Next Article in Journal
Green Chemistry for the Transformation of Chlorinated Wastes: Catalytic Hydrodechlorination on Pd-Ni and Pd-Fe Bimetallic Catalysts Supported on SiO2
Next Article in Special Issue
DOE-Assisted Formulation, Optimization, and Characterization of Tioconazole-Loaded Transferosomal Hydrogel for the Effective Treatment of Atopic Dermatitis: In Vitro and In Vivo Evaluation
Previous Article in Journal
Beyond Sol-Gel: Molecular Gels with Different Transitions
Previous Article in Special Issue
Development and Evaluation of Novel Encapsulated Isoeugenol-Liposomal Gel Carrier System for Methicillin-Resistant Staphylococcus aureus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Use of Hydrogels for the Treatment of Bone Osteosarcoma via Localized Drug-Delivery and Tissue Regeneration: A Narrative Review

1
College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA
2
Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
3
Department of Electrical and Computing Engineering, New York Institute of Technology, Old Westbury, NY 11568, USA
*
Author to whom correspondence should be addressed.
Submission received: 9 March 2023 / Revised: 22 March 2023 / Accepted: 23 March 2023 / Published: 25 March 2023
(This article belongs to the Special Issue Gel-Based Drug Delivery Systems for Cancer Treatment)

Abstract

:
Osteosarcoma is a malignant tumor of bone that leads to poor mortality and morbidity. Management of this cancer through conventional methods involves invasive treatment options that place patients at an increased risk of adverse events. The use of hydrogels to target osteosarcoma has shown promising results both in vitro and in vivo to eradicate tumor cells while promoting bone regeneration. The loading of hydrogels with chemotherapeutic drugs provides a route for site-specific targeted therapy for osteosarcoma. Current studies demonstrate tumor regression in vivo and lysis of tumor cells in vitro when exposed to doped hydrogel scaffolds. Additionally, novel stimuli-responsive hydrogels are able to react with the tissue microenvironment to facilitate the controlled release of anti-tumor drugs and with biomechanical properties that can be modulated. This narrative review of the current literature discusses both in vitro and in vivo studies of different hydrogels, including stimuli-responsive, designed to treat bone osteosarcoma. Future applications to address patient treatment for this bone cancer are also discussed.

1. Introduction

Bone malignancies are an uncommon sporadic form of cancer primarily affecting adolescents and older adults. Primary bone tumors are rare and account for less than 1% of all cancers. Rather, bone metastasis is a commonly seen phenomenon, with 30–75% of advanced malignant tumors presenting this way [1]. Of the primary bone tumors, osteosarcoma is the most common primary bone cancer overall in children and teens; however, it can also be seen in the elderly [2]. In adults, the most common primary bone tumor is chondrosarcoma, followed by chordoma and osteosarcoma [3]. Osteosarcoma has the lowest 5-year survival rate for bone cancers across all Surveillance, Epidemiology, and End Results (SEER) stages. Osteosarcoma has a 60% 5-year survival rate compared to 79% in chondrosarcoma and giant cell tumor of bone [4]. Osteosarcoma takes a severe toll on patients by a reduction of their quality of life, even with or without treatment.
Unfortunately, osteosarcoma is a difficult tumor to treat. Often, physicians recommend surgery, radiation, chemotherapy, or a combination of these options as aggressive treatment. Currently, neoadjuvant chemotherapy with surgical resection of the tumor, followed by additional adjuvant therapy, is one of the current regimens for osteosarcoma therapy [5]. In the past, severe osteosarcoma would require the removal of a large portion of bone, often resulting in limb amputation [6,7]. With the advancement of surgical precision and techniques, patients can undergo limb-sparing surgery with a prosthetic replacement or graft [8,9,10]. While nearly 85% of osteosarcomas are treated this way, surgery is highly invasive, and patients endure a loss of function in mobility and lifestyle [11]. The diagnosis, treatment, and management of osteosarcoma place a physical and psychological burden on patients and their families and a financial burden on the healthcare system. Additionally, surgical complications may predispose patients to infection, periprosthetic fractures, and dehiscence [12]. Coupled with the enormous side-effect profile of chemotherapeutic agents, patients often tolerate the treatment very poorly despite increased survival [5,13].
Current in vivo and in vitro studies focus on generating mouse models to identify and target ‘driver mutations’ for effective drug therapy. Numerous gene clusters have been identified to contribute to osteosarcoma: p53, Notch1, PTEN, BRCA2, FOS, RET, ATM, and FANCA amongst others [14]. Driver-dependent therapy targets constitutively active signaling pathways to downregulate tumor growth. In osteosarcoma, the PI3K/mTOR signaling pathway has been identified as common for cancer proliferation [15]. Unfortunately, in nearly 25% of patients, this altered pathway is affected, thus, limiting kinase-targeted treatment to just this subset of patients [15]. However, the most commonly seen mutation is in p53, but therapeutic targeting of p53 is difficult due to many mutations and downstream effects [14]. Investigation into the pharmaceutical modulation of p53 has been conducted with Nutlin-3a, a ubiquitin ligase antagonist (MDM2 antagonist) [16]. Preclinical sensitivity to osteosarcoma cells was observed; however, the clinical activity in patients with liposarcoma indicated an unsatisfactory response [17]. Novel MDM2 inhibitors have not been tested in treating osteosarcoma in preclinical or clinical settings. Therefore, current chemotherapy focuses on using conventional chemotherapeutic drugs such as methotrexate, doxorubicin, cisplatin, ifosfamide, cyclophosphamide, etoposide, or gemcitabine [18,19].
While patient tolerance to chemotherapy drugs and tissue loss post-surgery are concerns, this can potentially be addressed simultaneously with drug-loaded hydrogels directly targeting the affected osteosarcoma site [20]. In tissue engineering, hydrogels and polymeric implants or scaffolds have been used for their ability to simultaneously release preloaded drugs and induce native tissue regeneration [21,22]. In preclinical osteosarcoma models, hydrogels were studied for their capacity to reduce tumor size with enhanced drug release. In this review, we describe the use of drug-loaded hydrogels for osteosarcoma therapy and subsequent bone regeneration. Furthermore, in-depth details about the future applications of these implantable scaffolds to effectively treat osteosarcoma in animal models are also discussed.

2. Materials and Methods

PubMed was queried for articles with the search term “Hydrogel Bone Regeneration,” which yielded 2491 results. Of the 2491 hits, 1473 articles were studies conducted in vitro, and 866 articles were conducted in vivo. The search was further restricted to the terms “Hydrogel-osteosarcoma treatment,” “tissue engineering,” and “drug-delivery,” resulting in 113 results, of which 63 were in vitro studies and 30 in vivo studies (Figure 1). Herein, we summarize the results of these studies.

3. Potential Therapies for Osteosarcoma

The current etiology of osteosarcoma is unknown; however, it is associated with syndromes linked to tumor suppressor gene mutations such as Li-Fraumeni syndrome (p53 mutation), retinoblastoma (Rb mutation), and multiple genetic mutations [14,23,24]. Additionally, exposure to radiation is a risk factor for developing osteosarcoma [1]. Clinically, patients often present with localized bone pain, swelling, and limited range of motion in the affected region. Rarely, pathologic fractures may be present due to osteolytic tumor formation [25,26]. The diagnosis is made through gross imaging studies, such as X-rays, to detect bony tumor formation. However, small tumors are likely not detected through imaging modalities [27]. Recently, advances have been made in the detection of osteosarcoma through the use of nanomaterials in a preclinical setting. Radiolabeled nanomaterials are used to mark tumor cells which in turn increases imaging intensity [28]. Non-invasive imaging through fluorescence dyes attached to nanoparticles can target osteosarcoma and metastasis in mice [29]. While the primary treatment of osteosarcoma includes surgery, chemotherapy, and radiation, patients require multidisciplinary care; a team of surgeons, oncologists, physical therapists, and psychiatrists, among others, are necessary for the proper multifactorial treatment of osteosarcoma in order to address the patient’s medical and socioeconomic needs [30]. Despite extensive involvement in patient care and treatment options, outcomes are usually poor for osteosarcoma, and the exploration of novel treatment options is necessary to reduce patient morbidity and mortality.
Targeted therapy and immunotherapy are popular modalities for treating cancers by targeting driver mutations that result in overexpressed genes and have been performed with lung, thyroid, and skin cancers with promising results [31,32,33]. In osteosarcoma, genetic heterogeneity allows for a wide array of molecular targets for targeted therapy and immunotherapy [14]. Inhibitors against PI3K/mTOR have been developed and have shown anti-tumor effects in vitro; however, not all patients carry this mutation. Alternatively, PTEN is a negative regulator of PI3K/mTOR and can serve as a potential target in future trials [34]. Another common mutation is seen in vascular endothelial growth factor (VEGF), a major regulator of angiogenesis and cell growth. VEGF expression is often increased with metastasis. Anti-VEGF drugs have demonstrated anti-tumor activity against osteosarcoma in clinical trials for single tumors and metastasis [35,36,37,38,39]. Future advancements can place a more focused approach on VEGF mutations to greatly reduce treatment time and survivability in these patients. A promising method to target hematological cancers is known as chimeric antigen receptor (CAR) T-cell therapy [40]. This modality utilizes the patient’s own T-cells modified with CARs to directly target antigens expressed on the malignancy. A major limitation is the lack of use in solid malignancies such as osteosarcoma [40]. To date, multiple ongoing clinical trials are being conducted on CAR-T cell therapy for osteosarcoma; however, no published data have been reported [41]. Interestingly, CAR-T cell therapy against GD2, a glycolipid with low expression in normal tissue, is being explored [42]. GD2 is highly expressed in solid tumors, including osteosarcoma, and has been targeted prior as a treatment for neuroblastoma [43]. Additionally, preclinical studies have demonstrated efficacy in tumor death with GD2-targeted CAR-T cells [42,44]. As a result, CAR-T cell therapy is a viable option for osteosarcoma treatment once further research is conducted.

