinder their effectiveness, as the pH at most tumor sites may not be sufficiently low to induce drug release, potentially compromising therapeutic outcomes or causing off-target effects.
Table 2. pH-sensitive hydrogels for DDSs.
Table 2. pH-sensitive hydrogels for DDSs.
Loading DrugsPolymersDDS MechanismDrug Release pHDegradation and StabilitySterilization MethodIn Vitro/In Vivo AnalysisReference
DOXNitrogen-doped carbon quantum dots and hydroxyapatiteDisintegration of the Schiff base bond and dissolution of hydroxyapatite in acidic conditions6.5–4.0/The introduction of HA by either chemical bonding or physical incorporation can result in hydrogels with enhanced antimicrobial activityIn vitro[56]
DOXN-carboxyethyl chitosan (CEC) and Di benzaldehyde-terminated poly (ethylene glycol) (PEGDA)Disintegration of the Schiff base bond6.8–4.0Around 30% mass loss in PBS 7.4 and 50% mass loss in PBS 5.5 after 200 h/In vitro[51]
DOXSericin, rice bran albumin, and gellan gumDegradation in the ester bond and increase in the swelling ratio of the hydrogel5.0–4.0//In vitro[53]
ProspidineDextran phosphateDiffusion of the cytostatic bond and the destruction of the polymer4.0–1.2100% mass loss after 30 days in PBS/In vitro and in vivo[54]
DOXGlycol chitosan–Pluronic F127 and α-CDDissociation of the hydrogel5.0100% mass loss after 220 to 260 h in PBS /In vitro and in vivo[55]
BortezomibA ‘ABA’ triblock copolymer of phenylboronic acid-functionalized polycarbonate/poly (ethylene glycol)Dissociations of boronated ester bond5.8At pH 7.4, the BTZ release from the micelle/hydrogel composite remained low at 7%, an acidic environment, ∼85% of BTZ was released over 9 days/In vitro and in vivo[57]
DOXDextran-based nanogelDisintegration of the Schiff base bond5.0–2.0//In vitro[58]
DOXCarboxymethyl chitosanHydrolysis of ortho ester bond6.5–5.0Around 30% mass loss after 300 h in PBS 7.4 and around 40% mass loss after 300 h in PBS 5.0/In vitro[59]
Gemcitabine, paclitaxel (PTX)OE peptide (VKVKVOVK-VDPPT-KVEVKVKV-NH2)Disruption of the 3D network of peptide from beta-sheet to random coil5.8//In vitro and in vivo[60]
Drug release pH: the pH condition used to measure the drug release profile from the DDS. DDS, drug delivery system; DOX, doxorubicin.
In contrast to pH-responsive hydrogels, thermosensitive hydrogels represent a class of physically stimuli-responsive hydrogels, designed with a particular focus on injectability. These hydrogels remain in a liquid state before injection and undergo gelation to the solid state at physiological temperatures (37 °C) [61,62,63,64]. This property allows thermosensitive hydrogels to be directly injected into tumor sites, where they provide sustained drug release. The application of these hydrogels minimizes the toxicity caused by drug diffusion and, more importantly, avoids the extensive tissue damage caused by surgical implantation. Additionally, the swelling and collapsing processes of thermosensitive polymer networks near their critical temperature have been used as a mechanism for drug delivery [65] due to the fact that therapeutic agents are commonly encapsulated within hydrogels, which results in a drug release profile governed by constrained diffusion of the agents through hydrogel networks [66].
Poly (N-isopropylacrylamide) (PNIPAAm) is one of the most commonly used polymers for preparing thermosensitive hydrogels. The sol–gel transition is driven by reversible changes between the hydrated and dehydrated states of PNIPAAm as the temperature crosses its lower critical solution temperature (LCST), typically below 37 °C [67]. Liu et al. developed an alginate-g-poly (N-isopropylacrylamide) copolymer system by conjugating PNIPAAm to alginate. This system remains dissolved in water or phosphate-buffered saline buffer at room temperature but self-assembles into a hydrogel when heated to body temperature (37 °C). DOX loaded onto a hydrogel exhibited sustained drug release and achieved significant antitumor effects against multidrug-resistant AT3B-1 cells [61]. Similarly, Luo et al. synthesized a PNIPAAm-coacrylic acid-g-F68 copolymer hydrogel for triptolide delivery. This hydrogel maintained a liquid state at room temperature and showed a marked increase in storage modulus when the temperature exceeded 35 °C. The antitumor agent, triptolide, was encapsulated in nanomicelles at room temperature and released intratumorally after injection. In animal models, the hydrogel exhibited superior antitumor efficacy and low toxicity compared with that of free drug administration [62].
In addition to the sol–gel translation behavior of thermosensitive hydrogels, the swelling and shrinking properties of hydrogels at their LCST have been widely used to control drug release [65,68,69]. For instance, PNIPAAm-based hydrogels typically exhibit LCST below 37 °C, collapsing and shrinking above the LCST while swelling and expanding below the LCST. This reversible swelling and shrinking behavior can be used in DDSs to control drug release profiles [70]. Lei et al. further advanced the design of thermosensitive hydrogels by developing a near-infrared (NIR)-triggered hydrogel for on-demand drug delivery. This thermosensitive chitosan (chitosan-g-PNIPAAm) polymer was synthesized by grafting PNIPAAm onto chitosan (chitosan-g-PNIPAAm) via free-radical copolymerization, followed by UV-induced crosslinking with methacryloyl groups. The resulting composite hydrogels exhibited significant shrinkage at elevated temperatures (45 °C). By incorporating photothermal carbon, the hydrogel could trigger the shrinkage of polymer networks upon NIR irradiation and thus release the loaded drug [71]. Although the composite hydrogels exhibited NIR-triggered behavior, the burst release profile of DOX following each exposure to 45 °C may negatively impact treatment outcomes, and the lack of detailed experimental data limits the evaluation of its clinical applicability. A critical mechanism underlying drug release control via the LCST is that below the LCST, the swelling of the hydrogel increases the diffusion distance for the encapsulated drugs, thus slowing their release. Conversely, above the LCST, the hydrogel network collapses, reducing diffusion resistance and accelerating drug release [65,70]. Therefore, it is essential to clarify the diffusion rate of drugs at various environmental temperatures because high environmental temperatures may accelerate drug release, potentially leading to misleading conclusions regarding hydrogel performance. A detailed investigation of these factors is crucial for optimizing LCST-based hydrogel systems for clinical use.

2.2. Cargo-Based Drug Delivery Hydrogels

The properties of hydrogels can be designed for DDSs using different materials; however, the encapsulation efficiency and release rates of drugs from hydrogels are influenced by multiple factors, even when using the same hydrogel matrix. Properties, such as hydrophilicity or hydrophobicity [72], electrostatic interactions [73], and molecular weight [74] significantly affect the drug delivery efficiency and antitumor efficacy. Consequently, the design and preparation of hydrogel-based DDSs must consider the physicochemical properties of the drugs and their specific delivery environment.
Reportedly, over 40% of marketed drugs exhibit poor water solubility, and approximately 60% of the compounds emerging from pharmaceutical research laboratories are classified as insoluble [75]. Among the challenges of hydrogel-based DDSs, the delivery of hydrophobic drugs via high-water-content hydrogels remains one of the most significant obstacles. Poor drug encapsulation efficiency and a low release rate often result in reduced bioavailability and therapeutic outcomes [76,77].
Paclitaxel (PTX), a widely used first-line antitumor agent with broad-spectrum antitumor effects, addresses these challenges. PTX functions as an anti-microtubule agent, binding specifically to the β-subunit of tubulin to inhibit microtubule disassembly, thereby arresting mitosis and suppressing tumor proliferation. In contrast, DOX hydrochloride (DOX · HCl), a water-soluble anthracycline, can bond to nucleic acids and form complexes with DNA by intercalating to base pairs to inhibit the function of topoisomerase II activity and then present antimitotic and cytotoxic effects. Owing to the hydrophilic and hydrophobic differences between these drugs, their release behaviors in the same DDS are markedly different. For example, in a study by Xu et al., the cumulative release of PTX from a hydrogel was only approximately 10% after 24 h, whereas DOX showed a much faster release and resulted in higher cytotoxicity against tumors than PTX after 48 h of culture. Conversely, the inhibition of cell proliferation by PTX was notably weaker even after 72 h, likely because of its low drug release rate [76]. Similarly, docetaxel (DOC), another taxane derivative with potent antitumor effects, is commercially available only as an intravenous formulation owing to its poor water solubility [78]. To address these limitations, strategies, such as combining hydrogels with liposomes, NPs, or microspheres, are commonly used as delivery tools for hydrophobic drugs. Furthermore, modifying the water solubility of drugs is commonly used (Table 3).
Amphiphilic polymers are particularly advantageous for delivering hydrophobic drugs. Their ability to self-assemble via hydrophobic interactions facilitates hydrogel formation and enhances drug encapsulation within the hydrogel. Rezazadeh et al. encapsulated PTX into mixed polymeric micelles composed of PF127 and tocopherol polyethylene glycol 1000 succinate (TPGS), which were subsequently incorporated into a hyaluronic acid (HA)-based hydrogel for the co-delivery of PTX and DOX. TPGS, with its amphiphilic structure, formed micelles in aqueous media and achieved a PTX entrapment efficiency of approximately 51%. Although the initial PTX loading affected the release rate, more than 50% of the drug was released from the hydrogel within 60 h, demonstrating efficient drug loading and release capabilities [79]. Although the study did not directly evaluate the antitumor efficacy of the hydrogel, subsequent in vitro and in vivo studies of similar DDSs demonstrated significant therapeutic effects [80].
The host–guest effect of cyclodextrin (CD)–drug complexes has been extensively explored as a strategy to improve the water solubility of hydrophobic drugs. CDs are cyclic oligosaccharides composed of D-glucopyranoside units linked via alpha-1,4 glycosidic bonds. The most commonly used CDs are alpha-CDs, beta-CDs, and gamma-CDs, which consist of six, seven, and eight glucose repeating units, respectively. The arrangement of these molecular structures forms a hydrophobic inner cavity and hydrophilic exterior [81], enabling CDs to encapsulate hydrophobic guest molecules within the cavity, thereby enhancing their physicochemical properties and aqueous solubility [82,83,84].
Nieto et al. used a host–guest reaction to load PTX into beta-CDs to form PTX: beta-CD inclusion complexes, which were subsequently encapsulated within a gellan gum hydrogel. The hydrogel was synthesized in different buffer systems (acetate and phosphate) and crosslinked with varying concentrations of L-cysteine using the crosslinker EDC/NHS to provide redox-responsive properties to glutathione (GSH). In drug release studies, approximately 50% of PTX was released from the hydrogel within 50 h under physiological conditions. Furthermore, the presence of L-cysteine and GSH allowed for adjustable drug release rates, demonstrating promising control over drug delivery and significant antitumor efficacy in vitro [85]. In another study, Ma et al. developed a beta-CD/camptothecin inclusion complex that was incorporated into folic acid hydrogels. The folic acid was chemically modified with NaOH and then crosslinked with ZnCl2 using a metal-ion-induced gelation method. The prepared CPT-beta-CD complex was added to the folic acid solution to form drug-loaded hydrogels [86]. This system leverages the selective affinity of folic acid for tumor cells, enhancing the targeting and delivery efficiency of CPT while mitigating off-target effects.
Notably, preclinical studies have demonstrated that CD–drug inclusion complexes often exhibit superior therapeutic performance compared with that of free drug administration [87,88,89]. However, most CD-based formulations are administered intravenously rather than incorporated into hydrogels for local drug delivery. Intravenous administration typically results in uncontrolled drug release, leading to systemic drug accumulation in non-target tissues and organs, which increases the risk of toxicity [90,91]. In contrast, combining CD–drug complexes with hydrogels provides an opportunity to leverage the sustained and localized release characteristics of hydrogels. This approach can enhance antitumor efficacy while minimizing systemic toxicity, particularly for hydrophobic drugs that pose substantial formulation challenges.
Furthermore, the application of CDs in local drug delivery commonly involves the formation of host–guest supramolecular hydrogels rather than the direct modification of drug properties. These supramolecular hydrogels have shown substantial potential for tumor therapy by providing tunable release kinetics, responsive behavior, and enhanced mechanical stability [92,93,94]. Combining drug complexes with hydrogel systems may optimize their therapeutic performance and clinical utility.
Table 3. Hydrogels for hydrophobic drug delivery.
Table 3. Hydrogels for hydrophobic drug delivery.
Hydrophobic DrugsMaterials to Synthesize HydrogelsDrug-Loading MechanismDrug Entrapment EfficiencyDrug-Loading EfficiencyDegradation and StabilitySterilization MethodIn Vitro/In Vivo AnalysisReferences
PTXMixed polymeric micelle composed of PF127, tocopherol polyethylene glycol 1000 succinate (TPGS), and hyaluronic acid (HA)Micelle formation by hydrophobic interaction~51%~12%Hydrogel maintained 75% of its original weight over 5 days at 37 °C, low viscosity at 4 °C converted to a semisolid upon heating to 35 °C/[76] In vitro
[77] In vitro and in vivo
[79,80]
PTXGellan gumFormed CD: drug complex///Sterilized by UV radiationIn vitro[85]
PTXPEGylated star polymer and PNIPAAmFormed CD: drug complex~90%~3%Complex nanoparticles experienced fast size broadening under physiological salt conditions (150 mM) within 30 min/In vitro and in vivo[95]
PTXGellan gum modified by prednisoloneHydrophobic interaction by prednisolone~40%/Good stability/In vitro[96]
PTXIC1-R peptideBeta-folded structure formation by hydrophobic interaction~98%/Parameters set to a frequency of 0.1–100 rad/s, a sheer force of 1%, and a test time of 15 min. Fixed frequency of 6.28 rad/s, 0–5 min shear force set to 1%, and 5–7 min set to 100%. The shear force was set to 1% at 7–37 min, and the process was repeated at 37–70 min Filtration via 0.22 μm filterIn vitro and in vivo[97]
PTX, epirubicinHA and poly (e-caprolactone)-poly (ethylene glycol)-poly-(e-caprolactone) (PCL-PEG-PCL)Loading PTX to PCL-PEG-PCL nanoparticles~90%~10%The HA-Gel embedded subcutaneously in mice gradually degraded 3 days post-implantation, and the extent of degradation was significant on day 5. Almost no residual hydrogel on day 9/In vitro and in vivo[98]
PTXPEG-PCL-PEG/DDP + MPEG-PCL/PTXLoading PTX by monomethoxy PEG-PCL~99%~4%When the temperature exceeded 25 °C, the samples changed from a liquid to an elastic gel-like substance at the crossover point of G and G; the composite’s viscosity was the strongest at about 43 °CThe prepared PDMP hydrogel composite was sterilized using CO-60 (20 Gy) before injectionIn vitro and in vivo[99]
CurcuminThiolated chitosanEncapsulated by liposomes~88%~4%[2] The deformation loss ratio was 20.06% (CSSH Gel), 23.92% (100 μM), 20.95% (150 μM), and 17.95 % (200 μM) after five cycles of compression [19]. The best compressive performance is at 5%, the compressive modulus of 5% is 27.44 kPa, and the maximum compressive strength is 32 kPa/In vitro and in vivo[2,19]
CurcuminPeptide (MAX8)Beta-hairpin structure formation by hydrophobic interaction//Stable colloidal dispersion system and good mechanical properties/In vivo[100]
CurcuminPolyethylene glycol (PEG) and polycaprolactone (PCL) polymer PCL-PEG-PCLHydrophobic interaction by hydrophobic PCL47~74%2~9%//In vitro[101]
CurcuminAlginate/chitosan hydrogel microparticlesLoading curcumin by hyaluronic acid/zein nanoparticles~70%/Cur/MPS in simulated gastric fluid (SGF) (pH 2) has no obvious change; NMPs can decompose rapidly in colonic fluid/In vitro and in vivo[102]
Camptothecin (CPT)Pluronic F127 and alpha-CDsMicelle formation by hydrophobic interaction///Filtration via 0.45 μm filterIn vitro and in vivo[92]
CPTFolic acid and beta-CDsFormed CD: drug complex//Good rheological mechanical property and thermodynamic stability/In vitro[86]
TriptolidePNIPAm-g-pluronic F68Micelle formation by hydrophobic interaction~84%~5%0.45 mg/kg TPL-equivalent dose three times over 14 days in 4T1 tumor-bearing miceFiltration via 0.45 μm filterIn vitro and in vivo[62]
DOX (DOX·HCl deprotonated at pH 9.6)PEG-b-PCL and alpha-CDsMicelle formation by hydrophobic interaction72~74%~15%//In vitro[103]
DOCMixed polymeric micelle composed of PF127, PL121, and hyaluronic acid (HA)Micelle formation by hydrophobic interaction~99%~2%//In vivo[104]
DOC, docetaxel; DOX, doxorubicin; PTX, paclitaxel.
In addition to hydrophobic interactions and host–guest mechanisms, other advanced tools, such as NPs and liposomes, are commonly used in hydrogel-based DDSs [105,106]. Compared with the individual use of these tools, integration with high-water-content hydrogels for in situ delivery can provide dual benefits. The hydrogels enable localized and controlled drug release and simultaneously serve as a postoperative filler, offering additional benefits, such as promoting tissue regeneration or reducing inflammation at surgical sites.
Compared with free drug administration, the application of hydrogels is expected to fulfill at least one of the following three primary functions: (1) mitigating burst release to extend the therapeutic window and improve treatment efficacy; (2) minimizing systemic toxicity by reducing the drug dosage or enhancing local drug accumulation, thereby improving the survival curve; and (3) addressing drug incompatibility issues through spatiotemporal control of drug release. The first two functions are easily understood; however, the third requires an understanding of the complexity of tumor therapy. It is highly relevant to the common properties of tumors, including inherently heterogeneous and diverse genetic, epigenetic, and phenotypic variations that enable them to adapt and develop resistance to single-agent therapies. Moreover, not only can cancer cells develop drug resistance [107], but changes in the TMEs can contribute to single-drug resistance, thus leading to treatment failure (Figure 2) [108,109]. Therefore, combination therapy has been widely adopted for the treatment of various tumors in clinical settings. Established clinical protocols, such as FOLFOX (folinic acid, fluorouracil, and oxaliplatin) for colorectal cancer (CRC) and CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) for non-Hodgkin lymphoma, exemplify the effectiveness of multi-agent regimens that significantly improve patient outcomes [110,111].
Despite its advantages, combination therapy presents challenges, such as increased complexity in pharmacokinetic and pharmacodynamic interactions, heightened risk of overlapping toxicities, and the need for precise dose optimization to prevent adverse effects [112,113]. Therefore, the development of advanced DDSs, including hydrogel-based platforms, has become popular for ensuring the controlled and targeted release of therapeutic agents in combination regimens.

