Next Article in Journal
Design and Optimization of In Situ Gelling Mucoadhesive Eye Drops Containing Dexamethasone
Next Article in Special Issue
Development of Ciprofloxacin-Loaded Bilosomes In-Situ Gel for Ocular Delivery: Optimization, In-Vitro Characterization, Ex-Vivo Permeation, and Antimicrobial Study
Previous Article in Journal
Some Remarkable Rheological and Conducting Properties of Hybrid PVC Thermoreversible Gels/Organogels
Previous Article in Special Issue
Poloxamer-Based Scaffolds for Tissue Engineering Applications: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gene Regulations upon Hydrogel-Mediated Drug Delivery Systems in Skin Cancers—An Overview

by
Ramya Mathiyalagan
1,†,
Anjali Kariyarath Valappil
2,†,
Deok Chun Yang
1,2,
Se Chan Kang
1,2,* and
Thavasyappan Thambi
1,*
1
Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
2
Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Gels 2022, 8(9), 560; https://doi.org/10.3390/gels8090560
Submission received: 31 July 2022 / Revised: 24 August 2022 / Accepted: 30 August 2022 / Published: 2 September 2022
(This article belongs to the Special Issue Advances in Biomedical Hydrogels)

Abstract

:
The incidence of skin cancer has increased dramatically in recent years, particularly in Caucasian populations. Specifically, the metastatic melanoma is one of the most aggressive cancers and is responsible for more than 80% of skin cancer deaths around the globe. Though there are many treatment techniques, and drugs have been used to cure this belligerent skin cancer, the side effects and reduced bioavailability of drug in the targeted area makes it difficult to eradicate. In addition, cellular metabolic pathways are controlled by the skin cancer driver genes, and mutations in these genes promote tumor progression. Consequently, the MAPK (RAS–RAF–MEK–ERK pathway), WNT and PI3K signaling pathways are found to be important molecular regulators in melanoma development. Even though hydrogels have turned out to be a promising drug delivery system in skin cancer treatment, the regulations at the molecular level have not been reported. Thus, we aimed to decipher the molecular pathways of hydrogel drug delivery systems for skin cancer in this review. Special attention has been paid to the hydrogel systems that deliver drugs to regulate MAPK, PI3K–AKT–mTOR, JAK–STAT and cGAS-STING pathways. These signaling pathways can be molecular drivers of skin cancers and possible potential targets for the further research on treatment of skin cancers.

1. Introduction

Skin is one of the largest and complex organs in the human body and it has some distinct functions such as acting as a protective fence in defense against injury due to UV radiation, chemicals and infections by microorganisms, enables the ability to feel, adjust body temperature and sensation [1]. Structurally, skin is a multi-lamellar structure, and the layers are epidermis, dermis, and subcutaneous tissue (Figure 1). The outermost layer, i.e., epidermis, is composed of the stratum corneum (SC) and the viable epidermis. The SC is metabolically inactive and possesses 10–24 layers of non-viable, elongated corneocytes (keratinized). These corneocytes are found to be embedded in the lipid bilayer matrix and this structural arrangement is known as the “Brick and Mortar” arrangement [2]. The extracellular lipid is composed of crystalline and liquid lipid phases. Hence, the skin acts as a principal physiological barrier inhibiting the uptake of polar compounds with high molecular weight (>500 Da) [3]. The dermis is thicker (3–5 mm) than the epidermis and comprises collagen fibrils and elastic connective tissues. Dermis consist of fibroblasts, mast cells, macrophages, lymphocytes, and melanocytes along with blood vessels, nerves, sweat and sebaceous glands [4]. Structurally, dermis does not show the same resistance to drug penetration as the SC, however, permeation of lipophilic drugs may be reduced in this layer. The subcutaneous tissue is a specialized fat cell layer, inter-connected by collagen and elastin fibers. Large quantities of fat are produced and stored in this layer [5].
To date, many reviews have been done on the applications against skin cancers [6,7]. However, the information on the regulations of the hydrogel drug delivery system of the drugs on the molecular mechanisms and pathways-related skin cancers is limited. This article provides a brief about skin cancer and its molecular drivers, and current treatment drugs along with an emphasis on studies conducted on hydrogel delivery system regulating gene expressions in skin cancers.