4. Tissue Engineering for Bioresponsive Hydrogels

4.1. Synthesis, Fabrication, and Properties of Hydrogels

Hydrogels are polymeric hydrophilic structures that are capable of absorbing up to thousands of times their dry weight in water without losing their chemical or structural properties [45,46]. Hydrogels are physical gels when the internal networks are connected through molecular interactions or secondary forces, including hydrogen bonding, hydrophobic forces, or ionic forces [45]. Chemical hydrogels have permanent physical bonds since they contain covalently crosslinked internal networks [46]. Chemical hydrogels are generated by transforming hydrophobic polymers into hydrophilic polymers and crosslinking them to form a network [45]. Interestingly, physical hydrogels are less stable due to their dependence on external stimuli such as pH, temperature, and ionic strength. On the contrary, chemical hydrogels are less dependent on external stimuli and have a tendency for greater mechanical resistance [46]. Finally, the swelling of hydrogels reaches a limit once the cohesive and osmotic forces reach equilibrium [47,48].
In the preparation of a physical gel, polymer selection depends on two important criteria: (1) a large amount of water molecules must be held within the hydrogel, and (2) the interactions between the polymer chains must be strong enough to form a semi-permanent connection in the molecular network [49]. The forces involved in physical gel formation include hydrogen bonding, hydrophobic forces, and electrostatic interactions between polymer chains [50]. Physically crosslinked hydrogels are assembled without the use of chemical modifications or crosslinking materials [49]. For example, alginate can be crosslinked using divalent cations within a solution [48]. However, despite these advantages, physical hydrogels have inconsistent performance in vivo due to their inflexibility towards gel pore size, chemical functionalization, and degradation [49].
Chemically crosslinked hydrogels allow for drug release through diffusion without dissolution. Chemical crosslinking produces a permanent hydrogel through covalent bonding to connect the polymers [49]. The formation of crosslinks can be performed by the introduction of small cross-linker molecules, photosensitive agents, or by an enzyme-catalyzed reaction. The simplest form of crosslinking occurs between amino groups and aldehydes, forming a Schiff base [51]. In particular, glutaraldehyde can form imine bonds with the amino groups of chitosan through a Schiff reaction [52]. However, a disadvantage of small-molecule crosslinking methods is the possible toxicity of unreacted cross-linker agents in vivo. Photo crosslinking formation depends on the existence of photosensitive functional groups. Conjugating a photosensitive functional group to a polymer allows it to create crosslinks after UV irradiation. For example, azide groups can be incorporated into a polymeric chain of chitosan to create a photo-crosslinked chitosan hydrogel [49]. After UV exposure, the azide groups convert to nitrene groups that are capable of binding free amino groups present in chitosan and lead to in situ formation of a hydrogel within a minute [53]. Photo crosslinking also allows for the rapid formation of hydrogels at a low cost compared to chemical methods that generally require the expensive additions of initiators and/or catalysts [49,54]. However, a disadvantage of this technique is the requirement of prolonged irradiation, which can result in a local rise in temperature that can damage nearby cells and tissue [55]. Finally, enzymatic crosslinking is another approach to linking polymer chains to develop in situ hydrogels. Under physiological conditions, injectable hydrogels were developed by enzymatically crosslinking poly(L-glutamic acid) grafted with tyramine and poly(ethylene glycol), which was shown to have biomedical applications in tissue engineering scaffolds and drug delivery [56]. Advantages of this approach are that enzymatic-cross linked hydrogels are substrate specific, dynamic, and can be applied to controlled drug release systems [57]. Additionally, enzymatic reactions catalyzed by most enzymes occur at neutral pH, moderate temperatures, and aqueous environments, demonstrating applications for in situ formation of hydrogels. Disadvantages of this approach are limited mechanical properties of the gels, instability of some of the enzyme types such as transglutaminases, and few studies conducted utilizing enzymatically-cross linked hydrogels in vivo [57].
Hydrogels may be fabricated through a variety of methods. Novel methods for rapid and efficient production include using 3D printers to create 3D scaffolds for in vitro or in vivo testing. Most importantly, bioprinters can be loaded with different bioinks containing the biomaterial for fabrication [58]. A key benefit of this method is the encapsulation of cells within the bioink prior to printing, thus avoiding the variability in cell seeding in post-printed constructs [48]. Common printing methods include extrusion, inkjet, stereolithography, and laser printing. Each method carries benefits and risks to encapsulated cells in the bioink during the printing process. For example, extrusion printing exposes cells to high-pressure gradients resulting in the lysis of cells during printing. However, this process can print large concentrations of cells. In contrast, laser printing demonstrates no genotoxicity during printing; however, it is an expensive device with complex usage [59]. Overall, the rapid production of hydrogels through 3D printing provides a route for reproducible scaffolds for basic science testing.

4.2. Biomedical Applications of Various Hydrogels

Hydrogels have a wide range of biomedical applications as they are biocompatible and biomimetic. The biocompatibility of hydrogels comes from the lack of immunogenic antigens to stimulate a local immune response. The biomimetic capability replicates the extracellular matrix (ECM) with the structure and composition of the gel [60]. Hydrogels can be engineered to control molecular responses, cellular attachments, structural integrity, biocompatibility, and biodegradability. An ideal hydrogel scaffold should mimic the properties of the native tissue’s ECM, allow cell attachments, cell migration, and nutrient diffusion, and induce nearby cells to modify cell behavior [60]. To create bio-responsive scaffolds, researchers have developed synthetic and natural hydrogels that contain a high surface area to volume ratio that mimics the natural tissue micro and macroenvironment [61].
A common natural material used in hydrogels is collagen, the most abundant animal ECM protein. Collagen fibers are triple-stranded helical structures bounded by covalent bonds and hydrogens bonds rendering the fibrils to be self-aggregating [60]. Collagen is an ideal material for hydrogels as it is a natural ECM protein and contains binding sites to encourage cell adhesion and function. Collagen is degraded naturally by serine proteases and collagenases, allowing locally controlled degradation and ECM remodeling by the cells present in the target tissue [62]. Electrospinning, a viable technique used for generating homogenous ultra-thin fibers [60], generates well-aligned and biocompatible collagen fibers. Electrospinning is necessary to create hydrogels with high surface-area-to-volume ratios and porosity to mimic the ECM [63,64]. The heat and low pressure of electrospinning result in weak denaturation, allowing crosslinks between collagen fibers to form and thus increase biocompatibility [60]. Gelatin, a denatured form of collagen, is another natural material used in hydrogels as it retains important bioactive properties such as RGD sequences and metalloproteinase degradation sites [65]. Gelatin is an attractive polymer for tissue engineering due to its ease of modification and low levels of cytotoxicity and immunogenicity [66]. The biocompatibility of gelatin and gelatin-based biomaterials was improved by incorporating a surface-modified hydroxyapatite sponge scaffold and using an alternating electrospinning and soaking process [60]. The electrospinning and soaking processes are used to generate gelatin hydrogels that contain the correct porosity needed for biocompatibility. Additionally, the alternate soaking process was found to promote better cellular proliferation and adhesion [60].
Synthetic polymers such as poly(lactic acid), poly(glycolic acid), poly(ethylene oxide), and poly(ethylene glycol) offer scientists versatile materials with chemical and physical properties that can be altered. Poly(lactic acid) and poly(glycolic acid) are some of the most studied synthetic polyesters that have been shown to have satisfactory biocompatibility and are FDA approved for specific human clinical applications [60,64]. Recently, nano-hydroxyapatite/Poly(lactic acid) scaffolds have been found to be biocompatible with MG-63 cells and derived human bone marrow mesenchymal stem cells [67,68]. Poly(ethylene oxide) and poly(ethylene glycol) are also FDA-approved and are some of the most commonly used polymers utilized in hydrogel synthesis for tissue engineering [60]. Both poly(ethylene oxide) and poly(ethylene glycol) have adjustable mechanical properties that make them excellent scaffolds for producing 3D templates for tissue regeneration [69]. Overall, synthetic blends and polymers are easier to process than naturally derived polymers and have more predictable results. However, synthetic polymers are not as bioactive or biocompatible as naturally derived polymers [70].
Stimuli-responsive hydrogels, or “smart” hydrogels, are used to stimulate drug release based on external triggers such as temperature, pH, the concentration of biomolecules, electrical fields, and light [71]. Smart hydrogels respond to triggers in a manner that is predictable, reversible, intensity-scalable, and reproducible [72]. Additionally, stimuli-responsive hydrogels have the capability of returning to their original shape after the trigger is withdrawn [73]. Smart hydrogels have many advantages in drug delivery systems as they reduce dosing frequencies, maintain the correct therapeutic concentration in a single dose, and minimize drug side effects [74]. For example, pH-responsive hydrogels, which are high molecular polymers, undergo volume or phase transitions when the pH of the external medium is altered. pH-responsive hydrogels are useful because certain injuries, such as wounds and inflammation, cause pH variations that can be exploited for targeted drug delivery to specific tissues and organs [72]. Another example of a smart hydrogel is a temperature-responsive hydrogel that changes shape, volume, or size due to changes in physiological temperatures [75]. Most utilized temperature-responsive hydrogels are liquid or semi-solid at ambient temperatures and undergo a solid-to-gel transition at body temperature. This characteristic allows scientists to load a therapeutic compound onto a hydrogel in the liquid state and then easily solidify and administer it upon application. Through these advancements in tissue engineering, hydrogels are able to be developed to target osteosarcoma to induce tumor cell death and increase survivability (Figure 2).

5. Hydrogels for Osteosarcoma Therapy

5.1. Drug Delivery

Combination drug therapies are commonly administered to induce cancer cell death in osteosarcoma. However, only a small portion of intravenously administered drugs can reach their organ target in vivo, while the high dose can damage surrounding tissues and result in toxicity [77]. Further, neurotoxicity may also occur due to drug penetration of the blood-brain barrier leading to increased adverse events. These off-site adverse effects reduce the efficacy and bioavailability of existing chemotherapeutic agents [78]. Due to their extensive 3D polymerized structure, hydrogels are capable of being effective drug depositories to allow for local drug delivery with the ability to respond to exogenous and endogenous triggers. The use of hydrogels as a repository for drug delivery is an approach that has gained attention in the last decade as it proves to be an effective targeted therapy for localized pathology [77,79]. Additionally, hydrogels have the capability of being integrated with other drug delivery systems, such as liposomes or microspheres, which synergistically allows for better performance [80]. For example, Wu et al. (2018), photo crosslinked gemcitabine (GEM) hydrochloride loaded liposomes with a gelatin methacryloyl (GelMA) hydrogel to test its efficacy for osteosarcoma ablation [81]. The authors observed a controlled and sustained release of GEM from the hydrogel that lasted 4 days in vitro. Additionally, the hydrogel demonstrated the ability to inhibit osteosarcoma in vivo (BALB/c MG-63 bearing mice) [81].
Due to the diversity of tumor pathogenesis, the effect of a single chemotherapeutic drug may not be successful and synergistic chemotherapy is required to address this issue. A recent study by Zheng et al. (2017) loaded combretastatin A-4 (CA4) and docetaxel (DTX) into a thermosensitive injectable polypeptide hydrogel and reported the preferential release of CA4 in 80 eight-week-old BALB/c male mice with K7 osteosarcoma cells [82]. CA4 can disrupt blood vessel structure in tumors by binding endothelial cell tubulin and blocking tumor necrosis by inhibiting the exchange of oxygen and other nutrients [83]. The sustained release of DTX led to the destruction of tumor surface cells, resulting in apoptosis. The co-loading of both CA4 and DTX resulted in a significantly smallest tumor volume change compared to the control and the largest necrotic area of ~90.5%. Histological analyses of internal organs (heart, liver, spleen, lung, kidney) indicated no toxicity of the dual-loaded hydrogel [82]. Additionally, the in vitro release of CA4 and DTX indicated that within 48 days, 54% of the loaded DTX was released from the co-loaded hydrogel with no burst release observed (Figure 3). The constant release observed of DTX ensured that the effective drug concentration was met in situ, which avoided significant side effects. However, within the same time period, 90% of CA4 was released from the co-loaded gel. The scientists noted a slight initial burst release during the first 8 days as a result of its quick diffusion from the co-loaded gel surface [82].
Similarly, Sun et al. (2020) co-loaded Alendronate (ALN) and Oxaliplatin (OXA) onto a mPEG45-PLV19 thermosensitive hydrogel [84]. OXA is a widely used anti-tumor drug that contains platinum atoms that crosslink DNA to antagonize transcription and replication [85]. ALN is a bisphosphonate, an osteoclast inhibitor, that can target bone tumors based on its affinity for hydroxyapatite and inhibit bone destruction [86]. To establish osteosarcoma, the authors injected K7M2 osteosarcoma cells into the medullary cavity of the tibia of 20 four-week-old BALB/c female mice. The mice injected with the ALN-OXA dual-loaded hydrogel system possessed the smallest dissected tumor (weight 2.81 ± 0.26 g) compared to the other experimental groups. Additionally, the ALN-OXA group had a 60.4% tumor suppression rate, achieving the most statistically significant tumor inhibition effect. The degradation of the ALN-OXA hydrogel was found to be stable and with a degradation rate of 50% reached in approximately 27 days. Additionally, the OXA-ALN-loaded hydrogel was found to slowly release 40% of the loaded drugs within 15 days. Furthermore, histological analyses of the heart, liver, spleen, and kidneys showed no noticeable morphological changes in the osteosarcoma-bearing mice given the ALN-OXA hydrogel compared to the control group [84]. These findings clearly indicate that the co-loaded ALN-OXA hydrogel can inhibit osteosarcoma progression while limiting the toxic effects of chemotherapy.
Figure 3. (A) Scaffold degradation over 60 days with and without elastase. (B) Biocompatibility was assessed through subcutaneous injection of the gel, followed by histological analysis. (C) Release of DTX and CA4 from the hydrogels over 48 days. Reprinted with permission from [82].
Figure 3. (A) Scaffold degradation over 60 days with and without elastase. (B) Biocompatibility was assessed through subcutaneous injection of the gel, followed by histological analysis. (C) Release of DTX and CA4 from the hydrogels over 48 days. Reprinted with permission from [82].
Gels 09 00274 g003
Another study conducted by Tan et al. (2021) used Curcumin (Cur) microspheres loaded in hybrid methylcellulose hydrogels to target K7M2wt osteosarcoma cells [87]. Delayed release of Cur was observed in the hydrogels that were sustained for up to 14 days. With laser irradiation, Cur released from the hydrogels was nearly 30% within 2 days. However, there was nearly 15% release without irradiation in the same period. The slow release is a promising factor for the long-term treatment of osteosarcoma. Anti-tumor capability against K7M2wt cells was demonstrated through nearly a 75% reduction in viable cells after exposure to Cur and irradiation. There was a 50% reduction in viability when only exposed to irradiation and the hydrogel without Cur. Tumor ablation was assessed in vivo through 20 tumor-bearing female BALB/c mice. After treatment, the tumor size was the smallest (~0.5 g) in mice treated with Cur and irradiation [87]. These findings show that using Cur and irradiation is a potent combination to eradicate osteosarcoma cells in vitro and in vivo. Similarly, Ma et al. (2015) loaded doxorubicin, cisplatin, and methotrexate into a thermosensitive PLGA-PEG-PLGA hydrogel that demonstrated cytotoxicity against Saos-2 and MG-63 osteosarcoma cells in vitro and Saos-2 xenografts in vivo. The PLGA-PEG-PLGA copolymer had no cytotoxic effects against the tumor cell lines; however, the addition of the chemotherapeutic agents had a synergistic anti-tumor effect. The triple drug combination hydrogel had approximately an 80% reduction in viability in Saos-2 and MG-63 cells. This was supported by an upregulation in BAX and caspase-3 and a downregulation in Bcl-2. Additionally, the triple drug combination had the lowest tumor volume and weight in vivo when assessed in 6 male BALB/c mice. Drug release of nearly 80% of the loaded drugs was observed over 11 days in vitro; however, tumor suppression was observed for up to 16 days in vivo [88].