2.3. Drug Delivery Hydrogels for Pathological Characteristics of Tumors

The distinct TME can influence drug delivery efficiency and is increasingly recognized as a unique characteristic that can be used in the design of DDSs, such as hypoxia, acidity, ultrahigh GSH levels, and the overexpression of specific enzymes [114]. Another important aspect is tumor heterogeneity, which also affects drug delivery and treatment efficiency yet remains an unresolved challenge. Heterogeneity refers to the various genetic and non-genetic phenotypes that emerge during tumorigenesis, progression, and treatment, as well as interpatient variability, even within the same tumor type [115,116]. Tumor vasculature is characterized by chaotic organization, high permeability, and substantial heterogeneity [117]. Additionally, cancer genotypes can vary across different microenvironments, possibly because of the different distributions of cytokines around tumor sites [118]. Moreover, significant spatial discordance in programmed death ligand 1 (PD-L1) expression has been observed between primary tumors and lymph node metastases in surgically resected gastroesophageal adenocarcinoma [119]. The heterogeneity of tumors not only reduces drug delivery efficiency by the disorganized vasculature [120] but also contributes to treatment failure through the emergence of drug-resistant cells and metastasis [121]. It also results in conflicting therapeutic outcomes and the unpredictability of clinical prognosis [119]. In single-agent DDSs, tumor heterogeneity leads to difficulties in precise targeted therapy [122], thereby compromising the effectiveness of hydrogel-based DDSs and diminishing therapeutic returns. Therefore, the development of combination therapy strategies using hydrogels has become a prevailing direction in efforts to enhance treatment efficacy [123,124,125].

2.3.1. DDSs of Cytotoxic Drugs

DOX is a model drug widely used in DDSs and acts via multiple mechanisms [126]. Despite its effectiveness, the non-negligible cardiotoxicity of both dose- and schedule-dependent DOX significantly limits its clinical application to a substantial extent [127]. Additionally, short-term treatment with DOX demonstrated a low antitumor effect [128]; therefore, long-term treatment is recommended but is associated with cumulative toxicity, followed by reduced therapeutic utility and eventual exhaustion of antitumor effectiveness, as evidenced by tumor regrowth typically observed at approximately 20 days post-administration in most in vivo experiments [129,130,131]. Collectively, efforts to mitigate the systemic toxicity of DOX, such as reduction or altered administration schedules, show potential, but often compromise the required therapeutic efficacy. Conversely, high-dose administration enhances antitumor outcomes but exacerbates toxicity [127,130,132]. To balance these competing factors, the combination of DOX with other antineoplastic agents has emerged as a promising strategy, particularly for hydrogel-based DDSs (Table 4).
Zhao et al. developed an injectable hydrogel system composed of glycol chitosan (GC) and amphiphilic hydrogels (OHC-PEO-PPO-PEO-CHO) to co-deliver DOX and PTX. The system showed a burst release of DOX and a sustained release of PTX owing to differences in their water solubility. Although the combination of DOX and PTX did not yield significantly enhanced antitumor efficiency, which is likely due to the asynchronous release profiles of the two drugs, the toxicity profile was not exacerbated by the prolonged survival time in comparison with a single DDS [133]. In another study, PTX and DOX were loaded with PECT and PEPF, respectively, and NPs prepared using nanoprecipitation technology were incorporated into hydrogels. A hydrogel containing drug-loaded NPs was then synthesized and achieved better antitumor efficacy and reduced systemic toxicity compared with free drug administration [134].
In addition to DOX, cisplatin is another important chemotherapeutic agent used to treat various malignancies, including sarcoma, carcinoma, and lymphoma. Cisplatin, or cis-diamminedichloroplatinum (II) (CDDP), is a platinum-based chemotherapeutic agent that exerts antitumor effects by forming DNA crosslinks, thereby disrupting DNA synthesis and transcription to induce tumor cell apoptosis. Despite its antitumor effects, the widespread uptake of CDDP by both normal and tumor cells via ubiquitously expressed transporters results in significant side effects, thereby limiting its application [135]. For instance, the specific expression of organic cation transporter-2 (OCT2) in the kidneys may explain the nephrotoxicity of CDDP [136,137]. The high water solubility and rapid burst release of CDDP at the time of administration further contribute to its systemic toxicity and suboptimal antitumor efficacy. Hydrogel-based DDSs have been used to address these challenges by retarding the drug release rate and enabling the localized delivery of drugs to tumor sites, thus improving therapeutic outcomes while avoiding off-target effects [138,139,140]. For example, a study by Li et al. demonstrated that incorporating CDDP into thermosensitive hydrogels not only provided controlled and sustained release but also enhanced tumor suppression while significantly reducing renal toxicity in animal models [141].
As for the co-administration of CDDP and other therapeutic agents, Chen et al. developed a co-delivery system using a biodegradable temperature-sensitive hydrogel (PDLLA-PEG-PDLLA, PLEL) for combination chemotherapy of gastric tumors. The thermosensitive hydrogel not only enhanced local combination therapy with 5-fluorouracil (5-FU) and CDDP but also demonstrated significantly better antitumor efficacy than that of the free administration of 5-FU and CDDP. Moreover, the hydrogel promoted the local accumulation of 5-FU and CDDP. Moreover, the hydrogel system promotes the localized accumulation of drugs, reduces systemic exposure, and extends the overall survival time of treated animals with minimal tissue damage and reduced systemic toxicity [138]. Similarly, Shen et al. investigated a self-assembled poly (ethylene glycol) (PEG)/polyester copolymer hydrogel for the co-delivery of CDDP and PTX to ovarian tumor models. This hydrogel-based DDS demonstrated low systemic toxicity, as evidenced by minimal weight loss in the experimental animals. Although PTX showed a slower release rate than that of CDDP, both drugs maintained a steady release profile throughout the study, contributing to enhanced antitumor efficacy through sustained therapeutic concentrations at the tumor site [139].
To further enhance antitumor efficiency, other chemotherapeutic agents with distinct mechanisms, such as topotecan, SN-38, and 5-FU, have also been explored in hydrogel carrier systems [142,143,144,145]. These studies highlighted the multiple advantages of hydrogel-based DDSs for combination therapy. First, hydrogels mitigate burst release effects and reduce the systemic toxicity associated with free drug administration. By tailoring the hydrophilic and hydrophobic properties of the hydrogel matrix, controlled and localized drug delivery can be achieved, thereby improving the therapeutic index. Second, the differential solubility of the co-administered drugs can be leveraged to regulate their overall concentrations at the target site, ensuring sustained exposure and enhanced synergistic effects.
Additionally, hydrogels enable the sequential or simultaneous delivery of multiple drugs, facilitating combination therapies that synergistically exploit distinct antitumor mechanisms. This approach is particularly effective for overcoming drug resistance because it disrupts adaptive cellular responses and maintains therapeutic pressure. Furthermore, hydrogels can bridge pharmacokinetic gaps in drug metabolism cycles by providing controlled release and preventing therapeutic windows in which tumor cells can recover or proliferate.
By co-delivering hydrophilic and hydrophobic drugs on a single platform, hydrogel systems can maximize the efficacy of chemotherapeutic agents while minimizing off-target effects. These systems exemplify a powerful strategy for achieving precise and effective combination chemotherapy, paving the way for personalized and tolerable cancer treatments.
Table 4. Combinations of different antineoplastic agents.
Table 4. Combinations of different antineoplastic agents.
TumorDrugsMaterials to Synthesize HydrogelsEffectsReference
MelanomaDOX and PTXGlycol chitosan and benzaldehyde-terminated polymerHigh antitumor efficacy and safety[133]
Colon carcinomaDOX and DOCMicelles by PL121, PF127, and HAHigh antitumor efficacy and safety[104]
Bladder carcinomaDOX and CDDPPEG-b-PCL and alpha-CDsControllable drug release and injectable ability[103]
HepatomaDOX and CurcuminPCL-PEG-PCLHigh antitumor efficacy and safety[101]
Lung tumorPTX and CDDPPEG-PCL-PEG and PMEG-PCLHigh antitumor efficacy, controllable drug release, and safety[99]
Ovarian tumorPTX and CDDPBi-mPEG-PLGA-Pt (IV)High antitumor efficacy and safety[139]
Breast tumorPTX and Epirubicin (EPI)PCL-PEG-PCL and HAHigh antitumor efficacy, controllable drug release, and safety[98]
Breast tumorPTX and Gemcitabine (GEM)OE peptide hydrogelHigh antitumor efficacy[60]
Breast tumorPTX and HonokiolPLGA-PEG-PLGAHigh antitumor efficacy and safety[146]
Ovarian tumorPTX, Rapamaycin, LS301PLGA-b-PEG-b-PLGAHigh antitumor efficacy[147]
Colorectal peritoneal carcinomatosisCDDP and 5-FURing-opening copolymerization to synthesize ε-CL and PEG and chitosanHigh antitumor efficacy and safety[145]
Gastric tumorCDDP and 5-FUPDLLA-PEG-PDLLAHigh antitumor efficacy, safety, and high efficacy to inhibit tumor recurrence[138]
Pancreatic tumorCDDP and GEMPDLLA-PEG-PDLLAHigh antitumor efficacy[148]
Colorectal tumorOxaliplatin and HesperetinCationic Okra gumHigh cytotoxicity[149]
Brain tumorCarmustine and CurcuminPCL-PEGHigh efficacy to inhibit tumor recurrence[150]
DOC, docetaxel; DOX, doxorubicin; HA, hyaluronic acid; PTX, paclitaxel.