2. Skin Cancer

Skin cancer is the most common cancer among Caucasian populations and according to U.S. estimates, almost one in five Americans will develop some kind of skin cancer. DNA damage caused by ultraviolet (UV) radiation, followed by failure of DNA damage repair mechanisms, are the primary source of these neoplasms, and Fitzpatrick skin type and immunosuppression are also considered as potential risk factors for the development of skin cancer [8]. Skin cancers will develop in the outer most layers of the skin in the early stage and, if not treated, they may invade deeper into the skin with metastatic (secondary malignant growth far away from the primary origin) potential [7,9]. Based on their origin, skin cancers have been broadly classified into two types: non-melanoma skin cancer (NMSC) and melanoma.

2.1. NMSC

NMSCs are known to be the most common human cancers and UV radiation is the primary cause of them. UV-A (320–400 nm), UV-B (280–320 nm) and UV-C (100–280 nm) are the three regions of the UV spectrum. However, UV-B is considered as the most carcinogenic radiation and long term exposure to UV-B radiation will result in specific mutations in keratinocytes followed by NMSC [10]. Basel cell carcinoma (BSC) is the most common skin neoplasm, which develops in the basal layer of epidermis and is usually located on the face or the back of hands. Squamous cell carcinoma (SCC) is known to be the second most common skin cancer, develops in the squamous (spinosum) layer of epidermis and occurs mainly on neck and head areas (Figure 1). BCC shows slow growth and spreads locally with little or no metastasis, however, SCC may advance to invasive SCC and risk of metastasis is 2 to 6% [11,12]. Normally, earlier surgical removal of the metastatic NMSC and precancerous superficial lesions may inhibit its further progression into tumors.
Apoptosis is a crucial molecular mechanism in the development of malignancies in almost all type of cancers and an important cell survival pathway. The NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells), Bcl-2, p53, TNF-related apoptosis-inducing ligand (TRAIL), ubiquitin ligases, overexpression of COX-2, mitogen-activated protein kinase (MAPK/ERK) pathways have significant role in skin cancers. The NF-κB regulates different cell survival pathways which control cell growth. Clinical studies have proven that disrupting NF-κB signaling in the epidermis causes terminal differentiation and promotes the appearance of SCC [13]. Bcl-2 and Bcl-x shows the overexpression in BCCs and SCC.
The p53 gene, known as the “guardian of the genome” is a member of the Bcl-2 protein family which found mutated in most of the human cancers. Mutations in the p53 gene induced by UV radiation has been regarded as a critical factor for developing skin cancer, as the susceptibility to apoptosis reduction would favor survival and tumor formation of mutated keratinocytes. Mutations of p53 are found in majority of NMSCs (more than 90% in SCC and 50% in BCC). Skin cancer cells cannot express Fas, but they can simultaneously express FasL, and thereby infiltrate antitumor T cells that express Fas will be killed. Reduction in the expression of the death receptor for TRAIL (a ligand that induces apoptosis from TNF family) was also observed in malignant cells in SCC [13,14,15]. The extracellular ligand-binding domain of epidermal growth factor receptor (EGFR) can bind to different ligands, i.e., epiregulin, EGF and TGF. Mutations in the EGFR tyrosine kinase domain activates the antiapoptotic signaling pathways in PI3K/AKT, ERK/MAPK and JAK–STAT [16]. Agustí et al. evaluated EGFR amplification deviations majorly in SCC [17].
Variations in the MAPK ERK signaling pathway have been detected in SCC. In non-transformed epidermis, Ras and Raf inhibit differentiation, stimulate cell division, and increase the expression of integrins [16]. In that K-RAS and H-RAS shows lower and higher level of mutations in SCC [13]. Despite the low rate of mutant RAS genes, an increase in levels of Ras with active GTP is observed in most tissues of spontaneous human SCC.