5.2. Bone Regeneration

The post-surgical defect present after the resection of osteosarcoma is often large and difficult to heal. Osteogenesis in in vivo models can be stimulated through the addition of scaffolds, growth factors, stem cells, and other small molecules [89]. Hydrogels can target tumor remnants in bony defects and encourage natural bone regeneration at the site of injury due to their biocompatibility, bioactivity, biodegradability, and osteoinductive properties [90]. Biomimetic hydrogels strongly resemble the ECM and therefore provide the necessary support for mesenchymal stem cells (MSC) to proliferate and differentiate into osteoblasts and ultimately regenerate lost bone [91,92].
Hydrogels have also been developed that rapidly induce bone healing, limiting the necessity of bone grafts. Zhang et al. (2018) emphasized the importance of glucocorticoids and magnesium cofactors as they induced MSCs toward osteogenic differentiation [93]. A nanocomposite hydrogel of hyaluronic acid (HA) and pamidronate (PAM) carried encapsulated human MSCs, dexamethasone phosphorylate (DexP), and magnesium cofactor to the site of bone injury to induce a positive feedback loop of bone regeneration. The Mg2+ released from the hydrogel-activated alkaline phosphatase (ALP) dephosphorylated the DexP, activating it. This accelerated the release of dexamethasone and increased osteogenesis. To test the efficacy of the hydrogel in vivo, femoral bone defects were surgically created in skeletally mature female New Zealand white rabbits, and after 8 weeks post-hydrogel implantation, the bone volume/tissue volume (BV/TV) showed a 20% significant increase with the HA-PAM-Mg-DexP hydrogel [93]. Gene expression analysis demonstrated an increase in type 1 collagen, osteocalcin, and Runx2 with the HA-PAM-Mg-DexP hydrogel. On day 7, the expression level of type 1 collagen was 2.7 compared to the control, which had 1.5. The expression of osteocalcin and Runx2 also significantly increased in comparison to the control (2.2 vs. 1, respectively).
The development of hydrogels capable of targeting residual osteosarcoma cells while encouraging non-tumor cell growth is essential for a dual-targeted system for treatment. Yu et al. (2021) demonstrated the development of Cur-loaded chitosan nanoparticles encapsulated in a methacrylated silk fibroin/hyaluronic acid (CCNPs-SF/HAMA) hydrogel, which proved to be effective in reducing the viability of osteosarcoma cells and promoting the proliferation of osteoblasts in vitro [94]. Cur, a natural polyphenol derived from turmeric Curcuma longa, has shown anti-tumor, anti-oxidant, and anti-inflammatory activities against osteosarcoma, as well as increased osteoblast proliferation and bone formation [95,96,97]. The CCNPs-SF/HAMA hydrogels loaded with Cur demonstrated that 150 μg/mL of Cur demonstrated simultaneous anti-cancer effects in MG-63 cell cultures, significantly decreasing cell viability from 100% to approximately 56% and osteoblast proliferative effects in MC3T3-E1 cell cultures, significantly increasing the cell viability from 100% to 124.5% (Figure 4) [98].
Liao et al. (2021) fabricated a methacrylated gelatin/chondroitin sulfate hydrogel with hybrid gold nanorods (GNRs) and nanohydroxyapatite (nHA) and used photothermal therapy (PTT) to eradicate the tumor remnants while simultaneously depositing MSCs that encouraged bone growth. The eradication of the tumor composed of K7M2 cells was tested in vitro [92]. PTT was performed using multiple power densities, and a significant decrease in tumor cells was noted using 0.99 W/cm3 power density as the tumor cell viability dropped from 100% to 1.2%. In an in vivo demonstration, the experimental group (surgery + hydrogel + PTT) successfully eliminated tumor remnants using a 0.99 W/cm3 power density for the PTT. Additionally, the authors showed a significant decrease in tumor volume from approximately 2500 mm3 in the control group to 50 mm3 in the experimental group, with no further recurrence of the osteosarcoma. Additionally, a significant increase in bone volume was observed with microCT and H & E staining, measuring from 3.1 mm3 in the control group up to 5.4 mm3 in the surgery + hydrogel + PTT group [92]. A summary of the studies used to treat osteosarcoma are detailed in Table 1. A schematic of hydrogels of varying composition and loaded drugs is shown in Figure 5.
Figure 4. (A) Cell viability of MG-63 osteosarcoma cells when exposed to curcumin (Cur). (B) Cell viability of MC3T3-E1 pre-osteoblasts when exposed to Cur. (C) Live and dead images of MG-63 and MC3T3-E1 cells exposed to curcumin-loaded scaffolds (+) and without (−). * p < 0.05, ** p < 0.01, *** p < 0.001. Reprinted with permission from [94].
Figure 4. (A) Cell viability of MG-63 osteosarcoma cells when exposed to curcumin (Cur). (B) Cell viability of MC3T3-E1 pre-osteoblasts when exposed to Cur. (C) Live and dead images of MG-63 and MC3T3-E1 cells exposed to curcumin-loaded scaffolds (+) and without (−). * p < 0.05, ** p < 0.01, *** p < 0.001. Reprinted with permission from [94].
Gels 09 00274 g004

6. Non-Hydrogel Scaffolds for Osteosarcoma Therapy

While the use of hydrogels has shown success in the treatment of osteosarcoma and regeneration of bone defects, there also has been much research in the development of non-hydrogel scaffolds for osteosarcoma therapy [98]. Yang et al. (2018) worked on establishing a bifunctional scaffold, one that could treat post-resection osteosarcoma remnants via drug (doxorubicin) release and PTT while simultaneously encouraging bone regeneration in the defect using human bone-marrow MSCs (hBMSC) [99]. A mesoporous calcium silicate/chitosan (MCSC) scaffold consisting of M-type ferrite particles (SrFe12O19), mesoporous calcium silicate (CaSiO3), and chitosan was fabricated. SrFe12O19 enhanced the PTT efficacy using doxorubicin (DOX) as the chemotherapeutic agent [100]. The MCSC scaffolds were developed with two different ferrite/calcium silicate ratios: 1:7 and 1:3. hBMSCs demonstrated the highest proliferation on the MCSC 1:3 scaffolds and had the highest expression of osteogenic genes, bone morphogenetic protein (BMP)-2, phosphorylated Smad1/5 and Runx2, all of which indicate osteoblast differentiation [101,102]. Further, an in vivo experiment was conducted on rat bone defect models with four experimental groups: untreated control, chitosan (CS), MCSC 1:3, and MCSC 1:7 treated. The experimental group treated with MCSC 1:3 scaffolds and DOX, synergized via PTT, had the greatest signs of tumor decrease, bone formation, and decrease in the defect area, as confirmed by microCT. The relative tumor volume was determined by a ratio at day 12 (V) and day 0 (V0). The group treated with MCSC 1:3-DOX-PTT had a V/V0 of approximately 0.4, with an approximate 85% rate of necrosis, compared to the control group with a V/V0 of approximately 3.4 with an approximate 2% rate of necrosis. The BV/TV in rats treated with MCSC 1:3 scaffolds was 57.32  ±  3.53%, which was significantly higher compared to the other groups (Figure 6) [99].
Ma et al. (2018) designed a Fe-CaSiO3 (mass %: 30% CaSiO3 and 70% Fe) compound scaffold, named 30CS, via 3D printing and demonstrated tumor killing and bone regeneration in vivo [103]. The release of Fe ions by the scaffold encourages the production of reactive oxygen species (ROS), which results in tumor cell apoptosis via DNA damage [104]. Further, the scaffolds exhibited synergistic effects with PTT to further eradicate the tumor remnants after resection. The 30CS scaffolds also provided mechanical support for rabbit bone mesenchymal stem cells (rBMSC) and encouraged bone formation in vivo. In 30 tumor-bearing nude mice, the control CaSiO3 scaffold without PTT had an approximate relative tumor volume of 8, whereas the 30CS + PTT group had an approximate relative tumor value of 0.1. The authors further assessed bone regeneration in 12 New Zealand white rabbits with critical-size bone defects. The increase in bone volume was significantly higher, with the 30CS scaffold showing a BV/TV of approximately 16 vs. 11 for the Fe scaffold [103] (Figure 6). Additional studies also showed that Si ions released from these scaffolds stimulated collagen synthesis, osteoblast proliferation, and skeletal and vascular development [105,106]. Similarly, released Ca ions enhanced osteogenesis and bone mineralization [107,108]. Overall, these experiments proved the efficacy of the 30CS scaffold in tumor treatment and bone regeneration.
Another study integrated 2D niobium carbide (Nb2C) and MXene nanosheets (NS) into 3D printed porous bioactive glass scaffolds (BGS) designed with PTT-induced destruction of osteosarcoma cells and bone regeneration by encouraging neovascularization in the defect [109] (Figure 6). Nb2C bioactive glass scaffolds (NBGS) MXene NSs are biocompatible and biodegradable materials due to their intrinsic photoresponse in the second near-infrared (NIR-II) biological window for PTT [110]. The researchers explored the in vitro cytotoxic effects of this scaffold on the human osteosarcoma cell line Saos-2. In the NBGS + NIR-treated group, 50.6% of the tumor cells underwent apoptosis, as determined through immunofluorescence. The same procedure was conducted in vivo with five female BALB/c mice. NBGS + PTT showed the greatest decrease in tumor volume, measuring 0 cm3 on day 14, compared to NBGS, showing increased tumor volume up to approximately 1500 cm3. After validating the cytotoxicity of the scaffold with PTT on osteosarcoma cells, its capacity to promote angiogenesis and induce osteogenesis was evaluated. It is already well-known that blood vessels and angiogenesis play a pivotal role in osteogenic differentiation and bone healing [111]. The researchers compared the effects of the BGS and NBGS scaffolds in promoting angiogenesis in vitro using human umbilical endothelial vein cells (HUVEC) to confirm the NBGS increased the migration capacity of HUVECs as confirmed by the detection of VEGF-B and FGF2 expression [112]. Additionally, calvarial defects were created in 24 male Sprague–Dawley rats with hBMSC-seeded NBGS and BGS scaffolds to assess bone regeneration. Significant bone regeneration was demonstrated in the NBGS group, with 47% BV/TV, compared to the BGS (30%) group. Bone regeneration was also confirmed via the expression of bone markers, COL1, OCN, and OPN genes [113,114]. Overall, the experiment demonstrated that the NBGS scaffold effectively destroyed tumor cells via PTT and prompted bone regeneration at the site of the bone defect [109].
Figure 6. Schematic overview of some preclinical studies on the use of non-hydrogel scaffolds for the treatment of osteosarcoma [99,103,109].
Figure 6. Schematic overview of some preclinical studies on the use of non-hydrogel scaffolds for the treatment of osteosarcoma [99,103,109].
Gels 09 00274 g006