2.3.2. DDSs of Anti-Angiogenesis Drugs

Generally, tumor growth is characterized by uncontrolled proliferation, which leads to a high metabolic demand, rapid nutrient consumption, and increased production of waste products. Unlike blood cancers and other nonsolid tumors, the growth of solid tumors generally requires vascularized connective tissue stroma to sustain expansion beyond their minimum size [151]. It is widely accepted that the vasculature in tumors is chaotic with high permeability and heterogeneity compared with that in normal tissues [152,153]. As tumors rely on angiogenesis for their continued growth and metastasis, angiogenesis therapy has been recognized as a critical target in tumor treatment, promoting the development of numerous anti-angiogenic agents, such as bevacizumab, cediranib, axitinib, and regorafenib [154,155]. Other anti-angiogenic agents, such as inhibitors of tyrosine kinase receptors, including sunitinib, sorafenib, and pazopanib, have found broad clinical applications [156]. Anti-angiogenic strategies transiently normalize tumor vasculature and enhance the delivery and efficacy of chemotherapeutic agents [157,158]. Furthermore, targeting tumor angiogenesis facilitates metronomic chemotherapy, a regimen of chronic, low-dose drug administration aimed at preventing angiogenesis and inducing sustained antitumor effects [159]. According to these theories, the combination of anti-angiogenesis and chemotherapy has become a widely adopted therapeutic model [160].
To address the toxicity and limited efficiency of anti-angiogenesis therapies, hydrogels have been explored for localized drug delivery and the controlled release of the drugs. Li et al. used a classic injectable thermosensitive PLGA-PEG-PLGA hydrogel to co-deliver the vascular disruptive agents, combretastatin A-4 (CA4) disodium phosphate (CA-4DP) and epirubicin. Combination therapy not only exhibited superior antitumor effects compared with that of single-drug administration but also prolonged the survival of treated animals [161].
CA4P is a water-soluble prodrug of CA4, which demonstrates robust antitumor efficacy, particularly in combination therapies for platinum-resistant ovarian tumors. This is a mechanism that induces vascular congestion and reduces tumor blood flow by changing the endothelial cell morphology to achieve treatment outcomes [162]. However, the vascular effects of CA4 can negatively affect the efficacy of concurrently administered drugs, which necessitates high requirements for controlling the sequence and timing of drug release [163]. Wei et al. addressed this challenge by designing a polypeptide-based hydrogel capable of sequentially releasing CA4 and DOX. By utilizing the distinct hydrophobic properties of these drugs, their hydrogel facilitated the faster release of DOX and the slower release of CA4, minimizing potential negative interactions and optimizing therapeutic efficacy [63]. Conversely, Wang et al. reported a different release profile using an injectable PLGA-PEG-PLGA triblock polymer hydrogel. They found that CA4P, which is more hydrophilic than DOX, was released at a faster rate. Despite this reversed release sequence, combination therapy yielded superior antitumor effects compared with single-drug treatments, highlighting the versatility and potential of hydrogel-based co-delivery systems [164].
Regardless of the agent that should be released first, it is important to avoid drug interactions with anti-angiogenic and antineoplastic agents. Owing to the different mechanisms of anti-angiogenic and antineoplastic agents, the design and synthesis of hydrogels must be considered. In addition to enhancing the antitumor effect, avoiding high toxicity, preventing negative drug interactions, and the time-dependent induction of drug resistance by anti-angiogenic therapies [165,166] increase requirements for hydrogel-based DDSs. The preparation of smart hydrogels for the combination of antineoplastic and anti-angiogenic agents is urgently required.

2.3.3. DDSs of Immune Checkpoint Inhibitors

Cancer treatment includes various strategies beyond conventional chemotherapy, such as surgery, radiotherapy, immunotherapy, and gene therapy. Combination therapy leverages the complementary mechanisms of different approaches to enhance efficacy, mitigate resistance, and improve overall outcomes. In the case of advanced non-small-cell lung cancer, the combination of immune checkpoint inhibitors with chemotherapy in a first-line treatment prolongs the survival of patients in phase III trials [167]. Hydrogel-based DDSs can facilitate the combination of cytotoxic drugs for cancer treatment and other treatment approaches by providing controllable and localized release of different therapeutic agents. As an example of chemotherapy combined with immunotherapy, Chao et al. developed an alginate-based hydrogel to deliver the chemotherapeutic agent oxaliplatin (OXA) and immune adjuvant imiquimod (R837) as a “cocktail” of chemoimmunotherapeutic composites. The composite was administered intratumorally to form a hydrogel for localized treatment. In addition, an immune checkpoint blockade antibody (anti-PD-1) was included in the injection solution to enhance local immune modulation. The combination of OXA, R837, and anti-PD-1 within the hydrogel matrix showed superior antitumor efficiency compared with that of the free administration of these agents [168]. In another study, a nanogel was designed for combined chemoimmunotherapy by crosslinking carboxymethyl chitosan-derived polymetformin loaded with DOX. This system efficiently inhibited tumor growth, as polymetformin reshaped the TME and DOX killed the tumor cells more effectively [169].
Owing to the complexity of immune responses, designing hydrogels for combining chemotherapy and immunotherapy often requires meticulous consideration of both drug release profiles and treatment sequences. Animal studies typically involve multifaceted validation to assess the additive or synergistic effects of therapeutic strategies. Sun et al. used immunogenic cell death (ICD) to develop a hydrogel capable of administering immune adjuvants to tumors in a controlled manner during each cycle of chemotherapy and radiotherapy. The hydrogel is designed to release immunotherapeutic factors in response to low doses of oxaliplatin and X-rays. This approach results in a marked synergistic effect between chemotherapy and immunotherapy. By avoiding the simultaneous administration of chemotherapy and immunotherapy, this design effectively reduces systemic toxicity. More importantly, because ICD is induced by the release of chemotherapeutic agents or radiation therapy, it leads to the generation of a robust immune response, which has demonstrated a potent therapeutic effect in animal models. Hydrogel-based DDSs have been extensively reported as a means of minimizing the adverse effects associated with systemic immunotherapy. These systems facilitate the controlled release of therapeutic agents, thereby improving both safety and efficacy [170].
From a fundamental perspective, one of the most valuable attributes of hydrogel-based DDSs in combination therapies is their ability to control drug release based on the distinct solubility profiles of the incorporated drugs. Engineering hydrogels with appropriate and tunable affinities for specific drugs may be critical to achieve optimal DDS performance. Other types of hydrogels, such as hydrophobic hydrogels [171], tough adhesive hydrogels [172], and specialized formulations, are also under investigation. Owing to their high elasticity, water content, plasticity, and tunable gelation properties, hydrogels represent an indispensable DDS platform with immense potential for research and clinical applications. Furthermore, hydrogels can be used to carry NPs and serve as fillers for treatments. Magnetic NPs exhibit stronger thermal effects in hydrogels than conventional scaffold materials (Figure 3) [173], making hydrogels not only effective DDSs but also potentially transformative as post-surgical fillers with enhanced therapeutic outcomes. This multifunctionality broadens the scope of hydrogel applications in oncology, highlighting its versatility as both a drug carrier and therapeutic material.

3. Methods for In Vitro and In Vivo Evaluations

3.1. Method for In Vitro Antitumor Effect Evaluation

The evaluation of antitumor efficiency is critical for determining the applicability of drug-loaded hydrogels. Currently, four primary methods are commonly used to assess the cytotoxicity of drug-loaded hydrogels (Figure 4). The most widely used approach, reported in over 70% of the studies, involves treating cells with a medium containing hydrogel. Approximately 20% of the studies used a method in which cell suspensions were deposited directly into the hydrogel for co-culture. Among the 106 studies analyzed, six used hydrogel extracts for treatment and seven studies used Transwell systems to analyze cytotoxicity. Each of these methods have distinct advantages and limitations.
Adding hydrogels or their extracts directly to the culture medium is convenient, which is not different from methods that add hydrogel extracts with drugs into the culture medium and allow the simultaneous evaluation of hydrogel biocompatibility and cytotoxicity. However, these tests are typically conducted over a short period (48–72 h), which is considerably shorter than the extended duration of drug release observed in practical applications. Therefore, these methods may not accurately represent the actual antitumor efficacy of hydrogels. The method involving the deposition of cell suspensions on/into the hydrogels provides a closer approximation of the real-world applications of drug-loaded hydrogels. However, this approach inherently reflects the behavior of cells in direct contact with the hydrogel without considering the effects of remote DDSs. Moreover, drug diffusion from the hydrogel can significantly affect antitumor efficiency, necessitating the optimization of culture conditions, such as prolonging the incubation period, to better simulate in vivo drug release dynamics.
The Transwell assay focuses on assessing the cytotoxicity of drugs released from hydrogels. Although this provides valuable insights into drug delivery and release efficacy, it lacks the ability to directly evaluate hydrogel biocompatibility. The primary purpose of using a hydrogel-based DDS is to enhance the safety of drug administration and achieve localized drug concentrations; a comprehensive evaluation strategy is essential. It would be more reasonable to test the toxicity of the hydrogels and the optimal concentration of antitumor drugs.

3.2. Method for In Vivo Antitumor Effect Evaluation

The results of animal experiments for DDSs are more generalizable for clinical use than those of in vitro experiments. Although hydrogel-based DDSs have been extensively studied, some studies are still in the preclinical stage [174]. To reveal the real effects of hydrogel-based DDSs, in vivo experiments are indispensable before preclinical analysis.
Numerous studies have reported the antitumor effect of hydrogels in vivo, with some focusing on their efficacy in animal models of recurrence. Unlike free drug administration, the dosage regimen of hydrogels typically involves intratumoral, peritumoral, or subcutaneous injections. These methods require tailoring of the rheological properties of hydrogels to meet the requirements of injectability and sol–gel transition under in vivo conditions (Figure 5).
Peritumoral or intratumoral injections maximize local drug concentrations and antitumor efficacy, minimize drug loss through the circulatory system, and reduce accumulation in non-target organs, thereby lowering systemic toxicity compared to intravenous injections [134,144,175,176].
In contrast, in situ administration is more frequently used in brain tumor research [177,178,179]. For brain tumor treatments, the blood–brain barrier presents a considerable challenge, reducing effective drug concentrations at the target site and potentially causing off-target effects and systemic toxicity [180,181]. Compared with NP-based DDSs, in situ injection is more important for analyzing the function and availability of hydrogel-based DDSs in brain tumors, and subcutaneous tumor models cannot replicate the challenges posed by the unique environment of the brain.
Moreover, for specific therapies, such as immunotherapy, hydrogel carriers have demonstrated higher efficacy than that of free drug administration [182]. This may be attributed to the rapid clearance or degradation of immune-related agents, such as antibodies, during free administration, which reduces their therapeutic activity [183]. The application of hydrogels mitigates this issue by protecting these agents and enabling their sustained release, thereby enhancing their efficacy.
In addition to a single method of administration, in most studies, researchers have used more than one experimental method to evaluate the treatment potential of hydrogel-based DDSs. Jin et al. investigated the tumor ablation effects of injectable peptide hydrogels co-delivering DOX and Melttin. Their study compared intratumoral, peritumoral, and in situ injection methods and demonstrated significant antitumor effects across all approaches using hydrogel-based DDSs. In a footpad model used to study lymphatic metastasis, the in situ injection of hydrogels also achieved effective tumor suppression [184]. For the same hydrogel material, intratumoral and peritumoral injections showed similar results, likely due to the close proximity of the injection sites to the tumor [104]. In various studies, intratumoral or peritumoral administration consistently exhibited higher antitumor efficacy than that of free drug administration via intravenous injection [134,175,185]. Even when tumor growth inhibition was comparable, localized administration methods resulted in lower systemic toxicity, as reflected by the reduced body weight loss in treated animals [176]. These findings underscore the significant advantages of hydrogel-based DDSs in improving therapeutic outcomes while minimizing adverse effects.
Despite the gradual comprehensive analysis of hydrogel-based DDSs, challenges remain in translating these systems into practical applications. One issue is the discrepancy between the administration methods used in the experiments and those applicable to preclinical models. For disease treatment, intravenous, intramuscular, and subcutaneous injections remain the most common routes, whereas intratumoral injection is primarily used in antitumor immunotherapy at the preclinical stage [186,187,188]. Hydrogel injection would involve more problems than single immune agent injection. Furthermore, the feasibility of injection imposes additional constraints on the selection of hydrogel materials.
Apart from administration approaches, the variability in animal experiments in various studies poses substantial challenges. Differences in tumor volume at the start of experiments and research-dependent injection sites for peritumoral administration make it difficult to precisely evaluate treatment efficiency. Therefore, it is necessary to develop standardized and reliable animal models and experimental protocols to facilitate accurate comparison and reproducibility in hydrogel-based DDS research.
Finally, the lack of attention to sterilization methods in many studies is another issue. Only a few studies provided detailed descriptions of the sterilization process [189]. Although some hydrogels incorporate antibacterial drugs [190] or are composed of inherently antibacterial materials [191], most studies have not described sterilization methods, particularly for long-term animal experiments. Although Schiff base interactions have antibacterial effects [192], programmed sterilization remains essential for both in vivo studies and further applications.

4. Challenges and Developments of Hydrogel-Based DDSs

4.1. Preclinical Evaluation and Clinical Trials

No matter how innovative or imaginative a concept may be, it ultimately needs to be translated into practical applications. Only when favorable therapeutic efficacy is demonstrated can a hydrogel-based system proceed to clinical trials and eventual implementation. Currently, the major limitations hindering the clinical translation of hydrogel-based DDSs include suboptimal therapeutic efficiency, safety concerns, and insufficient objective preclinical evaluations.
For hydrogel-based DDSs, the most critical factor is drug delivery efficiency, which directly affects treatment effectiveness. Macroscopically, this efficiency is influenced by two key factors: the design of the hydrogel and the biological characteristics of the tumor. In terms of the hydrogel design, the mesh size of the hydrogel polymers, interactions between drugs and hydrogels, and drug concentration significantly affect the overall DDS performance. These factors have been extensively discussed in previous reviews [66,193,194]. However, pathophysiological characteristics of tumors also play a crucial role in drug delivery efficiency and cannot be ignored. The development of DDSs that adapt to the TME, along with combination therapy-based delivery strategies, is a major research topic in tumor treatment research. Specifically, with the application of novel materials and the development of innovative antitumor agents, the integration of bioactive compounds with hydrogel polymers offers expanded possibilities for tumor therapy.
Hydrogels exhibit excellent biocompatibility, supporting their broad clinical potential. However, the introduction of chemical crosslinking agents and the degradation of hydrogel polymers may inadvertently trigger undesirable biological responses, leading to safety concerns regarding their use [195,196]. For example, the incorporation of crosslinkers, such as glutaraldehyde, alters macrophage polarization during the foreign body response. This response is typically characterized by ECM deposition around the implanted biomaterial, which can impair device function and induce chronic inflammation [196,197]. Moreover, the degradation products of certain synthetic hydrogels, such as polyesters, poly (glycolic acid), and poly (L-lactic acid), may generate low-molecular-weight byproducts with cytotoxic or immunogenic properties [198,199]. These byproducts can induce oxidative stress, promote pro-inflammatory cytokine release, or interfere with local tissue homeostasis, raising concerns regarding long-term biocompatibility. However, most degradation analyses are conducted over short durations and the characterization of the degradation products is often lacking (Table 1, Table 2 and Table 3). Therefore, understanding and controlling the immunotoxicology profiles of both crosslinking agents and degradation products is essential for the safe translation of hydrogel-based DDSs into clinical use.
Despite these uncertainties, progress in clinical trials continues to demonstrate the strong potential of hydrogel-based DDSs for clinical applications. Currently, a hydrogel developed for the effective release of fibroblast growth factor-binding protein 1 (FGFBP1) inhibitors to treat pancreatic adenocarcinoma is in progress at CNR Nanotec Lecce [200]. Additionally, a thermosensitive hydrogel for 5-FU delivery for CRC treatment is in the recruitment phase [201]. The FDA has approved the chemotherapy gel, Jelmyto, for the treatment of low-grade upper tract urothelial cancer. Jelmyto is a thermally responsive hydrogel used to deliver the cytotoxic chemotherapeutic agent mitomycin [202]. Another promising candidate, UGN-102, a reverse thermal hydrogel with a mechanism similar to that of Jelmyto, has been developed for the primary nonsurgical treatment of low-grade, intermediate-risk, non-muscle-invasive bladder cancer. It has shown a high complete response rate in Phase III trials and may serve as a potential alternative to surgery [203].
Notably, both Jelmyto and UGN-102 offer minimally invasive approaches to chemotherapy [203]. Other non-drug delivery methods using hydrogels, such as the rectal hydrogel spacer system SpaceOAR [204], achieve a therapeutic effect by injection into the site of action. In line with the clinical requirements, most in vivo experiments have focused on the development of injectable hydrogels (Figure 5). However, their degradation behavior and immunogenicity have attracted less attention. More importantly, infection has also been reported as one of the complications associated with hydrogel injection, whereas sterilization methods are rarely described in hydrogel preparation protocols (Table 1, Table 2 and Table 3). In summary, although several hydrogel-based DDS candidates have entered clinical trials [205,206], insufficient preclinical data hinders the clinical translation of many promising systems. Completing in vivo studies with robust and sufficient data, particularly regarding the degradation safety and preparation methods, would be substantially beneficial for accelerating the entry of hydrogel-based DDSs into clinical use.