2.2. Melanoma

Melanoma is the deadliest skin cancer, originating from melanocytes, that predominantly affects younger and middle-aged people. Melanoma is not only developed on the skin, but also develops in the eyes, vagina, anus, sinus and oropharynx. However, occurrence in these areas consists of only 5% of total melanoma incidents. Cutaneous melanomas are classified as superficial spreading, lentigo malignant, nodular and acral lentiginous [18]. Because of their altering presentations, it is not easy to classify malignant melanomas. Though the incidence of melanoma is least, its often associated as an aggressive cancer condition for death, with increased tumor cell invasion and migration to other organs in the metastatic stage.
Various factors such as genetic and environmental factors such as prolonged sun exposure and sun burns by UV irradiation, lower melanin pigments, heredity, aging [10], more melanocytic nevi and immunosuppression in post-transplant patients can induce melanoma. The transformation of melanocytes (melanin producing cells present in the epidermal layer) into melanoma cells is a multistage process by occurrence of genomic alterations [19]. During the growth phase of the melanoma cells, it invades into the dermis/subcutaneous tissues followed by penetration into the capillaries and eventually enters the blood circulation to facilitate distant metastasis. The genetic mutations such as deletions, amplification, DNA methylation and translocations which drive melanoma were identified by genome-wide sequencing [20]. Therefore, the important genes that are known to be altered/mutated in melanoma and the molecular pathways involved in melanomagenesis are summarized in Table 1. Even though key genetic drivers are required for melanomagenesis, key microenvironmental factors play vital roles in modulating melanomagenesis and progression.
The MAPK pathway is most regularly triggered in cancer to facilitate rapid proliferation of tumor cells. Intracellular sequential activation of Ras, Raf, MEK, and ERK take place in with regard to extracellular binding of growth factors to receptor tyrosine kinases (RTKs) to regulate many oncogenic biological activities. The v-Raf murine sarcoma viral oncogene homologue B1 (BRAF) is one of the best-studied oncogenic mutations in melanoma. It encodes a serine/threonine protein kinase, a key regulator in the RAS–RAF–MEK–ERK MAPK pathway. Point mutation of BRAF by the substitution of valine to glutamic acid at codon 600 (V600E) occurs frequently (more than 50%) in melanoma than other types, which leads to a downstream MAPK pathway [47]. The wild-type BRAF melanomas have oncogenic mutations in upstream components of the MAPK pathway, such as NRAS (neuroblastoma RAS viral oncogene homolog), KIT (v-Kit Hardy–Zuckerman 4 feline sarcoma viral oncogene homolog), GNAQ (guanine nucleotide-binding protein, q polypeptide) and GNA11 (guanine nucleotide-binding protein, a11) [19].
Uncontrolled cell cycle is an important characteristic of melanoma development and p16INK4A is a key down-regulator of the cell cycle, which is induced by the expression of oncogenic BRAFV600E. CDKN2A locus encodes p16INK4A and is found mutated in 25% of melanoma types. The initiation of p16INK4A by the MAPK pathway along with activation of microphthalmia-associated transcription factor (MITF) locus amplification is found in 20–30% of melanomas [48]. Triggering of the PI3K (phosphoinositide 3-kinase)–AKT–mTOR (mammalian target of rapamycin) pathway occurs constitutively in BRAF-initiated melanogenesis which inactivate PTEN (phosphatase and tensin homolog), a down-regulator of this pathway [26,27]. Though NRAS and PTEN mutations are mutually exclusive in melanomas, an oncogenic RAS also can trigger the PI3K–AKT–mTOR pathway. By contrast, even though mutations in the catalytic subunit of PI3K, or AKT1, AKT2, and AKT3, are rare in melanoma, immunopositivity of AKT3 is common in melanoma, and could activate PI3K–AKT–mTOR in PTEN wild type tumors. Amplification or mutations in CDK4 is another genetic alteration found in melanoma, which is the binding partner of p16INK4a. Hyperactivation of ERK or loss of p16INK4A can dysregulate the CDK4 pathway. Both CDKN2A and CDK4 have important roles in controlling cell cycle, as both mutate, which disturbs the G1/S-phase checkpoint [49,50].