7. Future Applications of Hydrogels Designed for Osteosarcoma Therapy

While hydrogels have shown promising results in treating osteosarcoma, there are still unresolved research areas. Hydrogels are often prone to failure due to poor mechanical and structural stability and are unable to maintain complex structures [115]. Even though most hydrogels are fabricated to mimic the ECM, the lack of mechanical strength prevents them from providing substantial mechanical support to cells and tissues [116]. Natural sources of hydrogels such as collagen, gelatin, alginate, and chitosan are often considered to be good candidates for drug and cell-based therapy because of their biocompatibility but have limited use due to their lack of stability and weak mechanical qualities [117]. Alginate hydrogels are a primary example, as they require an additional crosslinking step to maintain their polymer structure; they also lose mechanical strength within a very short period of time [118]. A larger degree of crosslinking has been connected to a decrease in swelling ratios and an increase in the brittleness of the hydrogels, which may further indicate mechanical instability [117]. Hydrogels have low tensile strength and, as a result, can result in premature disintegration within the targeted site. In the case of drug delivery, the large pore size coupled with the large water content of hydrogels could limit the release of hydrophobic or highly specific homogenous drugs [119].
Despite these drawbacks, hydrogels have many advantageous and potential translational applications. Hydrogels allow for efficient drug delivery because they can be administered through parenteral, nasal, ocular, and topical routes [120]. Additionally, hydrogels make it possible to prevent off-site drug exposure to provide targeted therapy [121]. The site-specific delivery of drugs also allows the target site to receive higher doses of medication at the time of administration, leading to a reduced number of doses necessary to achieve the desired therapeutic effect [121]. Currently, regenerative hydrogels are being used in the United States and European Union to provide pain relief and functional improvement and help tissue regeneration [122].
Despite the translational applications, there are still limitations to using hydrogels in clinical trials. Many hydrogel systems, such as poly(phosphazene), pluronic, and poly(N-isopropylacrylamide), have non-biodegradable and non-biocompatible characteristics that may have unintended or unknown effects [122]. The release of therapeutic agents also presents a challenge for clinical trials. For drugs that require sustained release (such as protein-based drugs), the crosslinking mesh and solute elution cannot be disrupted as the drug will not be properly released at the target site [123]. Injectable hydrogels currently comprise 26% of all clinical trials involving bulk hydrogels, but there is a challenge to create hydrogels with a viscosity low enough to be delivered through a needle and syringe [123]. Additional properties of hydrogels that must be considered for use in clinical trials involve the molecular weight of polymer chains, the density of crosslinking, and the viscosity of the solution.
Even with the possible limitations of hydrogels in clinical trials, they may help to treat osteosarcoma less invasively. Current methods of treatment for osteosarcoma involve surgical resection and adjuvant chemotherapy. Some of the biggest challenges for osteosarcoma treatment are chemoresistance and metastasis prevention [124]. Studies have shown that hydrogels can treat tumors due to their biocompatibility and porous structures [80]. Hydrogels can be loaded with tumor therapy drugs that can be inserted intravenously to replace systemic chemotherapy. In one example, hydrogels loaded with PEG-g-chitosan improved the infiltration of T lymphocytes into the gel and allowed the controlled release of these cells to the tumor site [125]. Often, tumor pathogenesis requires the use of more than one drug, and the current method of administering multiple drugs is synergistic chemotherapy. Currently, there is an effort to produce hydrogel scaffolds with stronger mechanical and biological properties [124]. For osteosarcoma treatment, this entails using materials in the gels that improve the flexibility and structure of the hydrogels [126]. Some materials under consideration are chitosan and magnesium because these are known for their mechanical properties and biocompatibility [126]. Hydroxyapatite is being studied for its ability to promote bone growth and tissue repair, but it has not been frequently used in hydrogels because it is brittle [127]. However, carbon nanofillers such as carbon nanotubes, graphene oxides, and graphene oxide-carbon nanotubes are also being studied to examine if they can reduce the brittle properties of hydroxyapatite [128].
Hydrogels allow targeted drug release and aid in reducing the adverse effects of neoadjuvant chemotherapy and anti-cancer drugs, but inherently hydrogels carry some side effects. Hydrogels made with synthetic polymers such as polylactide (PLA), poly-lactide-co-glycolide (PLGA), polyglycolide (PGA), poly-(D,L-lactic acid) (PDLLA), polycaprolactone (PCL), poly-ethylene-glycol (PEG), poly(vinyl alcohol) (PVA), poly(N-isopropylacrylamide) (PNIPAM), and polyacrylamide (PAM) can induce an immune response in the body or expose toxicity to the cells [118]. In addition, many of these polymeric-based biomaterials cannot be incorporated into host tissues and are thus toxic foreign materials that must be degraded in the body. Hydrogels made with natural polymers do not have the same problems and do not cause immune or toxic reactions [118]. Thus, there is an emphasis on using natural hydrogels because of their non-toxic, biocompatible, and biodegradable properties. There are fewer long-term side effects caused by hydrogels because of their ability to degrade in the body after the release of the desired drug(s) [129].
Due to the convenience of 3D printing, the synthesis and fabrication of hydrogels are both cost-effective and time-efficient [59,130]. 3D printing also allows for complex crosslinking and structure formation without requiring long periods of time and costly materials. In addition, hydrogels are generally composed of easily accessible compounds such as hyaluronic acid, alginate, fibrin, collagen, gelatin, and chitosan, which also help to reduce overall production costs [59]. The major technical drawback of fabricating hydrogels is creating a stable structure despite the poor mechanical properties of most hydrogels [131]. Further research is needed to determine the best structure and material to use for maximizing the therapeutic potential of hydrogels, especially for osteosarcoma.

8. Hydrogel Use in Clinical Trials on Osteosarcoma

Examining clinical trial registries is fundamental for acquiring an approximate overview of the progress of ongoing research efforts. Our group performed a search in the National Institutes of Health’s (NIH) ClinicalTrials.gov registry (access dates up to and including 1 March 2023) for all registered clinical trials, using the following keywords: “hydrogels” and “cancer treatment.” No exemption criteria were applied to filter the initial results, and the search included both studies marked as completed and as actively enrolling. The search brought up 57 clinical trials in the area of hydrogels used in various cancer therapy studies, mostly located in Europe (16) and the United States (24). There were 2 phase 1 clinical trials, 11 phase 2 trials, 14 phase 3 trials, and 2 phase 4 clinical trials. The search included 5 studies based on a young population (0–17), 10 studies based on adults (18–64), and the remaining 42 studies focused on subjects 65 and older. However, most of the above-mentioned clinical trials investigate therapeutic options for conditions such as gastrointestinal cancer, carcinoma, or male genital cancers. None of the queried studies partake in the topic of bone malignancies such as osteosarcoma.
We also searched with the keywords “hydrogel” and “bone recovery”, with no exception criteria included. The search resulted in 19 completed or actively enrolling clinical trials that exclusively target the adult population. Overall, the clinical trials focus on therapeutic methods for conditions such as bone resorption, tooth loss, periodontitis, chondral defect, or articular cartilage defect, with no significant mention of bone malignancies. Despite the many preclinical studies investigating localized hydrogel therapy for osteosarcoma treatment as a means to replace systemic chemotherapy, the need for highly translational trials remains unmet. Further, the reduced number of ongoing clinical trials proves a significant gap between the successful implementation of a preclinical strategy that can translate into the rigor, limitations, and regulations of clinical trials.

9. Conclusions

While numerous advancements have been made clinically for treating and managing osteosarcoma, this tumor remains one of the most devastating bone cancers. Surgical management with chemotherapy has proven to be effective in preventing the recurrence of osteosarcoma; however, patient responses vary. The treatment of osteosarcoma with non-surgical management has been explored both in vitro and in vivo through the use of hydrogels loaded with chemotherapeutic drugs to assess tumor regression and subsequent bone regeneration. Synergistic scaffolds that can display cancer cytotoxicity while promoting bone regeneration of the defect are superior in treatment due to their dual-targeted approach. However, such biomaterials have not yet reached the market and are still performed in laboratory testing to fully assess the side-effect profile and to optimize successful functional parameters. The future applications of hydrogels for osteosarcoma treatment should emphasize in vivo revascularization of bone defects and stimulate neo-osteogenesis from post-surgical resection or direct chemotherapy-targeted osteosarcoma regression. Currently, there is a lack of data supporting increased neovascularization of bone, while there is data supporting bone formation. Furthermore, advancements in hydrogel-based therapy with novel stimuli-responsive hydrogels can pose a breakthrough in potential treatments that can be adjusted for tumor microenvironments to rapidly eradicate osteosarcoma cells without many side effects. Lastly, clinical trials are needed to potentially address the translational applications of the hydrogel’s dual-targeted approach.