4.2. Future Directions and Conclusions

In this review, we summarized the strategies and methods commonly used to prepare hydrogel-based DDSs for antitumor therapies. Unlike traditional administration, hydrogel-based DDSs not only avoid blast drug release but are also applicable for combination therapy design. Hydrogel-based DDSs represent a promising frontier in antitumor therapy owing to their unique properties, such as high water content, biocompatibility, tunable physical and chemical characteristics, and the ability to encapsulate a variety of therapeutic agents. These features make hydrogels well-suited for addressing key challenges in cancer treatment, including enhancing local drug concentrations, reducing systemic toxicity, and enabling controlled drug release. Despite significant progress, several avenues remain unexplored, providing opportunities for future research.
One critical area of advancement is the integration of smart and responsive hydrogel technologies. Hydrogels that respond to external stimuli, such as temperature, pH, and magnetic or electric fields, are increasingly being investigated for their potential to achieve spatiotemporal drug release. Future studies should focus on developing multi-responsive hydrogels capable of adapting to the dynamic TME, thereby improving the precision of drug delivery and therapeutic outcomes.
Another promising direction is the application of hydrogels based on the clinical demand for tumor treatment, such as the combination delivery of different antitumor drugs, the combination delivery of antitumor and anti-angiogenic agents, and the combination of chemotherapy and other therapeutics. These hybrid systems can leverage the advantages of each component to improve the targeting specificity and achieve multilevel drug release profiles. Additionally, because of their good biocompatibility and designable mechanical properties using different polymers, hydrogel systems can further broaden their application as postoperative fillers for tissue regeneration.
Despite rapid developments, several challenges need to be addressed for the translation of hydrogel-based DDSs from the laboratory to clinical settings. Biodegradability and biocompatibility must be meticulously evaluated to ensure that the hydrogel degradation products are nontoxic and do not elicit adverse immune responses. Furthermore, the scalability and reproducibility of hydrogel synthesis must be optimized for mass production under good manufacturing practice standards.
The versatility and adaptability of hydrogels make them well-positioned to play a pivotal role in personalized medicine. As research continues to reveal the complexities of tumor biology, hydrogel-based systems are expected to evolve into more sophisticated and effective tools for cancer therapy, with promising improvements and reduced treatment burdens for patients worldwide.

Author Contributions

Conceptualization, G.C. and N.K.; methodology, G.C., N.K., T.Y., Y.Y. and T.Z.; software, T.Z. and N.K.; validation, G.C., N.K., T.Z. and L.C.; formal analysis, T.Z. and L.C.; investigation, T.Z. and L.C.; resources, G.C., N.K. and T.Y.; data curation, T.Z. and L.C.; writing—original draft preparation, T.Z. and G.C.; writing—review and editing, T.Z., L.C., T.Y., N.K., Y.Y. and G.C.; visualization, T.Z. and L.C.; supervision, G.C.; project administration, G.C. and N.K.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by JSPS KAKENHI Grant Number 19H04475, and 24K03289.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bruschi, M.L. Modification of drug release. In Strategies to Modify the Drug Release from Pharmaceutical Systems; Woodhead Publishing: Cambridge, UK, 2015; pp. 15–28. [Google Scholar]
  2. Li, R.; Lin, Z.; Zhang, Q.; Zhang, Y.; Liu, Y.; Lyu, Y.; Li, X.; Zhou, C.; Wu, G.; Ao, N.; et al. Injectable and In Situ-Formable Thiolated Chitosan-Coated Liposomal Hydrogels as Curcumin Carriers for Prevention of In Vivo Breast Cancer Recurrence. ACS Appl. Mater. Interfaces 2020, 12, 17936–17948. [Google Scholar] [CrossRef]
  3. Xu, L.; Cooper, R.C.; Wang, J.; Yeudall, W.A.; Yang, H. Synthesis and Application of Injectable Bioorthogonal Dendrimer Hydrogels for Local Drug Delivery. ACS Biomater. Sci. Eng. 2017, 3, 1641–1653. [Google Scholar] [CrossRef]
  4. Shukla, A.; Singh, A.P.; Maiti, P. Injectable hydrogels of newly designed brush biopolymers as sustained drug-delivery vehicle for melanoma treatment. Signal Transduct. Target. Ther. 2021, 6, 63. [Google Scholar] [CrossRef]
  5. Sheykhisarem, R.; Dehghani, H. In Vitro biocompatibility evaluations of pH-sensitive Bi(2)MoO(6)/NH(2)-GO conjugated polyethylene glycol for release of daunorubicin in cancer therapy. Colloids Surf. B Biointerfaces 2023, 221, 113006. [Google Scholar] [CrossRef]
  6. O’Brien, M.E.; Wigler, N.; Inbar, M.; Rosso, R.; Grischke, E.; Santoro, A.; Catane, R.; Kieback, D.G.; Tomczak, P.; Ackland, S.P.; et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann. Oncol. 2004, 15, 440–449. [Google Scholar] [CrossRef]
  7. Poon, R.T.; Borys, N. Lyso-thermosensitive liposomal doxorubicin: A novel approach to enhance efficacy of thermal ablation of liver cancer. Expert Opin. Pharmacother. 2009, 10, 333–343. [Google Scholar] [CrossRef]
  8. Olusanya, T.O.B.; Haj Ahmad, R.R.; Ibegbu, D.M.; Smith, J.R.; Elkordy, A.A. Liposomal Drug Delivery Systems and Anticancer Drugs. Molecules 2018, 23, 907. [Google Scholar] [CrossRef]
  9. Sun, R.; Wang, M.; Zeng, T.; Chen, H.; Yoshitomi, T.; Takeguchi, M.; Kawazoe, N.; Yang, Y.; Chen, G. Scaffolds functionalized with matrix metalloproteinase-responsive release of miRNA for synergistic magnetic hyperthermia and sensitizing chemotherapy of drug-tolerant breast cancer. Bioact. Mater. 2025, 44, 205–219. [Google Scholar] [CrossRef]
  10. Liu, P.; Chen, G.; Zhang, J. A Review of Liposomes as a Drug Delivery System: Current Status of Approved Products, Regulatory Environments, and Future Perspectives. Molecules 2022, 27, 1372. [Google Scholar] [CrossRef]
  11. Hsu, H.J.; Bugno, J.; Lee, S.R.; Hong, S. Dendrimer-based nanocarriers: A versatile platform for drug delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol 2017, 9, e1409. [Google Scholar] [CrossRef]
  12. Parra-Nieto, J.; Del Cid, M.A.G.; de Carcer, I.A.; Baeza, A. Inorganic Porous Nanoparticles for Drug Delivery in Antitumoral Therapy. Biotechnol. J. 2021, 16, e2000150. [Google Scholar] [CrossRef]
  13. Tsoi, K.M.; MacParland, S.A.; Ma, X.Z.; Spetzler, V.N.; Echeverri, J.; Ouyang, B.; Fadel, S.M.; Sykes, E.A.; Goldaracena, N.; Kaths, J.M.; et al. Mechanism of hard-nanomaterial clearance by the liver. Nat. Mater. 2016, 15, 1212–1221. [Google Scholar] [CrossRef]
  14. Wilhelm, S.; Tavares, A.J.; Dai, Q.; Ohta, S.; Audet, J.; Dvorak, H.F.; Chan, W.C.W. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 2016, 1, 16014. [Google Scholar] [CrossRef]
  15. Couvreur, P. Nanoparticles in drug delivery: Past, present and future. Adv. Drug Deliv. Rev. 2013, 65, 21–23. [Google Scholar] [CrossRef]
  16. Sharma, A.; Sharma, U.S. Liposomes in drug delivery: Progress and limitations. Int. J. Pharm. 1997, 154, 123–140. [Google Scholar] [CrossRef]
  17. Zhang, H.; Liu, K.; Gong, Y.; Zhu, W.; Zhu, J.; Pan, F.; Chao, Y.; Xiao, Z.; Liu, Y.; Wang, X.; et al. Vitamin C supramolecular hydrogel for enhanced cancer immunotherapy. Biomaterials 2022, 287, 121673. [Google Scholar] [CrossRef]
  18. Zhang, N.; Zhang, C.; Liu, J.; Fan, C.; Yin, J.; Wu, T. An oral hydrogel carrier for delivering resveratrol into intestine-specific target released with high encapsulation efficiency and loading capacity based on structure-selected alginate and pectin. Food Funct. 2022, 13, 12051–12066. [Google Scholar] [CrossRef]
  19. Li, R.; Lyu, Y.; Luo, S.; Wang, H.; Zheng, X.; Li, L.; Ao, N.; Zha, Z. Fabrication of a multi-level drug release platform with liposomes, chitooligosaccharides, phospholipids and injectable chitosan hydrogel to enhance anti-tumor effectiveness. Carbohydr. Polym. 2021, 269, 118322. [Google Scholar] [CrossRef]
  20. Thiele, J.; Ma, Y.; Bruekers, S.M.; Ma, S.; Huck, W.T. 25th anniversary article: Designer hydrogels for cell cultures: A materials selection guide. Adv. Mater. 2014, 26, 125–147. [Google Scholar] [CrossRef]
  21. Balbir, S.; Anjali, S.; Amit, K. Hydrogels: Synthesis, classification, properties and potential applications-a brief review. J. Polym. Environ. 2021, 29, 3827–3841. [Google Scholar] [CrossRef]
  22. Cao, H.; Duan, L.; Zhang, Y.; Cao, J.; Zhang, K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct. Target. Ther. 2021, 6, 426. [Google Scholar] [CrossRef]
  23. Varghese, S.A.; Rangappa, S.M.; Siengchin, S.; Parameswaranpillai, J. Natural polymers and the hydrogels prepared from them. In Hydrogels Based on Natural Polymers; Elsevier: Amsterdam, The Netherlands, 2020; pp. 17–47. [Google Scholar]
  24. Wang, Z.; Zhang, Y.; Zhang, J.; Huang, L.; Liu, J.; Li, Y.; Zhang, G.; Kundu, S.C.; Wang, L. Exploring natural silk protein sericin for regenerative medicine: An injectable, photoluminescent, cell-adhesive 3D hydrogel. Sci. Rep. 2014, 4, 7064. [Google Scholar] [CrossRef]
  25. Masters, K.S.; Shah, D.N.; Walker, G.; Leinwand, L.A.; Anseth, K.S. Designing scaffolds for valvular interstitial cells: Cell adhesion and function on naturally derived materials. J. Biomed. Mater. Res. Part. A 2004, 71, 172–180. [Google Scholar] [CrossRef]
  26. Zhang, Y.; Liu, J.; Huang, L.; Wang, Z.; Wang, L. Design and performance of a sericin-alginate interpenetrating network hydrogel for cell and drug delivery. Sci. Rep. 2015, 5, 12374. [Google Scholar] [CrossRef]
  27. Aravamudhan, A.; Ramos, D.M.; Nada, A.A.; Kumbar, S.G. Natural Polymers. In Natural and Synthetic Biomedical Polymers; Elsevier: Amsterdam, The Netherlands, 2014; pp. 67–89. [Google Scholar]
  28. Reddy, M.S.B.; Ponnamma, D.; Choudhary, R.; Sadasivuni, K.K. A Comparative Review of Natural and Synthetic Biopolymer Composite Scaffolds. Polymers 2021, 13, 1105. [Google Scholar] [CrossRef] [PubMed]
  29. Sun, Z.; Song, C.; Wang, C.; Hu, Y.; Wu, J. Hydrogel-Based Controlled Drug Delivery for Cancer Treatment: A Review. Mol. Pharm. 2020, 17, 373–391. [Google Scholar] [CrossRef]
  30. Jahanban-Esfahlan, R.; Derakhshankhah, H.; Haghshenas, B.; Massoumi, B.; Abbasian, M.; Jaymand, M. A bio-inspired magnetic natural hydrogel containing gelatin and alginate as a drug delivery system for cancer chemotherapy. Int. J. Biol. Macromol. 2020, 156, 438–445. [Google Scholar] [CrossRef]
  31. Zeng, Y.; Zhang, C.; Du, D.; Li, Y.; Sun, L.; Han, Y.; He, X.; Dai, J.; Shi, L. Metal-organic framework-based hydrogel with structurally dynamic properties as a stimuli-responsive localized drug delivery system for cancer therapy. Acta Biomater. 2022, 145, 43–51. [Google Scholar] [CrossRef]
  32. Sheng, Y.; Gao, J.; Yin, Z.-Z.; Kang, J.; Kong, Y. Dual-drug delivery system based on the hydrogels of alginate and sodium carboxymethyl cellulose for colorectal cancer treatment. Carbohydr. Polym. 2021, 269, 118325. [Google Scholar] [CrossRef] [PubMed]
  33. Foroutan, R.; Mohammadzadeh, A.; Javanbakht, S.; Mohammadi, R.; Ghorbani, M. Alginate/magnetic hydroxyapatite bio-nanocomposite hydrogel bead as a pH-responsive oral drug carrier for potential colon cancer therapy. Results Chem. 2025, 15, 102177. [Google Scholar] [CrossRef]
  34. Yang, Z.; Liu, J.; Lu, Y. Doxorubicin and CD-CUR inclusion complex co-loaded in thermosensitive hydrogel PLGA-PEG-PLGA localized administration for osteosarcoma. Int. J. Oncol. 2020, 57, 433–444. [Google Scholar] [CrossRef] [PubMed]
  35. Qu, Y.; Chu, B.; Wei, X.; Lei, M.; Hu, D.; Zha, R.; Zhong, L.; Wang, M.; Wang, F.; Qian, Z. Redox/pH dual-stimuli responsive camptothecin prodrug nanogels for “on-demand” drug delivery. J. Control. Release 2019, 296, 93–106. [Google Scholar] [CrossRef]
  36. Lin, J.-T.; Ye, Q.-B.; Yang, Q.-J.; Wang, G.-H. Hierarchical bioresponsive nanocarriers for codelivery of curcumin and doxorubicin. Colloids Surf. B Biointerfaces 2019, 180, 93–101. [Google Scholar] [CrossRef]
  37. Yaghoubi, A.; Ramazani, A.; Sillanpaa, M.; Ghasemzadeh, H.; Mohammadi, E. Biocompatible porous PAM/CNT nanocomposite hydrogel films for sustained drug delivery and cancer therapy. Sci. Rep. 2025, 15, 22387. [Google Scholar] [CrossRef]
  38. Li, X.; Li, H.; Zhang, C.; Pich, A.; Xing, L.; Shi, X. Intelligent nanogels with self-adaptive responsiveness for improved tumor drug delivery and augmented chemotherapy. Bioact. Mater. 2021, 6, 3473–3484. [Google Scholar] [CrossRef]
  39. Qian, H.; Qian, K.; Cai, J.; Yang, Y.; Zhu, L.; Liu, B. Therapy for Gastric Cancer with Peritoneal Metastasis Using Injectable Albumin Hydrogel Hybridized with Paclitaxel-Loaded Red Blood Cell Membrane Nanoparticles. ACS Biomater. Sci. Eng. 2019, 5, 1100–1112. [Google Scholar] [CrossRef]
  40. Nazir, S.; Umar Aslam Khan, M.; Shamsan Al-Arjan, W.; Izwan Abd Razak, S.; Javed, A.; Rafiq Abdul Kadir, M. Nanocomposite hydrogels for melanoma skin cancer care and treatment: In-vitro drug delivery, drug release kinetics and anti-cancer activities. Arab. J. Chem. 2021, 14, 103120. [Google Scholar] [CrossRef]
  41. Ullah, K.; Sohail, M.; Murtaza, G.; Khan, S.A. Natural and synthetic materials based CMCh/PVA hydrogels for oxaliplatin delivery: Fabrication, characterization, In-Vitro and In-Vivo safety profiling. Int. J. Biol. Macromol. 2019, 122, 538–548. [Google Scholar] [CrossRef]
  42. Halder, S.; Das, T.; Kushwaha, R.; Misra, A.K.; Jana, K.; Das, D. Targeted and precise drug delivery using a glutathione-responsive ultra-short peptide-based injectable hydrogel as a breast cancer cure. Mater. Horiz. 2025, 12, 987–1001. [Google Scholar] [CrossRef]
  43. Sathi Devi, L.; Gigliobianco, M.R.; Gabrielli, S.; Agas, D.; Sabbieti, M.G.; Morelli, M.B.; Amantini, C.; Casadidio, C.; Di Martino, P.; Censi, R. Localized Cancer Treatment Using Thiol–Ene Hydrogels for Dual Drug Delivery. Biomacromolecules 2025, 26, 3234–3254. [Google Scholar] [CrossRef]
  44. Ahmadi, S.; Olad, A.; Fathi, M.; Molavi, O. An injectable chitosan based dual thermo/pH-responsive fast gelling hydrogel loaded by methotrexate/curcumin as local drug delivery system of breast cancer. J. Drug Deliv. Sci. Technol. 2025, 104, 106540. [Google Scholar] [CrossRef]
  45. Sionkowska, A. Current research on the blends of natural and synthetic polymers as new biomaterials: Review. Progress. Polym. Sci. 2011, 36, 1254–1276. [Google Scholar] [CrossRef]
  46. Fu, X.; Hosta-Rigau, L.; Chandrawati, R.; Cui, J. Multi-Stimuli-Responsive Polymer Particles, Films, and Hydrogels for Drug Delivery. Chemistry 2018, 4, 2084–2107. [Google Scholar] [CrossRef]
  47. Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef]
  48. Xiao, Y.; Yu, D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol. Ther. 2021, 221, 107753. [Google Scholar] [CrossRef]
  49. Neri, D.; Supuran, C.T. Interfering with pH regulation in tumours as a therapeutic strategy. Nat. Rev. Drug Discov. 2011, 10, 767–777. [Google Scholar] [CrossRef]
  50. Gaohua, L.; Miao, X.; Dou, L. Crosstalk of physiological pH and chemical pKa under the umbrella of physiologically based pharmacokinetic modeling of drug absorption, distribution, metabolism, excretion, and toxicity. Expert Opin. Drug Metab. Toxicol. 2021, 17, 1103–1124. [Google Scholar] [CrossRef]
  51. Qu, J.; Zhao, X.; Ma, P.X.; Guo, B. pH-responsive self-healing injectable hydrogel based on N-carboxyethyl chitosan for hepatocellular carcinoma therapy. Acta Biomater. 2017, 58, 168–180. [Google Scholar] [CrossRef]
  52. Yan, Q.; Liu, L.; Wang, T.; Wang, H. A pH-responsive hydrogel system based on cellulose and dopamine with controlled hydrophobic drug delivery ability and long-term bacteriostatic property. Colloid. Polym. Sci. 2019, 297, 705–717. [Google Scholar] [CrossRef]
  53. Arjama, M.; Mehnath, S.; Rajan, M.; Jeyaraj, M. Sericin/RBA embedded gellan gum based smart nanosystem for pH responsive drug delivery. Int. J. Biol. Macromol. 2018, 120, 1561–1571. [Google Scholar] [CrossRef]
  54. Solomevich, S.O.; Bychkovsky, P.M.; Yurkshtovich, T.L.; Golub, N.V.; Mirchuk, P.Y.; Revtovich, M.Y.; Shmak, A.I. Biodegradable pH-sensitive prospidine-loaded dextran phosphate based hydrogels for local tumor therapy. Carbohydr. Polym. 2019, 226, 115308. [Google Scholar] [CrossRef]