Author Contributions

Conceptualization, S.T. and A.G.; Literature search, I.R., A.P. and E.S.; writing—original draft preparation, S.T., A.G., I.R., A.P., E.S. and M.H.; writing—review and editing, M.H., E.P. and A.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sadykova, L.R.; Ntekim, A.I.; Muyangwa-Semenova, M.; Rutland, C.S.; Jeyapalan, J.N.; Blatt, N.; Rizvanov, A.A. Epidemiology and risk factors of osteosarcoma. Cancer Investig. 2020, 38, 259–269. [Google Scholar] [CrossRef] [PubMed]
  2. Freedman, I.G.; Dowd, H.N.; Dhodapkar, M.M.; Halperin, S.J.; Grauer, J.N. Second Primary Malignancies of the Bones and Joints: More Common than Expected in Osteosarcoma Patients. JAAOS Glob. Res. Rev. 2023, 7, e22. [Google Scholar] [CrossRef] [PubMed]
  3. Czarnecka, A.M.; Synoradzki, K.; Firlej, W.; Bartnik, E.; Sobczuk, P.; Fiedorowicz, M.; Grieb, P.; Rutkowski, P. Molecular biology of osteosarcoma. Cancers 2020, 12, 2130. [Google Scholar] [CrossRef] [PubMed]
  4. Cole, S.; Gianferante, D.M.; Zhu, B.; Mirabello, L. Osteosarcoma: A surveillance, epidemiology, and end results program-based analysis from 1975 to 2017. Cancer 2022, 128, 2107–2118. [Google Scholar] [CrossRef]
  5. Zhang, J.; Tan, Y.; Shou, Y.; Fang, L.; Li, X.; Lai, J. The relationship between adjuvant radiotherapy and survival of osteosarcoma: A case-control study. Biotechnol. Genet. Eng. Rev. 2023, 1–14. [Google Scholar] [CrossRef]
  6. Li, X.; Zhang, Y.; Wan, S.; Li, H.; Li, D.; Xia, J.; Yuan, Z.; Ren, M.; Yu, S.; Li, S. A comparative study between limb-salvage and amputation for treating osteosarcoma. J. Bone Oncol. 2016, 5, 15–21. [Google Scholar] [CrossRef] [Green Version]
  7. Han, G.; Bi, W.-Z.; Xu, M.; Jia, J.-P.; Wang, Y. Amputation versus limb-salvage surgery in patients with osteosarcoma: A meta-analysis. World J. Surg. 2016, 40, 2016–2027. [Google Scholar] [CrossRef]
  8. Ayerza, M.A.; Farfalli, G.L.; Aponte-Tinao, L.; Luis Muscolo, D. Does increased rate of limb-sparing surgery affect survival in osteosarcoma? Clin. Orthop. Relat. Res. 2010, 468, 2854–2859. [Google Scholar] [CrossRef] [Green Version]
  9. Papakonstantinou, E.; Stamatopoulos, A.; Athanasiadis, D.I.; Kenanidis, E.; Potoupnis, M.; Haidich, A.-B.; Tsiridis, E. Limb-salvage surgery offers better five-year survival rate than amputation in patients with limb osteosarcoma treated with neoadjuvant chemotherapy. A systematic review and meta-analysis. J. Bone Oncol. 2020, 25, 100319. [Google Scholar] [CrossRef]
  10. Xu, M.; Wang, Z.; Yu, X.C.; Lin, J.H.; Hu, Y.C. Guideline for limb-salvage treatment of osteosarcoma. Orthop. Surg. 2020, 12, 1021–1029. [Google Scholar] [CrossRef]
  11. Gil, S.; Fernandez-Pineda, I.; Rao, B.; Neel, M.D.; Baker, J.N.; Wu, H.; Wu, J.; Anghelescu, D.L. Role of amputation in improving mobility, pain outcomes, and emotional and psychological well-being in children with metastatic osteosarcoma. Am. J. Hosp. Palliat. Med. 2019, 36, 105–110. [Google Scholar] [CrossRef] [PubMed]
  12. Zhong, W.; Wu, Z.; Yuan, Y.; Luo, W. Meta-analysis of the prognosis after surgical treatment of osteosarcoma complicated by pathologic fracture. Am. J. Transl. Res. 2022, 14, 2580. [Google Scholar] [PubMed]
  13. Tang, L.; Liu, B. Lung and bone metastases patterns in osteosarcoma: Chemotherapy improves overall survival. Medicine 2023, 102, e32692. [Google Scholar] [CrossRef]
  14. Rickel, K.; Fang, F.; Tao, J. Molecular genetics of osteosarcoma. Bone 2017, 102, 69–79. [Google Scholar] [CrossRef]
  15. Perry, J.A.; Kiezun, A.; Tonzi, P.; Van Allen, E.M.; Carter, S.L.; Baca, S.C.; Cowley, G.S.; Bhatt, A.S.; Rheinbay, E.; Pedamallu, C.S. Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma. Proc. Natl. Acad. Sci. USA 2014, 111, E5564–E5573. [Google Scholar] [CrossRef] [Green Version]
  16. Synoradzki, K.J.; Bartnik, E.; Czarnecka, A.M.; Fiedorowicz, M.; Firlej, W.; Brodziak, A.; Stasinska, A.; Rutkowski, P.; Grieb, P. TP53 in Biology and Treatment of Osteosarcoma. Cancers 2021, 13, 4284. [Google Scholar] [CrossRef] [PubMed]
  17. Ray-Coquard, I.; Blay, J.-Y.; Italiano, A.; Le Cesne, A.; Penel, N.; Zhi, J.; Heil, F.; Rueger, R.; Graves, B.; Ding, M. Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: An exploratory proof-of-mechanism study. Lancet Oncol. 2012, 13, 1133–1140. [Google Scholar] [CrossRef]
  18. Zhang, B.; Zhang, Y.; Li, R.; Li, J.; Lu, X.; Zhang, Y. The efficacy and safety comparison of first-line chemotherapeutic agents (high-dose methotrexate, doxorubicin, cisplatin, and ifosfamide) for osteosarcoma: A network meta-analysis. J. Orthop. Surg. Res. 2020, 15, 51. [Google Scholar] [CrossRef] [Green Version]
  19. Yu, D.; Zhang, S.; Feng, A.; Xu, D.; Zhu, Q.; Mao, Y.; Zhao, Y.; Lv, Y.; Han, C.; Liu, R. Methotrexate, doxorubicin, and cisplatinum regimen is still the preferred option for osteosarcoma chemotherapy: A meta-analysis and clinical observation. Medicine 2019, 98, e15582. [Google Scholar] [CrossRef]
  20. Shan, H.; Li, K.; Zhao, D.; Chi, C.; Tan, Q.; Wang, X.; Yu, J.; Piao, M. Locally controlled release of methotrexate and alendronate by thermo-sensitive hydrogels for synergistic inhibition of osteosarcoma progression. Front. Pharmacol. 2020, 11, 573. [Google Scholar] [CrossRef]
  21. Wei, Z.; Volkova, E.; Blatchley, M.R.; Gerecht, S. Hydrogel vehicles for sequential delivery of protein drugs to promote vascular regeneration. Adv. Drug Deliv. Rev. 2019, 149, 95–106. [Google Scholar] [CrossRef] [PubMed]
  22. Liu, L.; Gao, Q.; Lu, X.; Zhou, H. In situ forming hydrogels based on chitosan for drug delivery and tissue regeneration. Asian J. Pharm. Sci. 2016, 11, 673–683. [Google Scholar] [CrossRef] [Green Version]
  23. Choi, J.; Curtis, S.J.; Roy, D.M.; Flesken-Nikitin, A.; Nikitin, A.Y. Local mesenchymal stem/progenitor cells are a preferential target for initiation of adult soft tissue sarcomas associated with p53 and Rb deficiency. Am. J. Pathol. 2010, 177, 2645–2658. [Google Scholar] [CrossRef] [PubMed]
  24. Hameed, M.; Mandelker, D. Tumor syndromes predisposing to osteosarcoma. Adv. Anat. Pathol. 2018, 25, 217. [Google Scholar] [CrossRef] [PubMed]
  25. Jeyarani, G.; Jayaraman, D.; Menon, G.; Harshavardhaan, J.G.; Rajendiran, S.; Murali, A. Telangiectatic Osteosarcoma in a Young Child–A Case Report and Review of the Literature. J. Orthop. Case Rep. 2021, 11, 72. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, R.K.; Chu, W.C.; Leung, J.H.; Cheng, F.W.; Li, C. Pathological fracture as the presenting feature in pediatric osteosarcoma. Pediatr. Blood Cancer 2013, 60, 1118–1121. [Google Scholar] [CrossRef]
  27. Barani, M.; Mukhtar, M.; Rahdar, A.; Sargazi, S.; Pandey, S.; Kang, M. Recent advances in nanotechnology-based diagnosis and treatments of human osteosarcoma. Biosensors 2021, 11, 55. [Google Scholar] [CrossRef]
  28. Hu, B.; Liu, Y.; Cheng, L.; Li, W.; Cao, X. SPECT/CT imaging of retroperitoneal extraskeletal osteosarcoma. Clin. Nucl. Med. 2014, 39, 200–202. [Google Scholar] [CrossRef]
  29. Zhou, H.; Yi, W.; Li, A.; Wang, B.; Ding, Q.; Xue, L.; Zeng, X.; Feng, Y.; Li, Q.; Wang, T. Specific Small-Molecule NIR-II Fluorescence Imaging of Osteosarcoma and Lung Metastasis. Adv. Healthc. Mater. 2020, 9, 1901224. [Google Scholar] [CrossRef]
  30. Beird, H.C.; Bielack, S.S.; Flanagan, A.M.; Gill, J.; Heymann, D.; Janeway, K.A.; Livingston, J.A.; Roberts, R.D.; Strauss, S.J.; Gorlick, R. Osteosarcoma. Nat. Rev. Dis. Prim. 2022, 8, 77. [Google Scholar] [CrossRef]
  31. Hu-Lieskovan, S.; Robert, L.; Moreno, B.H.; Ribas, A. Combining targeted therapy with immunotherapy in BRAF-mutant melanoma: Promise and challenges. J. Clin. Oncol. 2014, 32, 2248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Naylor, E.C.; Desani, J.K.; Chung, P.K. Targeted therapy and immunotherapy for lung cancer. Surg. Oncol. Clin. 2016, 25, 601–609. [Google Scholar] [CrossRef]
  33. Naoum, G.E.; Morkos, M.; Kim, B.; Arafat, W. Novel targeted therapies and immunotherapy for advanced thyroid cancers. Mol. Cancer 2018, 17, 51. [Google Scholar] [CrossRef] [PubMed]
  34. Hu, Z.; Wen, S.; Huo, Z.; Wang, Q.; Zhao, J.; Wang, Z.; Chen, Y.; Zhang, L.; Zhou, F.; Guo, Z. Current Status and Prospects of Targeted Therapy for Osteosarcoma. Cells 2022, 11, 3507. [Google Scholar] [CrossRef]
  35. Italiano, A.; Mir, O.; Mathoulin-Pelissier, S.; Penel, N.; Piperno-Neumann, S.; Bompas, E.; Chevreau, C.; Duffaud, F.; Entz-Werlé, N.; Saada, E. Cabozantinib in patients with advanced Ewing sarcoma or osteosarcoma (CABONE): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 446–455. [Google Scholar] [CrossRef]
  36. Gaspar, N.; Casanova, M.; Sirvent, F.J.B.; Venkatramani, R.; Morland, B.; Gambart, M.; Thebaud, E.; Strauss, S.J.; Locatelli, F.; Melcon, S.G. Single-agent expansion cohort of lenvatinib (LEN) and combination dose-finding cohort of LEN+ etoposide (ETP)+ ifosfamide (IFM) in patients (pts) aged 2 to ≤25 years with relapsed/refractory osteosarcoma (OS). J. Clin. Oncol. 2018, 36, 11527. [Google Scholar] [CrossRef]
  37. Gaspar, N.; Campbell-Hewson, Q.; Melcon, S.G.; Locatelli, F.; Venkatramani, R.; Hecker-Nolting, S.; Gambart, M.; Bautista, F.; Thebaud, E.; Aerts, I. Phase I/II study of single-agent lenvatinib in children and adolescents with refractory or relapsed solid malignancies and young adults with osteosarcoma (ITCC-050)☆. ESMO Open 2021, 6, 100250. [Google Scholar] [CrossRef]
  38. Duffaud, F.; Mir, O.; Boudou-Rouquette, P.; Piperno-Neumann, S.; Penel, N.; Bompas, E.; Delcambre, C.; Kalbacher, E.; Italiano, A.; Collard, O. Efficacy and safety of regorafenib in adult patients with metastatic osteosarcoma: A non-comparative, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2019, 20, 120–133. [Google Scholar] [CrossRef]
  39. Davis, L.E.; Bolejack, V.; Ryan, C.W.; Ganjoo, K.N.; Loggers, E.T.; Chawla, S.; Agulnik, M.; Livingston, M.B.; Reed, D.; Keedy, V. Randomized double-blind phase II study of regorafenib in patients with metastatic osteosarcoma. J. Clin. Oncol. 2019, 37, 1424. [Google Scholar] [CrossRef]
  40. Newick, K.; O’Brien, S.; Moon, E.; Albelda, S.M. CAR T cell therapy for solid tumors. Annu. Rev. Med. 2017, 68, 139–152. [Google Scholar] [CrossRef]
  41. Köksal, H.; Müller, E.; Inderberg, E.M.; Bruland, Ø.; Wälchli, S. Treating osteosarcoma with CAR T cells. Scand. J. Immunol. 2019, 89, e12741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Long, A.H.; Highfill, S.L.; Cui, Y.; Smith, J.P.; Walker, A.J.; Ramakrishna, S.; El-Etriby, R.; Galli, S.; Tsokos, M.G.; Orentas, R.J. Reduction of MDSCs with All-trans Retinoic Acid Improves CAR Therapy Efficacy for SarcomasMDSCs Limit GD2-CAR T-cell Therapy. Cancer Immunol. Res. 2016, 4, 869–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yu, A.L.; Gilman, A.L.; Ozkaynak, M.F.; London, W.B.; Kreissman, S.G.; Chen, H.X.; Smith, M.; Anderson, B.; Villablanca, J.G.; Matthay, K.K. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N. Engl. J. Med. 2010, 363, 1324–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chulanetra, M.; Morchang, A.; Sayour, E.; Eldjerou, L.; Milner, R.; Lagmay, J.; Cascio, M.; Stover, B.; Slayton, W.; Chaicumpa, W. GD2 chimeric antigen receptor modified T cells in synergy with sub-toxic level of doxorubicin targeting osteosarcomas. Am. J. Cancer Res. 2020, 10, 674. [Google Scholar]
  45. Hoffman, A.S. Hydrogels for biomedical applications. Adv. Drug Deliv. Rev. 2012, 64, 18–23. [Google Scholar] [CrossRef]
  46. Sánchez-Cid, P.; Jiménez-Rosado, M.; Rubio-Valle, J.F.; Romero, A.; Ostos, F.J.; Rafii-El-Idrissi Benhnia, M.; Perez-Puyana, V. Biocompatible and Thermoresistant Hydrogels Based on Collagen and Chitosan. Polymers 2022, 14, 272. [Google Scholar] [CrossRef] [PubMed]
  47. Peppas, N.A.; Khare, A.R. Preparation, structure and diffusional behavior of hydrogels in controlled release. Adv. Drug Deliv. Rev. 1993, 11, 1–35. [Google Scholar] [CrossRef] [Green Version]
  48. Tharakan, S.; Khondkar, S.; Lee, S.; Ahn, S.; Mathew, C.; Gresita, A.; Hadjiargyrou, M.; Ilyas, A. 3D Printed Osteoblast–Alginate/Collagen Hydrogels Promote Survival, Proliferation and Mineralization at Low Doses of Strontium Calcium Polyphosphate. Pharmaceutics 2022, 15, 11. [Google Scholar] [CrossRef] [PubMed]
  49. Parhi, R. Cross-linked hydrogel for pharmaceutical applications: A review. Adv. Pharm. Bull. 2017, 7, 515–530. [Google Scholar] [CrossRef]
  50. Berger, J.; Reist, M.; Mayer, J.M.; Felt, O.; Gurny, R. Structure and interactions in chitosan hydrogels formed by complexation or aggregation for biomedical applications. Eur. J. Pharm. Biopharm. 2004, 57, 35–52. [Google Scholar] [CrossRef]
  51. Ali, F.; Khan, I.; Chen, J.; Akhtar, K.; Bakhsh, E.M.; Khan, S.B. Emerging fabrication strategies of hydrogels and its applications. Gels 2022, 8, 205. [Google Scholar] [CrossRef]