  55. Liu, Z.; Xu, G.; Wang, C.; Li, C.; Yao, P. Shear-responsive injectable supramolecular hydrogel releasing doxorubicin loaded micelles with pH-sensitivity for local tumor chemotherapy. Int. J. Pharm. 2017, 530, 53–62. [Google Scholar] [CrossRef]
  56. Turk, S.; Altinsoy, I.; Efe, G.C.; Ipek, M.; Ozacar, M.; Bindal, C. A novel multifunctional NCQDs-based injectable self-crosslinking and in situ forming hydrogel as an innovative stimuli responsive smart drug delivery system for cancer therapy. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 121, 111829. [Google Scholar] [CrossRef]
  57. Lee, A.L.Z.; Voo, Z.X.; Chin, W.; Ono, R.J.; Yang, C.; Gao, S.; Hedrick, J.L.; Yang, Y.Y. Injectable Coacervate Hydrogel for Delivery of Anticancer Drug-Loaded Nanoparticles in vivo. ACS Appl. Mater. Interfaces 2018, 10, 13274–13282. [Google Scholar] [CrossRef]
  58. Su, H.; Zhang, W.; Wu, Y.; Han, X.; Liu, G.; Jia, Q.; Shan, S. Schiff base-containing dextran nanogel as pH-sensitive drug delivery system of doxorubicin: Synthesis and characterization. J. Biomater. Appl. 2018, 33, 170–181. [Google Scholar] [CrossRef]
  59. Hu, L.; Zhang, P.; Wang, X.; Cheng, X.; Qin, J.; Tang, R. pH-sensitive carboxymethyl chitosan hydrogels via acid-labile ortho ester linkage for potential biomedical applications. Carbohydr. Polym. 2017, 178, 166–179. [Google Scholar] [CrossRef]
  60. Liu, Y.; Ran, Y.; Ge, Y.; Raza, F.; Li, S.; Zafar, H.; Wu, Y.; Paiva-Santos, A.C.; Yu, C.; Sun, M.; et al. pH-Sensitive Peptide Hydrogels as a Combination Drug Delivery System for Cancer Treatment. Pharmaceutics 2022, 14, 652. [Google Scholar] [CrossRef]
  61. Liu, M.; Song, X.; Wen, Y.; Zhu, J.L.; Li, J. Injectable Thermoresponsive Hydrogel Formed by Alginate-g-Poly(N-isopropylacrylamide) That Releases Doxorubicin-Encapsulated Micelles as a Smart Drug Delivery System. ACS Appl. Mater. Interfaces 2017, 9, 35673–35682. [Google Scholar] [CrossRef]
  62. Luo, Y.; Li, J.; Hu, Y.; Gao, F.; Pak-Heng Leung, G.; Geng, F.; Fu, C.; Zhang, J. Injectable thermo-responsive nano-hydrogel loading triptolide for the anti-breast cancer enhancement via localized treatment based on “two strikes” effects. Acta Pharm. Sin. B 2020, 10, 2227–2245. [Google Scholar] [CrossRef]
  63. Wei, L.; Chen, J.; Zhao, S.; Ding, J.; Chen, X. Thermo-sensitive polypeptide hydrogel for locally sequential delivery of two-pronged antitumor drugs. Acta Biomater. 2017, 58, 44–53. [Google Scholar] [CrossRef]
  64. Darge, H.F.; Andrgie, A.T.; Hanurry, E.Y.; Birhan, Y.S.; Mekonnen, T.W.; Chou, H.Y.; Hsu, W.H.; Lai, J.Y.; Lin, S.Y.; Tsai, H.C. Localized controlled release of bevacizumab and doxorubicin by thermo-sensitive hydrogel for normalization of tumor vasculature and to enhance the efficacy of chemotherapy. Int. J. Pharm. 2019, 572, 118799. [Google Scholar] [CrossRef]
  65. Hu, Y.; Darcos, V.; Monge, S.; Li, S.; Zhou, Y.; Su, F. Thermo-responsive release of curcumin from micelles prepared by self-assembly of amphiphilic P(NIPAAm-co-DMAAm)-b-PLLA-b-P(NIPAAm-co-DMAAm) triblock copolymers. Int. J. Pharm. 2014, 476, 31–40. [Google Scholar] [CrossRef]
  66. Li, J.; Mooney, D.J. Designing hydrogels for controlled drug delivery. Nat. Rev. Mater. 2016, 1, 16071. [Google Scholar] [CrossRef]
  67. Yang, L.; Fan, X.; Zhang, J.; Ju, J. Preparation and Characterization of Thermoresponsive Poly(N-Isopropylacrylamide) for Cell Culture Applications. Polymers 2020, 12, 389. [Google Scholar] [CrossRef]
  68. Li, W.; Huang, L.; Ying, X.; Jian, Y.; Hong, Y.; Hu, F.; Du, Y. Antitumor Drug Delivery Modulated by A Polymeric Micelle with an Upper Critical Solution Temperature. Angew. Chem. Int. Ed. 2015, 54, 3126–3131. [Google Scholar] [CrossRef]
  69. Le, M.; Huang, W.; Chen, K.-F.; Lin, C.; Cai, L.; Zhang, H.; Jia, Y.-G. Upper critical solution temperature polymeric drug carriers. Chem. Eng. J. 2022, 432, 134354. [Google Scholar] [CrossRef]
  70. Park, T.G.; Hoffman, A.S. Deswelling characteristics of poly(N-isopropylacrylamide) hydrogel. J. Appl. Polym. Sci. 1994, 52, 85–89. [Google Scholar] [CrossRef]
  71. Wang, L.; Li, B.; Xu, F.; Xu, Z.; Wei, D.; Feng, Y.; Wang, Y.; Jia, D.; Zhou, Y. UV-crosslinkable and thermo-responsive chitosan hybrid hydrogel for NIR-triggered localized on-demand drug delivery. Carbohydr. Polym. 2017, 174, 904–914. [Google Scholar] [CrossRef]
  72. Burger, K.N.; Staffhorst, R.W.; de Vijlder, H.C.; Velinova, M.J.; Bomans, P.H.; Frederik, P.M.; de Kruijff, B. Nanocapsules: Lipid-coated aggregates of cisplatin with high cytotoxicity. Nat. Med. 2002, 8, 81–84. [Google Scholar] [CrossRef]
  73. Zhang, P.; Ling, G.; Sun, J.; Zhang, T.; Yuan, Y.; Sun, Y.; Wang, Z.; He, Z. Multifunctional nanoassemblies for vincristine sulfate delivery to overcome multidrug resistance by escaping P-glycoprotein mediated efflux. Biomaterials 2011, 32, 5524–5533. [Google Scholar] [CrossRef] [PubMed]
  74. Gupta, S.; Singh, M.; Reddy, A.; Yavvari, P.S.; Srivastava, A.; Bajaj, A. Interactions governing the entrapment of anticancer drugs by low-molecular-weight hydrogelator for drug delivery applications. RSC Adv. 2016, 6, 19751–19757. [Google Scholar] [CrossRef]
  75. Fahr, A.; Liu, X. Drug delivery strategies for poorly water-soluble drugs. Expert Opin. Drug Deliv. 2007, 4, 403–416. [Google Scholar] [CrossRef]
  76. Xu, X.; Chen, X.; Wang, Z.; Jing, X. Ultrafine PEG-PLA fibers loaded with both paclitaxel and doxorubicin hydrochloride and their in vitro cytotoxicity. Eur. J. Pharm. Biopharm. 2009, 72, 18–25. [Google Scholar] [CrossRef]
  77. Allen, T.M.; Cullis, P.R. Drug delivery systems: Entering the mainstream. Science 2004, 303, 1818–1822. [Google Scholar] [CrossRef]
  78. Shah, B.; Dong, X. Design and Evaluation of Two-Step Biorelevant Dissolution Methods for Docetaxel Oral Formulations. AAPS PharmSciTech 2022, 23, 113. [Google Scholar] [CrossRef]
  79. Rezazadeh, M.; Akbari, V.; Amuaghae, E.; Emami, J. Preparation and characterization of an injectable thermosensitive hydrogel for simultaneous delivery of paclitaxel and doxorubicin. Res. Pharm. Sci. 2018, 13, 181–191. [Google Scholar] [CrossRef]
  80. Akbari, V.; Hejazi, E.; Minaiyan, M.; Emami, J.; Lavasanifar, A.; Rezazadeh, M. An injectable thermosensitive hydrogel/nanomicelles composite for local chemo-immunotherapy in mouse model of melanoma. J. Biomater. Appl. 2022, 551–562. [Google Scholar] [CrossRef]
  81. Yu, A.C.; Stapleton, L.M.; Mann, J.L.; Appel, E.A. Self-assembled biomaterials using host-guest interactions. In Self-Assembling Biomaterials; Woodhead Publishing: Cambridge, UK, 2018; pp. 205–231. [Google Scholar]
  82. Angelova, S.; Nikolova, V.; Pereva, S.; Spassov, T.; Dudev, T. alpha-Cyclodextrin: How Effectively Can Its Hydrophobic Cavity Be Hydrated? J. Phys. Chem. B 2017, 121, 9260–9267. [Google Scholar] [CrossRef]
  83. Miranda, J.C.D.; Martins, T.E.A.; Veiga, F.; Ferraz, H.G. Cyclodextrins and ternary complexes: Technology to improve solubility of poorly soluble drugs. Braz. J. Pharm. Sci. 2011, 47, 665–681. [Google Scholar] [CrossRef]
  84. Jalalvandi, E.; Cabral, J.; Hanton, L.R.; Moratti, S.C. Cyclodextrin-polyhydrazine degradable gels for hydrophobic drug delivery. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 69, 144–153. [Google Scholar] [CrossRef]
  85. Nieto, C.; Vega, M.A.; Rodríguez, V.; Esteban, P.P.; Martín del Valle, E.M. Biodegradable gellan gum hydrogels loaded with paclitaxel for HER2+ breast cancer local therapy. Carbohydr. Polym. 2022, 294, 119732. [Google Scholar] [CrossRef]
  86. Ma, M.; Shang, W.; Jia, R.; Chen, R.; Zhao, M.; Wang, C.; Tian, M.; Yang, S.; Hao, A. A novel folic acid hydrogel loading β-cyclodextrin/camptothecin inclusion complex with effective antitumor activity. J. Incl. Phenom. Macrocycl. Chem. 2019, 96, 169–179. [Google Scholar] [CrossRef]
  87. Jensen, G.; Hwang, J.; Schluep, T. Antitumor activity of IT-101, a cyclodextrin-containing polymer-camptothecin nanoparticle, in combination with various anticancer agents in human ovarian cancer xenografts. Cancer Res. 2008, 68, 767. [Google Scholar]
  88. Cheng, J.; Khin, K.T.; Davis, M.E. Antitumor activity of beta-cyclodextrin polymer-camptothecin conjugates. Mol. Pharm. 2004, 1, 183–193. [Google Scholar] [CrossRef]
  89. Davis, M.E. Design and development of IT-101, a cyclodextrin-containing polymer conjugate of camptothecin. Adv. Drug Deliv. Rev. 2009, 61, 1189–1192. [Google Scholar] [CrossRef]
  90. Schluep, T.; Hwang, J.; Cheng, J.; Heidel, J.D.; Bartlett, D.W.; Hollister, B.; Davis, M.E. Preclinical efficacy of the camptothecin-polymer conjugate IT-101 in multiple cancer models. Clin. Cancer Res. 2006, 12, 1606–1614. [Google Scholar] [CrossRef]
  91. Schluep, T.; Cheng, J.; Khin, K.T.; Davis, M.E. Pharmacokinetics and biodistribution of the camptothecin-polymer conjugate IT-101 in rats and tumor-bearing mice. Cancer Chemother. Pharmacol. 2006, 57, 654–662. [Google Scholar] [CrossRef]
  92. Zhang, W.; Zhou, X.; Liu, T.; Ma, D.; Xue, W. Supramolecular hydrogels co-loaded with camptothecin and doxorubicin for sustainedly synergistic tumor therapy. J. Mater. Chem. B 2015, 3, 2127–2136. [Google Scholar] [CrossRef] [PubMed]
  93. Song, X.; Zhang, Z.; Zhu, J.; Wen, Y.; Zhao, F.; Lei, L.; Phan-Thien, N.; Khoo, B.C.; Li, J. Thermoresponsive Hydrogel Induced by Dual Supramolecular Assemblies and Its Controlled Release Property for Enhanced Anticancer Drug Delivery. Biomacromolecules 2020, 21, 1516–1527. [Google Scholar] [CrossRef]
  94. Fu, C.; Lin, X.; Wang, J.; Zheng, X.; Li, X.; Lin, Z.; Lin, G. Injectable micellar supramolecular hydrogel for delivery of hydrophobic anticancer drugs. J. Mater. Sci. Mater. Med. 2016, 27, 73. [Google Scholar] [CrossRef]
  95. Cheng, H.; Fan, X.; Wang, X.; Ye, E.; Loh, X.J.; Li, Z.; Wu, Y.L. Hierarchically Self-Assembled Supramolecular Host-Guest Delivery System for Drug Resistant Cancer Therapy. Biomacromolecules 2018, 19, 1926–1938. [Google Scholar] [CrossRef]
  96. D’Arrigo, G.; Navarro, G.; Di Meo, C.; Matricardi, P.; Torchilin, V. Gellan gum nanohydrogel containing anti-inflammatory and anti-cancer drugs: A multi-drug delivery system for a combination therapy in cancer treatment. Eur. J. Pharm. Biopharm. 2014, 87, 208–216. [Google Scholar] [CrossRef]
  97. Zhu, Y.; Wang, L.; Li, Y.; Huang, Z.; Luo, S.; He, Y.; Han, H.; Raza, F.; Wu, J.; Ge, L. Injectable pH and redox dual responsive hydrogels based on self-assembled peptides for anti-tumor drug delivery. Biomater. Sci. 2020, 8, 5415–5426. [Google Scholar] [CrossRef]
  98. Leng, Q.; Li, Y.; Zhou, P.; Xiong, K.; Lu, Y.; Cui, Y.; Wang, B.; Wu, Z.; Zhao, L.; Fu, S. Injectable hydrogel loaded with paclitaxel and epirubicin to prevent postoperative recurrence and metastasis of breast cancer. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 129, 112390. [Google Scholar] [CrossRef]
  99. Wu, Z.; Zou, X.; Yang, L.; Lin, S.; Fan, J.; Yang, B.; Sun, X.; Wan, Q.; Chen, Y.; Fu, S. Thermosensitive hydrogel used in dual drug delivery system with paclitaxel-loaded micelles for in situ treatment of lung cancer. Colloids Surf. B Biointerfaces 2014, 122, 90–98. [Google Scholar] [CrossRef] [PubMed]
  100. Altunbas, A.; Lee, S.J.; Rajasekaran, S.A.; Schneider, J.P.; Pochan, D.J. Encapsulation of curcumin in self-assembling peptide hydrogels as injectable drug delivery vehicles. Biomaterials 2011, 32, 5906–5914. [Google Scholar] [CrossRef] [PubMed]
  101. Sheng, S.; Wei, C.; Ma, T.; Zhang, Y.; Zhu, D.; Dong, X.; Lv, F. Multiplex fluorescence imaging-guided programmed delivery of doxorubicin and curcumin from a nanoparticles/hydrogel system for synergistic chemotherapy. J. Polym. Sci. 2021, 60, 1557–1570. [Google Scholar] [CrossRef]
  102. Zhang, C.; Wang, X.; Xiao, M.; Ma, J.; Qu, Y.; Zou, L.; Zhang, J. Nano-in-micro alginate/chitosan hydrogel via electrospray technology for orally curcumin delivery to effectively alleviate ulcerative colitis. Mater. Des. 2022, 221, 110894. [Google Scholar] [CrossRef]
  103. Zhu, W.; Li, Y.; Liu, L.; Chen, Y.; Xi, F. Supramolecular hydrogels as a universal scaffold for stepwise delivering Dox and Dox/cisplatin loaded block copolymer micelles. Int. J. Pharm. 2012, 437, 11–19. [Google Scholar] [CrossRef]
  104. Sheu, M.T.; Jhan, H.J.; Su, C.Y.; Chen, L.C.; Chang, C.E.; Liu, D.Z.; Ho, H.O. Codelivery of doxorubicin-containing thermosensitive hydrogels incorporated with docetaxel-loaded mixed micelles enhances local cancer therapy. Colloids Surf. B Biointerfaces 2016, 143, 260–270. [Google Scholar] [CrossRef]
  105. Suri, A.; Campos, R.; Rackus, D.G.; Spiller, N.J.S.; Richardson, C.; Pålsson, L.-O.; Kataky, R. Liposome-doped hydrogel for implantable tissue. Soft Matter 2011, 7, 7071–7077. [Google Scholar] [CrossRef]
  106. Mauri, E.; Negri, A.; Rebellato, E.; Masi, M.; Perale, G.; Rossi, F. Hydrogel-Nanoparticles Composite System for Controlled Drug Delivery. Gels 2018, 4, 74. [Google Scholar] [CrossRef]
  107. Zeng, T.; Xu, M.; Zhang, W.; Gu, X.; Zhao, F.; Liu, X.; Zhang, X. Autophagy inhibition and microRNA-199a-5p upregulation in paclitaxel-resistant A549/T lung cancer cells. Oncol. Rep. 2021, 46, 149. [Google Scholar] [CrossRef]
  108. Zeng, T.; Chen, H.; Yoshitomi, T.; Kawazoe, N.; Yang, Y.; Chen, G. Effect of Hydrogel Stiffness on Chemoresistance of Breast Cancer Cells in 3D Culture. Gels 2024, 10, 202. [Google Scholar] [CrossRef] [PubMed]
  109. Zeng, T.; Lu, C.; Wang, M.; Chen, H.; Yoshitomi, T.; Kawazoe, N.; Yang, Y.; Chen, G. The effect of microenvironmental viscosity on the emergence of colon cancer cell resistance to doxorubicin. J. Mater. Chem. B 2025, 13, 2180–2191. [Google Scholar] [CrossRef] [PubMed]
  110. Mohelnikova-Duchonova, B.; Melichar, B.; Soucek, P. FOLFOX/FOLFIRI pharmacogenetics: The call for a personalized approach in colorectal cancer therapy. World J. Gastroenterol. 2014, 20, 10316–10330. [Google Scholar] [CrossRef] [PubMed]
  111. Linschoten, M.; Kamphuis, J.A.M.; van Rhenen, A.; Bosman, L.P.; Cramer, M.J.; Doevendans, P.A.; Teske, A.J.; Asselbergs, F.W. Cardiovascular adverse events in patients with non-Hodgkin lymphoma treated with first-line cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP with rituximab (R-CHOP): A systematic review and meta-analysis. Lancet Haematol. 2020, 7, e295–e308. [Google Scholar] [CrossRef]
  112. Chua, W.; Goldstein, D.; Lee, C.K.; Dhillon, H.; Michael, M.; Mitchell, P.; Clarke, S.J.; Iacopetta, B. Molecular markers of response and toxicity to FOLFOX chemotherapy in metastatic colorectal cancer. Br. J. Cancer 2009, 101, 998–1004. [Google Scholar] [CrossRef]
  113. Herold, M.; Hieke, K. Costs of toxicity during chemotherapy with CHOP, COP/CVP, and fludarabine. Eur. J. Health Econ. 2002, 3, 166–172. [Google Scholar] [CrossRef]
  114. Zhang, Z.; Ding, C.; Sun, T.; Wang, L.; Chen, C. Tumor Therapy Strategies Based on Microenvironment-Specific Responsive Nanomaterials. Adv. Healthc. Mater. 2023, 12, 2300153. [Google Scholar] [CrossRef]
  115. Zhou, S.; Zheng, J.; Zhai, W.; Chen, Y. Spatio-temporal heterogeneity in cancer evolution and tumor microenvironment of renal cell carcinoma with tumor thrombus. Cancer Lett. 2023, 572, 216350. [Google Scholar] [CrossRef]
  116. Proietto, M.; Crippa, M.; Damiani, C.; Pasquale, V.; Sacco, E.; Vanoni, M.; Gilardi, M. Tumor heterogeneity: Preclinical models, emerging technologies, and future applications. Front. Oncol. 2023, 13, 1164535. [Google Scholar] [CrossRef] [PubMed]
  117. Reitan, N.K.; Thuen, M.; Goa, P.L.E.; de Lange Davies, C. Characterization of tumor microvascular structure and permeability: Comparison between magnetic resonance imaging and intravital confocal imaging. J. Biomed. Opt. 2010, 15, 036004. [Google Scholar] [CrossRef] [PubMed]
  118. Yuan, Y. Spatial Heterogeneity in the Tumor Microenvironment. Cold Spring Harb. Perspect. Med. 2016, 6, a026583. [Google Scholar] [CrossRef]
  119. Zhou, K.I.; Peterson, B.; Serritella, A.; Thomas, J.; Reizine, N.; Moya, S.; Tan, C.; Wang, Y.; Catenacci, D.V.T. Spatial and Temporal Heterogeneity of PD-L1 Expression and Tumor Mutational Burden in Gastroesophageal Adenocarcinoma at Baseline Diagnosis and after Chemotherapy. Clin. Cancer Res. 2020, 26, 6453–6463. [Google Scholar] [CrossRef]
  120. Hida, K.; Maishi, N.; Sakurai, Y.; Hida, Y.; Harashima, H. Heterogeneity of tumor endothelial cells and drug delivery. Adv. Drug Deliv. Rev. 2016, 99, 140–147. [Google Scholar] [CrossRef]
  121. Jin, Z.; Zhou, Q.; Cheng, J.-N.; Jia, Q.; Zhu, B. Heterogeneity of the tumor immune microenvironment and clinical interventions. Front. Med. 2023, 17, 617–648. [Google Scholar] [CrossRef]
  122. Wang, B.; Song, B.; Li, Y.; Zhao, Q.; Tan, B. Mapping spatial heterogeneity in gastric cancer microenvironment. Biomed. Pharmacother. 2024, 172, 116317. [Google Scholar] [CrossRef] [PubMed]
  123. Lin, M.; Liu, X.; Li, J.; Zou, H.; Wang, J.; Yan, Z.; Liu, Y.; Lyu, Y.; Feng, N. Hybrid NIR-responsive liposome/hydrogel platform mediating chemo-photothermal therapy of retinoblastoma enhanced by quercetin as an adjuvant. Theranostics 2025, 15, 3995–4015. [Google Scholar] [CrossRef]
  124. Arvejeh, P.M.; Chermahini, F.A.; Marincola, F.; Taheri, F.; Mirzaei, S.A.; Alizadeh, A.; Deris, F.; Jafari, R.; Amiri, N.; Soltani, A.; et al. A novel approach for the co-delivery of 5-fluorouracil and everolimus for breast cancer combination therapy: Stimuli-responsive chitosan hydrogel embedded with mesoporous silica nanoparticles. J. Transl. Med. 2025, 23, 382. [Google Scholar] [CrossRef]
  125. Feng, C.; Xiao, S.; Mu, M.; Wang, X.; Yu, W.; Pan, S.; Li, H.; Fan, R.; Han, B.; Guo, G. In situ TLR7 activation with biocompatible injectable hydrogel for synergistic chemo-immunotherapy. J. Mater. Sci. Technol. 2026, 241, 93–106. [Google Scholar] [CrossRef]
  126. Gewirtz, D.A. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem. Pharmacol. 1999, 57, 727–741. [Google Scholar] [CrossRef]
  127. Bielack, S.S.; Erttmann, R.; Winkler, K.; Landbeck, G. Doxorubicin: Effect of different schedules on toxicity and anti-tumor efficacy. Eur. J. Cancer Clin. Oncol. 1989, 25, 873–882. [Google Scholar] [CrossRef]
  128. Dong, X.; Sun, Z.; Wang, X.; Zhu, D.; Liu, L.; Leng, X. Simultaneous monitoring of the drug release and antitumor effect of a novel drug delivery system-MWCNTs/DOX/TC. Drug Deliv. 2017, 24, 143–151. [Google Scholar] [CrossRef]
  129. Ogawara, K.; Un, K.; Tanaka, K.; Higaki, K.; Kimura, T. In vivo anti-tumor effect of PEG liposomal doxorubicin (DOX) in DOX-resistant tumor-bearing mice: Involvement of cytotoxic effect on vascular endothelial cells. J. Control. Release 2009, 133, 4–10. [Google Scholar] [CrossRef] [PubMed]
  130. Sun, Y.; Zou, W.; Bian, S.; Huang, Y.; Tan, Y.; Liang, J.; Fan, Y.; Zhang, X. Bioreducible PAA-g-PEG graft micelles with high doxorubicin loading for targeted antitumor effect against mouse breast carcinoma. Biomaterials 2013, 34, 6818–6828. [Google Scholar] [CrossRef]
  131. Rihova, B.; Srogl, J.; Jelinkova, M.; Hovorka, O.; Buresova, M.; Subr, V.; Ulbrich, K. HPMA-based biodegradable hydrogels containing different forms of doxorubicin. Antitumor effects and biocompatibility. Ann. N. Y. Acad. Sci. 1997, 831, 57–71. [Google Scholar] [CrossRef] [PubMed]
  132. Chun, C.; Lee, S.M.; Kim, C.W.; Hong, K.Y.; Kim, S.Y.; Yang, H.K.; Song, S.C. Doxorubicin-polyphosphazene conjugate hydrogels for locally controlled delivery of cancer therapeutics. Biomaterials 2009, 30, 4752–4762. [Google Scholar] [CrossRef] [PubMed]
  133. Zhao, L.; Zhu, L.; Liu, F.; Liu, C.; Shan, D.; Wang, Q.; Zhang, C.; Li, J.; Liu, J.; Qu, X.; et al. pH triggered injectable amphiphilic hydrogel containing doxorubicin and paclitaxel. Int. J. Pharm. 2011, 410, 83–91. [Google Scholar] [CrossRef]
  134. Wang, W.; Song, H.; Zhang, J.; Li, P.; Li, C.; Wang, C.; Kong, D.; Zhao, Q. An injectable, thermosensitive and multicompartment hydrogel for simultaneous encapsulation and independent release of a drug cocktail as an effective combination therapy platform. J. Control. Release 2015, 203, 57–66. [Google Scholar] [CrossRef]
  135. Wensing, K.U.; Ciarimboli, G. Saving ears and kidneys from cisplatin. Anticancer Res. 2013, 33, 4183–4188. [Google Scholar] [PubMed]
  136. Ciarimboli, G. Membrane transporters as mediators of cisplatin side-effects. Anticancer Res. 2014, 34, 547–550. [Google Scholar] [CrossRef]
  137. Taguchi, T.; Nazneen, A.; Abid, M.R.; Razzaque, M.S. Cisplatin-associated nephrotoxicity and pathological events. Contrib. Nephrol. 2005, 148, 107–121. [Google Scholar] [CrossRef]
  138. Chen, W.; Shi, K.; Liu, J.; Yang, P.; Han, R.; Pan, M.; Yuan, L.; Fang, C.; Yu, Y.; Qian, Z. Sustained co-delivery of 5-fluorouracil and cis-platinum via biodegradable thermo-sensitive hydrogel for intraoperative synergistic combination chemotherapy of gastric cancer. Bioact. Mater. 2023, 23, 1–15. [Google Scholar] [CrossRef]
  139. Shen, W.; Chen, X.; Luan, J.; Wang, D.; Yu, L.; Ding, J. Sustained Codelivery of Cisplatin and Paclitaxel via an Injectable Prodrug Hydrogel for Ovarian Cancer Treatment. ACS Appl. Mater. Interfaces 2017, 9, 40031–40046. [Google Scholar] [CrossRef]
  140. Peng, H.; Huang, Q.; Yue, H.; Li, Y.; Wu, M.; Liu, W.; Zhang, G.; Fu, S.; Zhang, J. The antitumor effect of cisplatin-loaded thermosensitive chitosan hydrogel combined with radiotherapy on nasopharyngeal carcinoma. Int. J. Pharm. 2019, 556, 97–105. [Google Scholar] [CrossRef]
  141. Li, J.; Gong, C.; Feng, X.; Zhou, X.; Xu, X.; Xie, L.; Wang, R.; Zhang, D.; Wang, H.; Deng, P.; et al. Biodegradable thermosensitive hydrogel for SAHA and DDP delivery: Therapeutic effects on oral squamous cell carcinoma xenografts. PLoS ONE 2012, 7, e33860. [Google Scholar] [CrossRef] [PubMed]
  142. Sharon Gabbay, R.; Rubinstein, A. Synchronizing the release rates of topotecan and paclitaxel from a self-eroding crosslinked chitosan—PLGA platform. Int. J. Pharm. 2022, 623, 121945. [Google Scholar] [CrossRef] [PubMed]
  143. Cocarta, A.I.; Hobzova, R.; Trchova, M.; Svojgr, K.; Kodetova, M.; Pochop, P.; Uhlik, J.; Sirc, J. 2-Hydroxyethyl Methacrylate Hydrogels for Local Drug Delivery: Study of Topotecan and Vincristine Sorption/Desorption Kinetics and Polymer-Drug Interaction by ATR-FTIR Spectroscopy. Macromol. Chem. Phys. 2021, 222, 2100086. [Google Scholar] [CrossRef]
  144. Bai, R.; Deng, X.; Wu, Q.; Cao, X.; Ye, T.; Wang, S. Liposome-loaded thermo-sensitive hydrogel for stabilization of SN-38 via intratumoral injection: Optimization, characterization, and antitumor activity. Pharm. Dev. Technol. 2018, 23, 106–115. [Google Scholar] [CrossRef] [PubMed]
  145. 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]
  146. Lu, X.; Lu, X.; Yang, P.; Zhang, Z.; Lv, H. Honokiol nanosuspensions loaded thermosensitive hydrogels as the local delivery system in combination with systemic paclitaxel for synergistic therapy of breast cancer. Eur. J. Pharm. Sci. 2022, 175, 106212. [Google Scholar] [CrossRef]
  147. McKenzie, M.; Betts, D.; Suh, A.; Bui, K.; Tang, R.; Liang, K.; Achilefu, S.; Kwon, G.S.; Cho, H. Proof-of-Concept of Polymeric Sol-Gels in Multi-Drug Delivery and Intraoperative Image-Guided Surgery for Peritoneal Ovarian Cancer. Pharm. Res. 2016, 33, 2298–2306. [Google Scholar] [CrossRef]
  148. Shi, K.; Xue, B.; Jia, Y.; Yuan, L.; Han, R.; Yang, F.; Peng, J.; Qian, Z. Sustained co-delivery of gemcitabine and cis-platinum via biodegradable thermo-sensitive hydrogel for synergistic combination therapy of pancreatic cancer. Nano Res. 2019, 12, 1389–1399. [Google Scholar] [CrossRef]
  149. Hodaei, M.; Varshosaz, J. Cationic Okra gum coated nanoliposomes as a pH-sensitive carrier for co-delivery of hesperetin and oxaliplatin in colorectal cancers. Pharm. Dev. Technol. 2022, 27, 773–784. [Google Scholar] [CrossRef] [PubMed]
  150. Wanjale, M.V.; Sunil Jaikumar, V.; Sivakumar, K.C.; Ann Paul, R.; James, J.; Kumar, G.S.V. Supramolecular Hydrogel Based Post-Surgical Implant System for Hydrophobic Drug Delivery Against Glioma Recurrence. Int. J. Nanomed. 2022, 17, 2203–2224. [Google Scholar] [CrossRef]
  151. Dvorak, H.F. Tumor Stroma, Tumor Blood Vessels, and Antiangiogenesis Therapy. Cancer J. 2015, 21, 237–243. [Google Scholar] [CrossRef] [PubMed]
  152. Forster, J.C.; Harriss-Phillips, W.M.; Douglass, M.J.; Bezak, E. A review of the development of tumor vasculature and its effects on the tumor microenvironment. Hypoxia 2017, 5, 21–32. [Google Scholar] [CrossRef]
  153. Jain, R.K. Determinants of Tumor Blood Flow: A Review1. Cancer Res. 1988, 48, 2641–2658. [Google Scholar]
  154. Tozer, G.M.; Kanthou, C.; Baguley, B.C. Disrupting tumour blood vessels. Nat. Rev. Cancer 2005, 5, 423–435. [Google Scholar] [CrossRef] [PubMed]
  155. Martin, J.D.; Seano, G.; Jain, R.K. Normalizing Function of Tumor Vessels: Progress, Opportunities, and Challenges. Annu. Rev. Physiol. 2019, 81, 505–534. [Google Scholar] [CrossRef] [PubMed]
  156. Giuliano, S.; Pages, G. Mechanisms of resistance to anti-angiogenesis therapies. Biochimie 2013, 95, 1110–1119. [Google Scholar] [CrossRef]
  157. Maj, E.; Papiernik, D.; Wietrzyk, J. Antiangiogenic cancer treatment: The great discovery and greater complexity (Review). Int. J. Oncol. 2016, 49, 1773–1784. [Google Scholar] [CrossRef]
  158. Jain, R.K. Normalizing tumor microenvironment to treat cancer: Bench to bedside to biomarkers. J. Clin. Oncol. 2013, 31, 2205–2218. [Google Scholar] [CrossRef]
  159. Pasquier, E.; Kavallaris, M.; Andre, N. Metronomic chemotherapy: New rationale for new directions. Nat. Rev. Clin. Oncol. 2010, 7, 455–465. [Google Scholar] [CrossRef]
  160. Liu, Y.; Kim, Y.J.; Siriwon, N.; Rohrs, J.A.; Yu, Z.; Wanga, P. Combination drug delivery via multilamellar vesicles enables targeting of tumor cells and tumor vasculature. Biotechnol. Bioeng. 2018, 115, 1403–1415. [Google Scholar] [CrossRef]
  161. Li, F.; Shao, X.; Liu, D.; Jiao, X.; Yang, X.; Yang, W.; Liu, X. Vascular Disruptive Hydrogel Platform for Enhanced Chemotherapy and Anti-Angiogenesis through Alleviation of Immune Surveillance. Pharmaceutics 2022, 14, 1809. [Google Scholar] [CrossRef]
  162. Westin, S.N.; Sood, A.K.; Coleman, R.L. Targeted Therapy and Molecular Genetics. In Clinical Gynecologic Oncology; Elsevier: Amsterdam, The Netherlands, 2012; pp. 539–560.e536. [Google Scholar]
  163. Drzyzga, A.; Cichon, T.; Czapla, J.; Jarosz-Biej, M.; Pilny, E.; Matuszczak, S.; Wojcieszek, P.; Urbas, Z.; Smolarczyk, R. The Proper Administration Sequence of Radiotherapy and Anti-Vascular Agent-DMXAA Is Essential to Inhibit the Growth of Melanoma Tumors. Cancers 2021, 13, 3924. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, T.; Yang, L.; Xie, Y.; Cheng, S.; Xiong, M.; Luo, X. An injectable hydrogel/staple fiber composite for sustained release of CA4P and doxorubicin for combined chemotherapy of xenografted breast tumor in mice. Nan Fang Yi Ke Da Xue Xue Bao 2022, 42, 625–632. [Google Scholar] [CrossRef] [PubMed]
  165. Sengupta, S.; Eavarone, D.; Capila, I.; Zhao, G.; Watson, N.; Kiziltepe, T.; Sasisekharan, R. Temporal targeting of tumour cells and neovasculature with a nanoscale delivery system. Nature 2005, 436, 568–572. [Google Scholar] [CrossRef] [PubMed]