  52. Shim, W.S.; Kim, J.-H.; Kim, K.; Kim, Y.-S.; Park, R.-W.; Kim, I.-S.; Kwon, I.C.; Lee, D.S. pH-and temperature-sensitive, injectable, biodegradable block copolymer hydrogels as carriers for paclitaxel. Int. J. Pharm. 2007, 331, 11–18. [Google Scholar] [CrossRef] [PubMed]
  53. Ono, K.; Saito, Y.; Yura, H.; Ishikawa, K.; Kurita, A.; Akaike, T.; Ishihara, M. Photocrosslinkable chitosan as a biological adhesive. J. Biomed. Mater. Res. Off. J. Soc. Biomater. Jpn. Soc. Biomater. 2000, 49, 289–295. [Google Scholar] [CrossRef]
  54. Elluru, M.; Ma, H.; Hadjiargyrou, M.; Hsiao, B.S.; Chu, B. Synthesis and characterization of biocompatible hydrogel using Pluronics-based block copolymers. Polymer 2013, 54, 2088–2095. [Google Scholar] [CrossRef]
  55. Łukaszczyk, J.; Śmiga, M.; Jaszcz, K.; Adler, H.J.P.; Jähne, E.; Kaczmarek, M. Evaluation of oligo (ethylene glycol) dimethacrylates effects on the properties of new biodegradable bone cement compositions. Macromol. Biosci. 2005, 5, 64–69. [Google Scholar] [CrossRef]
  56. Ren, K.; He, C.; Cheng, Y.; Li, G.; Chen, X. Injectable enzymatically crosslinked hydrogels based on a poly (l-glutamic acid) graft copolymer. Polym. Chem. 2014, 5, 5069–5076. [Google Scholar] [CrossRef]
  57. Teixeira, L.S.M.; Feijen, J.; van Blitterswijk, C.A.; Dijkstra, P.J.; Karperien, M. Enzyme-catalyzed crosslinkable hydrogels: Emerging strategies for tissue engineering. Biomaterials 2012, 33, 1281–1290. [Google Scholar] [CrossRef]
  58. Irvine, S.A.; Venkatraman, S.S. Bioprinting and differentiation of stem cells. Molecules 2016, 21, 1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Tharakan, S.; Khondkar, S.; Ilyas, A. Bioprinting of Stem Cells in Multimaterial Scaffolds and Their Applications in Bone Tissue Engineering. Sensors 2021, 21, 7477. [Google Scholar] [CrossRef]
  60. Naahidi, S.; Jafari, M.; Logan, M.; Wang, Y.; Yuan, Y.; Bae, H.; Dixon, B.; Chen, P. Biocompatibility of hydrogel-based scaffolds for tissue engineering applications. Biotechnol. Adv. 2017, 35, 530–544. [Google Scholar] [CrossRef]
  61. Chan, B.; Leong, K. Scaffolding in tissue engineering: General approaches and tissue-specific considerations. Eur. Spine J. 2008, 17, 467–479. [Google Scholar] [CrossRef] [Green Version]
  62. Lee, C.H.; Singla, A.; Lee, Y. Biomedical applications of collagen. Int. J. Pharm. 2001, 221, 1–22. [Google Scholar] [CrossRef] [PubMed]
  63. Eom, S.; Park, S.M.; Hong, H.; Kwon, J.; Oh, S.-R.; Kim, J.; Kim, D.S. Hydrogel-assisted electrospinning for fabrication of a 3D complex tailored nanofiber macrostructure. ACS Appl. Mater. Interfaces 2020, 12, 51212–51224. [Google Scholar] [CrossRef] [PubMed]
  64. Chiu, J.B.; Luu, Y.K.; Fang, D.; Hsiao, B.S.; Chu, B.; Hadjiargyrou, M. Electrospun nanofibrous scaffolds for biomedical applications. J. Biomed. Nanotechnol. 2005, 1, 115–132. [Google Scholar] [CrossRef]
  65. Elzoghby, A.O. Gelatin-based nanoparticles as drug and gene delivery systems: Reviewing three decades of research. J. Control. Release 2013, 172, 1075–1091. [Google Scholar] [CrossRef]
  66. Panduranga Rao, K. Recent developments of collagen-based materials for medical applications and drug delivery systems. J. Biomater. Sci. Polym. Ed. 1996, 7, 623–645. [Google Scholar] [CrossRef]
  67. Cecen, B.; Kozaci, D.; Yuksel, M.; Erdemli, D.; Bagriyanik, A.; Havitcioglu, H. Biocompatibility of MG-63 cells on collagen, poly-L-lactic acid, hydroxyapatite scaffolds with different parameters. J. Appl. Biomater. Funct. Mater. 2015, 13, 10–16. [Google Scholar] [CrossRef]
  68. Zong, C.; Qian, X.; Tang, Z.; Hu, Q.; Chen, J.; Gao, C.; Tang, R.; Tong, X.; Wang, J. Biocompatibility and bone-repairing effects: Comparison between porous poly-lactic-co-glycolic acid and nano-hydroxyapatite/poly (lactic acid) scaffolds. J. Biomed. Nanotechnol. 2014, 10, 1091–1104. [Google Scholar] [CrossRef]
  69. Kolewe, K.W.; Kalasin, S.; Shave, M.; Schiffman, J.D.; Santore, M.M. Mechanical properties and concentrations of poly (ethylene glycol) in hydrogels and brushes direct the surface transport of Staphylococcus aureus. ACS Appl. Mater. Interfaces 2018, 11, 320–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. 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]
  71. Bordbar-Khiabani, A.; Gasik, M. Smart hydrogels for advanced drug delivery systems. Int. J. Mol. Sci. 2022, 23, 3665. [Google Scholar] [CrossRef] [PubMed]
  72. Kopač, T.; Krajnc, M.; Ručigaj, A. A mathematical model for pH-responsive ionically crosslinked TEMPO nanocellulose hydrogel design in drug delivery systems. Int. J. Biol. Macromol. 2021, 168, 695–707. [Google Scholar] [CrossRef]
  73. Askari, E.; Seyfoori, A.; Amereh, M.; Gharaie, S.S.; Ghazali, H.S.; Ghazali, Z.S.; Khunjush, B.; Akbari, M. Stimuli-responsive hydrogels for local post-surgical drug delivery. Gels 2020, 6, 14. [Google Scholar] [CrossRef] [PubMed]
  74. Davoodi, P.; Lee, L.Y.; Xu, Q.; Sunil, V.; Sun, Y.; Soh, S.; Wang, C.-H. Drug delivery systems for programmed and on-demand release. Adv. Drug Deliv. Rev. 2018, 132, 104–138. [Google Scholar] [CrossRef] [PubMed]
  75. Rafael, D.; Melendres, M.M.R.; Andrade, F.; Montero, S.; Martinez-Trucharte, F.; Vilar-Hernandez, M.; Durán-Lara, E.F.; Schwartz Jr, S.; Abasolo, I. Thermo-responsive hydrogels for cancer local therapy: Challenges and state-of-art. Int. J. Pharm. 2021, 606, 120954. [Google Scholar] [CrossRef]
  76. SMART—Servier Medical Art. Available online: https://smart.servier.com/ (accessed on 21 March 2023).
  77. Oliva, N.; Conde, J.; Wang, K.; Artzi, N. Designing hydrogels for on-demand therapy. Acc. Chem. Res. 2017, 50, 669–679. [Google Scholar] [CrossRef]
  78. Bae, Y.H.; Park, K. Targeted drug delivery to tumors: Myths, reality and possibility. J. Control. Release 2011, 153, 198. [Google Scholar] [CrossRef] [Green Version]
  79. Melnyk, Y.; Stetsyshyn, Y.; Skorokhoda, V.; Nastishin, Y. Polyvinylpyrrolidone-graft-poly (2-hydroxyethylmethacrylate) hydrogel membranes for encapsulated forms of drugs. J. Polym. Res. 2020, 27, 354. [Google Scholar] [CrossRef]
  80. Zhang, J.; Tian, H.; Wu, H.; Feng, N.; Zuo, J. Recent advances in hydrogels-based osteosarcoma therapy. Front. Bioeng. Biotechnol. 2022, 10, 1938. [Google Scholar]
  81. Wu, W.; Dai, Y.; Liu, H.; Cheng, R.; Ni, Q.; Ye, T.; Cui, W. Local release of gemcitabine via in situ UV-crosslinked lipid-strengthened hydrogel for inhibiting osteosarcoma. Drug Deliv. 2018, 25, 1642–1651. [Google Scholar] [CrossRef] [Green Version]
  82. Zheng, Y.; Cheng, Y.; Chen, J.; Ding, J.; Li, M.; Li, C.; Wang, J.-C.; Chen, X. Injectable hydrogel–microsphere construct with sequential degradation for locally synergistic chemotherapy. ACS Appl. Mater. Interfaces 2017, 9, 3487–3496. [Google Scholar] [CrossRef]
  83. Pérez-Pérez, M.-J.S.; Priego, E.-M.; Bueno, O.; Martins, M.S.; Canela, M.-D.; Liekens, S. Blocking blood flow to solid tumors by destabilizing tubulin: An approach to targeting tumor growth. J. Med. Chem. 2016, 59, 8685–8711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Sun, Y.; Li, K.; Li, C.; Zhang, Y.; Zhao, D. Thermogel delivers oxaliplatin and alendronate in situ for synergistic osteosarcoma therapy. Front. Bioeng. Biotechnol. 2020, 8, 573962. [Google Scholar] [CrossRef] [PubMed]
  85. Hato, S.V.; Khong, A.; de Vries, I.J.M.; Lesterhuis, W.J. Molecular pathways: The immunogenic effects of platinum-based chemotherapeutics. Clin. Cancer Res. 2014, 20, 2831–2837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Blanco, E.; Kessinger, C.W.; Sumer, B.D.; Gao, J. Multifunctional micellar nanomedicine for cancer therapy. Exp. Biol. Med. 2009, 234, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Tan, B.; Wu, Y.; Wu, Y.; Shi, K.; Han, R.; Li, Y.; Qian, Z.; Liao, J. Curcumin-microsphere/IR820 hybrid bifunctional hydrogels for in situ osteosarcoma chemo-co-thermal therapy and bone reconstruction. ACS Appl. Mater. Interfaces 2021, 13, 31542–31553. [Google Scholar] [CrossRef]
  88. Ma, H.; He, C.; Cheng, Y.; Yang, Z.; Zang, J.; Liu, J.; Chen, X. Localized co-delivery of doxorubicin, cisplatin, and methotrexate by thermosensitive hydrogels for enhanced osteosarcoma treatment. ACS Appl. Mater. Interfaces 2015, 7, 27040–27048. [Google Scholar] [CrossRef]
  89. Velasco, M.A.; Narváez-Tovar, C.A.; Garzón-Alvarado, D.A. Design, materials, and mechanobiology of biodegradable scaffolds for bone tissue engineering. BioMed Res. Int. 2015, 2015, 729076. [Google Scholar] [CrossRef] [Green Version]
  90. Feng, Q.; Xu, J.; Zhang, K.; Yao, H.; Zheng, N.; Zheng, L.; Wang, J.; Wei, K.; Xiao, X.; Qin, L. Dynamic and cell-infiltratable hydrogels as injectable carrier of therapeutic cells and drugs for treating challenging bone defects. ACS Cent. Sci. 2019, 5, 440–450. [Google Scholar] [CrossRef] [Green Version]
  91. Xu, B.; Ye, J.; Yuan, F.-Z.; Zhang, J.-Y.; Chen, Y.-R.; Fan, B.-S.; Jiang, D.; Jiang, W.-B.; Wang, X.; Yu, J.-K. Advances of stem cell-laden hydrogels with biomimetic microenvironment for osteochondral repair. Front. Bioeng. Biotechnol. 2020, 8, 247. [Google Scholar] [CrossRef] [Green Version]
  92. Liao, J.; Shi, K.; Jia, Y.; Wu, Y.; Qian, Z. Gold nanorods and nanohydroxyapatite hybrid hydrogel for preventing bone tumor recurrence via postoperative photothermal therapy and bone regeneration promotion. Bioact. Mater. 2021, 6, 2221–2230. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, K.; Jia, Z.; Yang, B.; Feng, Q.; Xu, X.; Yuan, W.; Li, X.; Chen, X.; Duan, L.; Wang, D. Adaptable hydrogels mediate cofactor-assisted activation of biomarker-responsive drug delivery via positive feedback for enhanced tissue regeneration. Adv. Sci. 2018, 5, 1800875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Yu, Q.; Meng, Z.; Liu, Y.; Li, Z.; Sun, X.; Zhao, Z. Photocuring hyaluronic acid/silk fibroin hydrogel containing curcumin loaded CHITOSAN nanoparticles for the treatment of MG-63 cells and ME3T3-E1 cells. Polymers 2021, 13, 2302. [Google Scholar] [CrossRef] [PubMed]
  95. Zahedipour, F.; Bolourinezhad, M.; Teng, Y.; Sahebkar, A. The multifaceted therapeutic mechanisms of curcumin in osteosarcoma: State-of-the-art. J. Oncol. 2021, 2021, 3006853. [Google Scholar] [CrossRef] [PubMed]
  96. Jiang, Q.; Lei, Y.H.; Krishnadath, D.C.; Zhu, B.Y.; Zhou, X.W. Curcumin regulates EZH2/Wnt/β-Catenin pathway in the mandible and femur of ovariectomized osteoporosis rats. Kaohsiung J. Med. Sci. 2021, 37, 513–519. [Google Scholar] [CrossRef]
  97. Das, R.K.; Kasoju, N.; Bora, U. Encapsulation of curcumin in alginate-chitosan-pluronic composite nanoparticles for delivery to cancer cells. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 153–160. [Google Scholar] [CrossRef]
  98. Liao, J.; Han, R.; Wu, Y.; Qian, Z. Review of a new bone tumor therapy strategy based on bifunctional biomaterials. Bone Res. 2021, 9, 18. [Google Scholar] [CrossRef]
  99. Yang, F.; Lu, J.; Ke, Q.; Peng, X.; Guo, Y.; Xie, X. Magnetic mesoporous calcium sillicate/chitosan porous scaffolds for enhanced bone regeneration and photothermal-chemotherapy of osteosarcoma. Sci. Rep. 2018, 8, 7345. [Google Scholar] [CrossRef] [Green Version]
  100. Zi, Z.; Sun, Y.; Zhu, X.; Yang, Z.; Song, W. Structural and magnetic properties of SrFe12O19 hexaferrite synthesized by a modified chemical co-precipitation method. J. Magn. Magn. Mater. 2008, 320, 2746–2751. [Google Scholar] [CrossRef]
  101. Hartung, A.; Bitton-Worms, K.; Rechtman, M.M.; Wenzel, V.; Boergermann, J.H.; Hassel, S.; Henis, Y.I.; Knaus, P. Different routes of bone morphogenic protein (BMP) receptor endocytosis influence BMP signaling. Mol. Cell Biol. 2006, 26, 7791–7805. [Google Scholar] [CrossRef] [Green Version]
  102. Lee, J.S.; Kim, M.E.; Seon, J.K.; Kang, J.Y.; Yoon, T.R.; Park, Y.-D.; Kim, H.K. Bone-forming peptide-3 induces osteogenic differentiation of bone marrow stromal cells via regulation of the ERK1/2 and Smad1/5/8 pathways. Stem Cell Res. 2018, 26, 28–35. [Google Scholar] [CrossRef] [PubMed]
  103. Ma, H.; Li, T.; Huan, Z.; Zhang, M.; Yang, Z.; Wang, J.; Chang, J.; Wu, C. 3D printing of high-strength bioscaffolds for the synergistic treatment of bone cancer. NPG Asia Mater. 2018, 10, 31–44. [Google Scholar] [CrossRef] [Green Version]
  104. Gao, L.; Hua, W.; Tian, L.; Zhou, X.; Wang, D.; Yang, Y.; Ni, G. Molecular Mechanism of Ferroptosis in Orthopedic Diseases. Cells 2022, 11, 2979. [Google Scholar] [CrossRef]
  105. Wang, C.; Lin, K.; Chang, J.; Sun, J. Osteogenesis and angiogenesis induced by porous β-CaSiO3/PDLGA composite scaffold via activation of AMPK/ERK1/2 and PI3K/Akt pathways. Biomaterials 2013, 34, 64–77. [Google Scholar] [CrossRef] [PubMed]
  106. Valerio, P.; Pereira, M.M.; Goes, A.M.; Leite, M.F. The effect of ionic products from bioactive glass dissolution on osteoblast proliferation and collagen production. Biomaterials 2004, 25, 2941–2948. [Google Scholar] [CrossRef] [PubMed]