  166. Montemagno, C.; Pages, G. Resistance to Anti-angiogenic Therapies: A Mechanism Depending on the Time of Exposure to the Drugs. Front. Cell Dev. Biol. 2020, 8, 584. [Google Scholar] [CrossRef] [PubMed]
  167. Tamiya, A. Long-term survival of patients with advanced non-small cell lung cancer treated using immune checkpoint inhibitors. Respir. Investig. 2024, 62, 85–89. [Google Scholar] [CrossRef]
  168. Chao, Y.; Liang, C.; Tao, H.; Du, Y.; Wu, D.; Dong, Z.; Jin, Q.; Chen, G.; Xu, J.; Xiao, Z.; et al. Localized cocktail chemoimmunotherapy after in situ gelation to trigger robust systemic antitumor immune responses. Sci. Adv. 2020, 6, eaaz4204. [Google Scholar] [CrossRef] [PubMed]
  169. Tang, L.; Fu, C.; Liu, H.; Yin, Y.; Cao, Y.; Feng, J.; Zhang, A.; Wang, W. Chemoimmunotherapeutic Nanogel for Pre- and Postsurgical Treatment of Malignant Melanoma by Reprogramming Tumor-Associated Macrophages. Nano Lett. 2024, 24, 1717–1728. [Google Scholar] [CrossRef]
  170. Sun, L.; Shen, F.; Tian, L.; Tao, H.; Xiong, Z.; Xu, J.; Liu, Z. ATP-Responsive Smart Hydrogel Releasing Immune Adjuvant Synchronized with Repeated Chemotherapy or Radiotherapy to Boost Antitumor Immunity. Adv. Mater. 2021, 33, e2007910. [Google Scholar] [CrossRef] [PubMed]
  171. Guo, H.; Nakajima, T.; Hourdet, D.; Marcellan, A.; Creton, C.; Hong, W.; Kurokawa, T.; Gong, J.P. Hydrophobic Hydrogels with Fruit-Like Structure and Functions. Adv. Mater. 2019, 31, 1900702. [Google Scholar] [CrossRef]
  172. Cui, W.; Zhu, R.; Zheng, Y.; Mu, Q.; Pi, M.; Chen, Q.; Ran, R. Transforming non-adhesive hydrogels to reversible tough adhesives via mixed-solvent-induced phase separation. J. Mater. Chem. A 2021, 9, 9706–9718. [Google Scholar] [CrossRef]
  173. Wang, M.; Sun, R.; Chen, H.; Liu, X.; Yoshitomi, T.; Takeguchi, M.; Kawazoe, N.; Yang, Y.; Chen, G. Influence of hydrogel and porous scaffold on the magnetic thermal property and anticancer effect of Fe3O4 nanoparticles. Microstructures 2023, 3, 2023042. [Google Scholar] [CrossRef]
  174. Constantinou, A.P.; Georgiou, T.K. Pre-clinical and clinical applications of thermoreversible hydrogels in biomedical engineering: A review. Polym. Int. 2021, 70, 1433–1448. [Google Scholar] [CrossRef]
  175. Xu, S.; Wang, W.; Li, X.; Liu, J.; Dong, A.; Deng, L. Sustained release of PTX-incorporated nanoparticles synergized by burst release of DOXHCl from thermosensitive modified PEG/PCL hydrogel to improve anti-tumor efficiency. Eur. J. Pharm. Sci. 2014, 62, 267–273. [Google Scholar] [CrossRef]
  176. Gao, Y.; Ren, F.; Ding, B.; Sun, N.; Liu, X.; Ding, X.; Gao, S. A thermo-sensitive PLGA-PEG-PLGA hydrogel for sustained release of docetaxel. J. Drug Target. 2011, 19, 516–527. [Google Scholar] [CrossRef] [PubMed]
  177. Turabee, M.H.; Jeong, T.H.; Ramalingam, P.; Kang, J.H.; Ko, Y.T. N,N,N-trimethyl chitosan embedded in situ Pluronic F127 hydrogel for the treatment of brain tumor. Carbohydr. Polym. 2019, 203, 302–309. [Google Scholar] [CrossRef]
  178. Zhu, Y.; Jia, J.; Zhao, G.; Huang, X.; Wang, L.; Zhang, Y.; Zhang, L.; Konduru, N.; Xie, J.; Yu, R.; et al. Multi-responsive nanofibers composite gel for local drug delivery to inhibit recurrence of glioma after operation. J. Nanobiotechnol. 2021, 19, 198. [Google Scholar] [CrossRef]
  179. Zhao, Z.; Shen, J.; Zhang, L.; Wang, L.; Xu, H.; Han, Y.; Jia, J.; Lu, Y.; Yu, R.; Liu, H. Injectable postoperative enzyme-responsive hydrogels for reversing temozolomide resistance and reducing local recurrence after glioma operation. Biomater. Sci. 2020, 8, 5306–5316. [Google Scholar] [CrossRef]
  180. Li, J.; Zhao, J.; Tan, T.; Liu, M.; Zeng, Z.; Zeng, Y.; Zhang, L.; Fu, C.; Chen, D.; Xie, T. Nanoparticle Drug Delivery System for Glioma and Its Efficacy Improvement Strategies: A Comprehensive Review. Int. J. Nanomed. 2020, 15, 2563–2582. [Google Scholar] [CrossRef]
  181. Gregory, J.V.; Kadiyala, P.; Doherty, R.; Cadena, M.; Habeel, S.; Ruoslahti, E.; Lowenstein, P.R.; Castro, M.G.; Lahann, J. Systemic brain tumor delivery of synthetic protein nanoparticles for glioblastoma therapy. Nat. Commun. 2020, 11, 5687. [Google Scholar] [CrossRef]
  182. Kim, J.; Francis, D.M.; Thomas, S.N. In Situ Crosslinked Hydrogel Depot for Sustained Antibody Release Improves Immune Checkpoint Blockade Cancer Immunotherapy. Nanomaterials 2021, 11, 471. [Google Scholar] [CrossRef]
  183. Zhang, Y.; Tian, S.; Huang, L.; Li, Y.; Lu, Y.; Li, H.; Chen, G.; Meng, F.; Liu, G.L.; Yang, X.; et al. Reactive oxygen species-responsive and Raman-traceable hydrogel combining photodynamic and immune therapy for postsurgical cancer treatment. Nat. Commun. 2022, 13, 4553. [Google Scholar] [CrossRef] [PubMed]
  184. Jin, H.; Wan, C.; Zou, Z.; Zhao, G.; Zhang, L.; Geng, Y.; Chen, T.; Huang, A.; Jiang, F.; Feng, J.P.; et al. Tumor Ablation and Therapeutic Immunity Induction by an Injectable Peptide Hydrogel. ACS Nano 2018, 12, 3295–3310. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, W.; Deng, L.; Xu, S.; Zhao, X.; Lv, N.; Zhang, G.; Gu, N.; Hu, R.; Zhang, J.; Liu, J.; et al. A reconstituted “two into one” thermosensitive hydrogel system assembled by drug-loaded amphiphilic copolymer nanoparticles for the local delivery of paclitaxel. J. Mater. Chem. B 2013, 1, 552–563. [Google Scholar] [CrossRef]
  186. Jin, J.F.; Zhu, L.L.; Chen, M.; Xu, H.M.; Wang, H.F.; Feng, X.Q.; Zhu, X.P.; Zhou, Q. The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection. Patient Prefer. Adherence 2015, 9, 923–942. [Google Scholar] [CrossRef]
  187. Melero, I.; Castanon, E.; Alvarez, M.; Champiat, S.; Marabelle, A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat. Rev. Clin. Oncol. 2021, 18, 558–576. [Google Scholar] [CrossRef]
  188. Marabelle, A.; Tselikas, L.; de Baere, T.; Houot, R. Intratumoral immunotherapy: Using the tumor as the remedy. Ann. Oncol. 2017, 28, xii33–xii43. [Google Scholar] [CrossRef]
  189. Ruel-Gariépy, E.; Shive, M.; Bichara, A.; Berrada, M.; Le Garrec, D.; Chenite, A.; Leroux, J.-C. A thermosensitive chitosan-based hydrogel for the local delivery of paclitaxel. Eur. J. Pharm. Biopharm. 2004, 57, 53–63. [Google Scholar] [CrossRef]
  190. Duffy, C.V.; David, L.; Crouzier, T. Covalently-crosslinked mucin biopolymer hydrogels for sustained drug delivery. Acta Biomater. 2015, 20, 51–59. [Google Scholar] [CrossRef] [PubMed]
  191. Davoodi, P.; Ng, W.C.; Yan, W.C.; Srinivasan, M.P.; Wang, C.H. Double-Walled Microparticles-Embedded Self-Cross-Linked, Injectable, and Antibacterial Hydrogel for Controlled and Sustained Release of Chemotherapeutic Agents. ACS Appl. Mater. Interfaces 2016, 8, 22785–22800. [Google Scholar] [CrossRef] [PubMed]
  192. Chen, N.; Wang, H.; Ling, C.; Vermerris, W.; Wang, B.; Tong, Z. Cellulose-based injectable hydrogel composite for pH-responsive and controllable drug delivery. Carbohydr. Polym. 2019, 225, 115207. [Google Scholar] [CrossRef]
  193. Tahir, D.; Ardiansyah, A.; Heryanto, H.; Noor, E.E.M.; Mohamed, M.A. Chitosan-based hydrogels: A comprehensive review of transdermal drug delivery. Int. J. Biol. Macromol. 2025, 298, 140010. [Google Scholar] [CrossRef] [PubMed]
  194. Borchers, A.; Pieler, T. Programming pluripotent precursor cells derived from Xenopus embryos to generate specific tissues and organs. Genes 2010, 1, 413–426. [Google Scholar] [CrossRef]
  195. Frumento, D.; Țălu, Ș. Immunomodulatory Potential and Biocompatibility of Chitosan–Hydroxyapatite Biocomposites for Tissue Engineering. J. Compos. Sci. 2025, 9, 305. [Google Scholar] [CrossRef]
  196. Pagar, R.R.; Musale, S.R.; Pawar, G.; Kulkarni, D.; Giram, P.S. Comprehensive Review on the Degradation Chemistry and Toxicity Studies of Functional Materials. ACS Biomater. Sci. Eng. 2022, 8, 2161–2195. [Google Scholar] [CrossRef]
  197. Witherel, C.E.; Abebayehu, D.; Barker, T.H.; Spiller, K.L. Macrophage and Fibroblast Interactions in Biomaterial-Mediated Fibrosis. Adv. Healthc. Mater. 2019, 8, 1801451. [Google Scholar] [CrossRef]
  198. Taylor, M.S.; Daniels, A.U.; Andriano, K.P.; Heller, J. Six bioabsorbable polymers: In vitro acute toxicity of accumulated degradation products. J. Appl. Biomater. 1994, 5, 151–157. [Google Scholar] [CrossRef]
  199. Kim, M.-N.; Lee, B.-Y.; Lee, I.-M.; Lee, H.-S.; Yoon, J.-S. TOXICITY AND BIODEGRADATION OF PRODUCTS FROM POLYESTER HYDROLYSIS. J. Environ. Sci. Health Part. A 2001, 36, 447–463. [Google Scholar] [CrossRef] [PubMed]
  200. Innovative Therapeutic Treatments to Inhibit Perineural Invasion in Pancreatic Adenocarcinoma. 2024. Available online: https://www.clinicaltrials.gov/study/NCT06616688?term=inhibit&viewType=Table&checkSpell=&rank=7 (accessed on 31 July 2025).
  201. Fluorouracil Treatment Via Colon for Colorectal Cancer: An Exploratory Study. 2024. Available online: https://clinicaltrials.gov/study/NCT06385418 (accessed on 31 July 2025).
  202. Harris, E. Industry update: The latest developments in the field of therapeutic delivery, January 2025. Ther. Deliv. 2025, 16, 407–414. [Google Scholar] [CrossRef]
  203. Prasad, S.M.; Shishkov, D.; Mihaylov, N.V.; Khuskivadze, A.; Genov, P.; Terzi, V.; Kates, M.; Huang, W.C.; Louie, M.J.; Raju, S.; et al. Primary Chemoablation of Recurrent Low-Grade Intermediate-Risk Nonmuscle-Invasive Bladder Cancer with UGN-102: A Single-Arm, Open-Label, Phase 3 Trial (ENVISION). J. Urol. 2025, 213, 205–216. [Google Scholar] [CrossRef] [PubMed]
  204. Yates, A.H.; Power, J.W.; Dempsey, P.J.; Agnew, A.; Murphy, B.D.; Moore, R.; El Bassiouni, M.; McNicholas, M.M.J. SpaceOAR hydrogel distribution and early complications in patients undergoing radiation therapy for prostate cancer. Br. J. Radiol. 2023, 96, 20220947. [Google Scholar] [CrossRef]
  205. A Randomized, Controlled, Open-Label Study of the Efficacy, Durability, and Safety of UGN-102 with or Without TURBT in Patients With Low-Grade Intermediate-Risk Non-Muscle Invasive Bladder Cancer (LG-IR-NMIBC). 2020. Available online: https://clinicaltrials.gov/study/NCT04688931 (accessed on 31 July 2025).
  206. Feasibility and Safety of Local Immunomodulation Combined With Radiofrequency Ablation for Unresectable Colorectal Liver Metastases: A Monocentric Phase I Trial. 2019. Available online: https://clinicaltrials.gov/study/NCT04062721 (accessed on 31 July 2025).
Figure 1. Schematic illustration of various strategies used to prepare hydrogel-based drug delivery systems (DDSs) for antitumor therapy. The illustration was created using BioRender, https://app.biorender.com/ (accessed on 27 June 2025).
Figure 1. Schematic illustration of various strategies used to prepare hydrogel-based drug delivery systems (DDSs) for antitumor therapy. The illustration was created using BioRender, https://app.biorender.com/ (accessed on 27 June 2025).
Biomedicines 13 01899 g001
Figure 2. Extracellular matrix (ECM) stiffness affects chemoresistance of breast cancer cells to doxorubicin. Following encapsulation and culture in hydrogels, both cell viability and P-gp mRNA expression were found to decrease in the soft hydrogel, whereas they increased in the hard hydrogel. Copyright: Guoping Chen. There is no copyright issue [108].
Figure 2. Extracellular matrix (ECM) stiffness affects chemoresistance of breast cancer cells to doxorubicin. Following encapsulation and culture in hydrogels, both cell viability and P-gp mRNA expression were found to decrease in the soft hydrogel, whereas they increased in the hard hydrogel. Copyright: Guoping Chen. There is no copyright issue [108].
Biomedicines 13 01899 g002
Figure 3. Anticancer experimental scheme of free Fe3O4 NPs (A), agarose/Fe3O4 hydrogels (BD), and gelatin/Fe3O4 porous scaffolds (EG). Three modes (sitting, Transwell, and adhesion modes) were used to simulate the cells near or far away from or directly adhered to the matrices. Copyright: Guoping Chen. There is no copyright issue [173].
Figure 3. Anticancer experimental scheme of free Fe3O4 NPs (A), agarose/Fe3O4 hydrogels (BD), and gelatin/Fe3O4 porous scaffolds (EG). Three modes (sitting, Transwell, and adhesion modes) were used to simulate the cells near or far away from or directly adhered to the matrices. Copyright: Guoping Chen. There is no copyright issue [173].
Biomedicines 13 01899 g003
Figure 4. Summary of the methods used to analyze antitumor effects at the cellular level.
Figure 4. Summary of the methods used to analyze antitumor effects at the cellular level.
Biomedicines 13 01899 g004
Figure 5. Summary of the methods used for animal experiments.
Figure 5. Summary of the methods used for animal experiments.
Biomedicines 13 01899 g005
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

Zeng, T.; Chen, L.; Yoshitomi, T.; Kawazoe, N.; Yang, Y.; Chen, G. Research Strategies and Methods of Hydrogels for Antitumor Drug Delivery. Biomedicines 2025, 13, 1899. https://doi.org/10.3390/biomedicines13081899

AMA Style

Zeng T, Chen L, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Research Strategies and Methods of Hydrogels for Antitumor Drug Delivery. Biomedicines. 2025; 13(8):1899. https://doi.org/10.3390/biomedicines13081899

Chicago/Turabian Style

Zeng, Tianjiao, Lusi Chen, Toru Yoshitomi, Naoki Kawazoe, Yingnan Yang, and Guoping Chen. 2025. "Research Strategies and Methods of Hydrogels for Antitumor Drug Delivery" Biomedicines 13, no. 8: 1899. https://doi.org/10.3390/biomedicines13081899

APA Style

Zeng, T., Chen, L., Yoshitomi, T., Kawazoe, N., Yang, Y., & Chen, G. (2025). Research Strategies and Methods of Hydrogels for Antitumor Drug Delivery. Biomedicines, 13(8), 1899. https://doi.org/10.3390/biomedicines13081899

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