  107. Maeno, S.; Niki, Y.; Matsumoto, H.; Morioka, H.; Yatabe, T.; Funayama, A.; Toyama, Y.; Taguchi, T.; Tanaka, J. The effect of calcium ion concentration on osteoblast viability, proliferation and differentiation in monolayer and 3D culture. Biomaterials 2005, 26, 4847–4855. [Google Scholar] [CrossRef]
  108. Gu, H.; Guo, F.; Zhou, X.; Gong, L.; Zhang, Y.; Zhai, W.; Chen, L.; Cen, L.; Yin, S.; Chang, J. The stimulation of osteogenic differentiation of human adipose-derived stem cells by ionic products from akermanite dissolution via activation of the ERK pathway. Biomaterials 2011, 32, 7023–7033. [Google Scholar] [CrossRef]
  109. Yin, J.; Pan, S.; Guo, X.; Gao, Y.; Zhu, D.; Yang, Q.; Gao, J.; Zhang, C.; Chen, Y. Nb 2 C MXene-Functionalized Scaffolds Enables Osteosarcoma Phototherapy and Angiogenesis/Osteogenesis of Bone Defects. Nano-Micro Lett. 2021, 13, 30. [Google Scholar] [CrossRef]
  110. Han, X.; Jing, X.; Yang, D.; Lin, H.; Wang, Z.; Ran, H.; Li, P.; Chen, Y. Therapeutic mesopore construction on 2D Nb2C MXenes for targeted and enhanced chemo-photothermal cancer therapy in NIR-II biowindow. Theranostics 2018, 8, 4491. [Google Scholar] [CrossRef]
  111. Hadjiargyrou, M.; O’Keefe, R.J. The convergence of fracture repair and stem cells: Interplay of genes, aging, environmental factors and disease. J. Bone Miner. Res. 2014, 29, 2307–2322. [Google Scholar] [CrossRef]
  112. Melincovici, C.S.; Boşca, A.B.; Şuşman, S.; Mărginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.-M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)-key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018, 59, 455–467. [Google Scholar] [PubMed]
  113. Guneta, V.; Zhou, Z.; Tan, N.S.; Sugii, S.; Wong, M.; Choong, C. Recellularization of decellularized adipose tissue-derived stem cells: Role of the cell-secreted extracellular matrix in cellular differentiation. Biomater. Sci. 2018, 6, 168–178. [Google Scholar] [CrossRef] [PubMed]
  114. Huang, W.; Yang, S.; Shao, J.; Li, Y.-P. Signaling and transcriptional regulation in osteoblast commitment and differentiation. Front. Biosci. A J. Virtual Libr. 2007, 12, 3068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Lin, S.; Yuk, H.; Zhang, T.; Parada, G.A.; Koo, H.; Yu, C.; Zhao, X. Stretchable hydrogel electronics and devices. Adv. Mater. 2016, 28, 4497–4505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Shin, S.R.; Bae, H.; Cha, J.M.; Mun, J.Y.; Chen, Y.-C.; Tekin, H.; Shin, H.; Zarabi, S.; Dokmeci, M.R.; Tang, S. Carbon nanotube reinforced hybrid microgels as scaffold materials for cell encapsulation. ACS Nano 2011, 6, 362–372. [Google Scholar] [CrossRef] [Green Version]
  117. Aswathy, S.; Narendrakumar, U.; Manjubala, I. Commercial hydrogels for biomedical applications. Heliyon 2020, 6, e03719. [Google Scholar] [CrossRef] [PubMed]
  118. Bao, W.; Li, M.; Yang, Y.; Wan, Y.; Wang, X.; Bi, N.; Li, C. Advancements and frontiers in the high performance of natural hydrogels for cartilage tissue engineering. Front. Chem. 2020, 8, 53. [Google Scholar] [CrossRef] [Green Version]
  119. Hoare, T.R.; Kohane, D.S. Hydrogels in drug delivery: Progress and challenges. Polymer 2008, 49, 1993–2007. [Google Scholar] [CrossRef] [Green Version]
  120. Ghasemiyeh, P.; Mohammadi-Samani, S. Hydrogels as drug delivery systems; pros and cons. Trends Pharm. Sci. 2019, 5, 7–24. [Google Scholar]
  121. Correa, S.; Grosskopf, A.K.; Lopez Hernandez, H.; Chan, D.; Yu, A.C.; Stapleton, L.M.; Appel, E.A. Translational applications of hydrogels. Chem. Rev. 2021, 121, 11385–11457. [Google Scholar] [CrossRef]
  122. Øvrebø, Ø.; Perale, G.; Wojciechowski, J.P.; Echalier, C.; Jeffers, J.R.; Stevens, M.M.; Haugen, H.J.; Rossi, F. Design and clinical application of injectable hydrogels for musculoskeletal therapy. Bioeng. Transl. Med. 2022, 7, e10295. [Google Scholar] [CrossRef]
  123. Mandal, A.; Clegg, J.R.; Anselmo, A.C.; Mitragotri, S. Hydrogels in the clinic. Bioeng. Transl. Med. 2020, 5, e10158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Shao, R.; Wang, Y.; Li, L.; Dong, Y.; Zhao, J.; Liang, W. Bone tumors effective therapy through functionalized hydrogels: Current developments and future expectations. Drug Deliv. 2022, 29, 1631–1647. [Google Scholar] [CrossRef]
  125. Jiang, Y.-W.; Gao, G.; Hu, P.; Liu, J.-B.; Guo, Y.; Zhang, X.; Yu, X.-W.; Wu, F.-G.; Lu, X. Palladium nanosheet-knotted injectable hydrogels formed via palladium–sulfur bonding for synergistic chemo-photothermal therapy. Nanoscale 2020, 12, 210–219. [Google Scholar] [CrossRef] [PubMed]
  126. Li, C.; Zhang, W.; Wang, R.; Du, X.-F.; Jiang, D.; Liu, B.; Nie, Y.; Liao, J.; Chen, Y.; Liang, X. Nanocomposite multifunctional hydrogel for suppressing osteosarcoma recurrence and enhancing bone regeneration. Chem. Eng. J. 2022, 435, 134896. [Google Scholar] [CrossRef]
  127. Jyoti, J.; Kiran, A.; Sandhu, M.; Kumar, A.; Singh, B.P.; Kumar, N. Improved nanomechanical and in-vitro biocompatibility of graphene oxide-carbon nanotube hydroxyapatite hybrid composites by synergistic effect. J. Mech. Behav. Biomed. Mater. 2021, 117, 104376. [Google Scholar] [CrossRef] [PubMed]
  128. Hu, H.; Li, Z.; Sun, W.; Li, R.; Li, H.; Khor, K.A. Friction and wear behaviors of reduced graphene oxide-and carbon nanotube-reinforced hydroxyapatite Bioceramics. Front. Mater. 2020, 7, 564624. [Google Scholar] [CrossRef]
  129. Zagórska-Dziok, M.; Sobczak, M. Hydrogel-based active substance release systems for cosmetology and dermatology application: A review. Pharmaceutics 2020, 12, 396. [Google Scholar] [CrossRef] [PubMed]
  130. Li, J.; Wu, C.; Chu, P.K.; Gelinsky, M. 3D printing of hydrogels: Rational design strategies and emerging biomedical applications. Mater. Sci. Eng. R Rep. 2020, 140, 100543. [Google Scholar] [CrossRef]
  131. Billiet, T.; Vandenhaute, M.; Schelfhout, J.; Van Vlierberghe, S.; Dubruel, P. A review of trends and limitations in hydrogel-rapid prototyping for tissue engineering. Biomaterials 2012, 33, 6020–6041. [Google Scholar] [CrossRef]
Figure 1. Flowchart of literature search using PubMed.
Figure 1. Flowchart of literature search using PubMed.
Gels 09 00274 g001
Figure 2. Example schematic of hydrogel fabrication and site-specific incorporation to treat osteosarcoma. Hydrogels may be stimuli-responsive by self-regulation in response to pH, temperature, or mechanical stress. The hydrogels may also include properties that are pro-osteogenic while suppressing or even reducing tumor growth. This figure was generated using the Servier Medical Art. Servier Medical Art is licensed under a Creative Commons Attribution 3.0 Unported License [76].
Figure 2. Example schematic of hydrogel fabrication and site-specific incorporation to treat osteosarcoma. Hydrogels may be stimuli-responsive by self-regulation in response to pH, temperature, or mechanical stress. The hydrogels may also include properties that are pro-osteogenic while suppressing or even reducing tumor growth. This figure was generated using the Servier Medical Art. Servier Medical Art is licensed under a Creative Commons Attribution 3.0 Unported License [76].
Gels 09 00274 g002
Figure 5. Schematic of hydrogel combination drug therapies, commonly administered to induce slow drug release and cancer cell death in osteosarcoma [81,82,84,92,93,94]. This figure was generated using the Servier Medical Art. Servier Medical Art is licensed under a Creative Commons Attribution 3.0 Unported License [76].
Figure 5. Schematic of hydrogel combination drug therapies, commonly administered to induce slow drug release and cancer cell death in osteosarcoma [81,82,84,92,93,94]. This figure was generated using the Servier Medical Art. Servier Medical Art is licensed under a Creative Commons Attribution 3.0 Unported License [76].
Gels 09 00274 g005
Table 1. Summary of studies treating osteosarcoma.
Table 1. Summary of studies treating osteosarcoma.
Author/YearExperimentStrainHydrogel CompositionBenefitsDrawbacksMethodsResults
Wu et al., 2018 [80]In vivo, in vitroMG-63 cells, BALB/c mice injected with MG-63 cells (n = N/A)Gemcitabine (GEM) loaded liposomes with gelatin methacryloyl (GelMA)Rapid cell death within 4 h, sustained release of gemcitabineInflammatory reaction in vivo, low cell death in vivoLive/dead, degradation %, drug release %, neutrophil counting, H & E stainingGEM-GelMA scaffolds demonstrated cytotoxicity in vitro but marginal tumor suppression in vivo. An inflammatory reaction was present in vivo.
Zheng et al., 2017 [81]In vivo, in vitroBALB/c male mice injected with K7 cells (n = 80)Poly(L-alanine-co-L-phenylalanine)-block-poly(ethylene
glycol)-co-poly(L-alanine-co-L-phenylalanine))
hydrogel co-loaded with combretastatin A-4 and docetaxel
Biocompatible, biodegradable, rapid release and diffusion of drugs, apoptosis of osteosarcoma, blockage of cell proliferationSafety concerns due to rapid weight loss, accelerated diffusion rate within 1 week may risk drug overloadingDegradation %, drug release %, H & E staining, tumor volume measurements, the survival rateCA4-DTX co-loaded hydrogels caused a reduction in osteosarcoma tumor size and did not display any adverse effects on any internal organs. The hydrogel demonstrated sustained release over 48 days of DTX.
Sun et al., 2020 [83]In vivo, in vitroBALB/c female mice injected with K7M2 cells (n = 20)mPEG45–PLV19 co-loaded with oxaliplatin and alendronateDecrease in tumor weight, tumor suppression, no organ toxicity, biocompatibleSafety concerns due to rapid weight loss, the burst effect of drug release may influence temperature-sensitive hydrogelMTT assay, degradation %, drug release %, tumor volume measurements, H & E stain, microCTALN-OXA-loaded hydrogels reduced tumor size and growth. The hydrogel degraded by 50% within 27 days and released 40% of the drugs within 15 days. No toxicity was shown to the internal organs of the mice.
Tan et al., 2021 [86]In vivo, in vitroNIH3T3 cells, K7M2wt cells, BALB/c female mice (n = 24)Curcumin-loaded PLGA microspheres in methylcellulose and IR820Biodegradable, biocompatible, bone regrowthNecessary use of irradiation with Cur for optimal effects, inflammatory reaction in vivoLive/dead, CCK-8 viability assay, H & E stain, microCT, tumor volume measurementsCur-loaded PLGA/Methylcellulose scaffolds demonstrated cell cytotoxicity; however, prominent effects were seen with the addition of irradiation. Tumor suppression was seen in vivo.
Ma et al., 2015 [87]In vivo, in vitroSaos-2 cells, MG-63 cells, BALB/c male mice with Saos-2 xenografts (n = 48)PLGA-PEG-PLGA triblock copolymer loaded with doxorubicin, cisplatin, and methotrexateSlow release, pro-apoptotic, no organ toxicityPLGA-PEG-PLGA copolymer has minimal anti-cancer effects alone. Bone regeneration was not assessedDrug release %, MTT assay, PCR, tumor volume measurements, H & E stainThe addition of chemotherapeutic drugs is necessary for cytotoxicity. Tumor volume and cell viability were suppressed. No toxicity was shown to the internal organs of the mice.
Liao et al., 2021 [91]in vivo, in vitroK7M2 cells, Mice MSCs, and BALB/c mice for the in vivo assessment (n = 20)Methacrylated gelatin/methacrylated chondroitin sulfate hydrogel with hybrid gold nanorods (GNRs) and nanohydroxyapatite (nHA)Decrease in osteosarcoma and increased deposition of stem cells for dual-purpose treatment, minimal destruction of healthy tissue.Potential inflammatory response due to macrophage and lymphocyte infiltration, bone grew for 2 weeks and was not assessed for longer.Live/dead, H & E stain, microCT, bone volume measurementsThe methacrylated gelatin/methacrylated chondroitin sulfate hydrogel with hybrid gold nanorods (GNRs) and nanohydroxyapatite (nHA) eliminated the tumor remnants with no further recurrence of the osteosarcoma. An increase in bone volume was seen.
Zhang et al., 2018 [92]in vivo, in vitroHuman MSCs, female New Zealand white rabbits (n = 3)Hyaluronic acid (HA), pamidronate (PAM), Mg2+ cofactor, and dexamethasone phosphorylate (DexP)Flexible hydrogel with a non-restricted microenvironment for cells, quick molding to bone defect, slow degradation with steady, sustained releaseAngiogenesis and bone growth were not addressedFluorescent staining, PCR, microCT, H & E stainHydrogel loaded with human MSCs, dexamethasone, and magnesium showed increased bone regeneration in a rabbit femur defect. Greater bone volume was demonstrated in the treated femur.
Yu et al., 2021 [93]in vitroMG-63 and MC3T3-E1 cellsCurcumin-loaded chitosan particles encapsulated in a methacrylated silk fibroin/hyaluronic acid hydrogelLow immunogenicity, can handle high compression forces, adherence to tissueNo data regarding how the hydrogel would perform in vivoDrug release %, Live/Dead stain, MTS assayCCNP-SF/HAMA hydrogels demonstrated anti-tumor activity and osteoblastic proliferation in vitro with varying Cur concentrations.
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

Tharakan, S.; Raja, I.; Pietraru, A.; Sarecha, E.; Gresita, A.; Petcu, E.; Ilyas, A.; Hadjiargyrou, M. The Use of Hydrogels for the Treatment of Bone Osteosarcoma via Localized Drug-Delivery and Tissue Regeneration: A Narrative Review. Gels 2023, 9, 274. https://0-doi-org.brum.beds.ac.uk/10.3390/gels9040274

AMA Style

Tharakan S, Raja I, Pietraru A, Sarecha E, Gresita A, Petcu E, Ilyas A, Hadjiargyrou M. The Use of Hydrogels for the Treatment of Bone Osteosarcoma via Localized Drug-Delivery and Tissue Regeneration: A Narrative Review. Gels. 2023; 9(4):274. https://0-doi-org.brum.beds.ac.uk/10.3390/gels9040274

Chicago/Turabian Style

Tharakan, Shebin, Iman Raja, Annette Pietraru, Elina Sarecha, Andrei Gresita, Eugen Petcu, Azhar Ilyas, and Michael Hadjiargyrou. 2023. "The Use of Hydrogels for the Treatment of Bone Osteosarcoma via Localized Drug-Delivery and Tissue Regeneration: A Narrative Review" Gels 9, no. 4: 274. https://0-doi-org.brum.beds.ac.uk/10.3390/gels9040274

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