2.3. Current Treatments and Drugs for Skin Cancers

Early diagnosis and immediate treatment are important in any type of skin cancer. Surgical and non-surgical treatments such as topical therapies are mostly carried out for skin cancer lesions [51]. Depending on the cancer progression stage, skin cancers can be treated by surgery [52], immunotherapy [53], cryosurgery [54], laser therapy [55], curettage, desiccation [56], dermabrasion [57], targeted therapy, photodynamic therapy [58], chemotherapy and radiotherapy [59]. Commonly used medications against melanoma include 5-fluorouracil (5-FU, Efudex), Imiquimod, Resiquimod, Ingenol mebutate, Diclofenac, cisplatin, etc. (Table 2). However, these conventional treatment methods and drugs can induce side effects such as hypopigmentation, scars, loss of hair, edema, gastrointestinal irritabilities such as chronic ulcer formation, blister formation and radiodermatitis.
SC acts as a major barrier to the penetration and delivery of adequate concentration of anti-skin cancer drugs to the targeted site when administrated topically. Many drugs that are currently used to treat cancer have a limited distribution in the tumor area and there are multiple factors, such as solid tumor development far away from the blood vessels, extracellular matrix composition, and elevated interstitial fluid, which cause the limited drug distribution [76]. Tumor microenvironment is a highly dynamic network of cells and molecules that create a favorable condition for the development of tumors. Immunosuppressive cells such as regulatory T lymphocytes and tumor-associated macrophages are general characteristic of the tumor microenvironment that are associated with extracellular matrix destruction, angiogenesis and metastasis [77,78]. The immune system of hosts has significant importance in response against cancer. However, most of the common cancer therapies that are used at present have immunosuppressive effects. For instance, myelosuppression is a common cause of chemotherapy and ionizing radiation [58,79]. Even though there are different immunotherapeutic techniques such as cytokines and monoclonal antibody-based therapies, antitumor vaccines and T cell therapies for treating SKs, disadvantages such as systemic side effects and cost of the therapies make immunotherapies still in their infancy [80].

3. Hydrogel: Promising Drug Delivery Systems to Treat Skin Cancers

To overcome the side effects associated with anti-neoplastic drugs in skin cancer treatment, hydrogel-based drug delivery systems could be promising drug candidates [81,82,83,84,85]. The hydrogels are three-dimensional, highly crosslinked polymers that can retain a substantial quantity of water in their swollen state due to the hydrophilic –OH, –CONH–, –CONH2, –COOH, and –SO3H groups [86,87,88,89]. Classification of hydrogels can be done based on the nature of material (synthetic, natural or hybrid) [90], mechanism of gel formation (chemically or physically crosslinked) [91], nature of side group (cationic, anionic or neutral) [92,93], biodegradability (degradable or non-degradable) [94,95], and the degree of swelling (low, high or superabsorbent) [96] and porosity (micro-, macro- or super porous) [97]. Hence, the compositions and synthesis conditions determine hydrogel properties and structures. For instance, highly porous structures permit more adherence and entrap the therapeutic agents in it, and the release of therapeutic agents can be controlled by regulating its porous structure [98,99,100]. These high swelling properties of biocarriers have a comparable degree of elasticity to natural tissues, and can undergo gel–solid phase transitions in response to various types of stimuli such as temperature, light, pressure, electric field, magnetic field, ionic strength and pH [101,102,103]. The natural polymers have reached a considerable significance in drug delivery applications, due to their characteristics such as biocompatibility, biodegradability, bifunctionality, biochemical stability, improved drug solubility, controlled drug release, cost effectiveness and nontoxicity [104,105,106,107]. These advantages, identical to the native extracellular matrix (ECM), and tunable physical and mechanical properties, aid in a vast variety of biomedical applications [108,109,110].
Unique characteristics of hydrogel such as tunable porous structure, high strength and stimulus response have made them widely used as carriers or vehicles for different biomolecules such as drugs, nucleic acids, antibodies, metal ions and enzymes, and are reported to be effective against different types of cancers such as lung cancer [111], leukemia [112], colon cancer [113], breast cancer [114], melanoma [115,116], hepatocellular carcinoma [116,117], etc. In addition, it is applied in the delivery of antibodies and other immune modulatory molecules at tumor sites in immunotherapy [118], drug carriers for the eye [119] and drug-delivering contact lenses for glaucoma therapy, in tissue engineering and mesenchymal stem/stromal cells [120]. Hence, it has been used to treat osteoporosis [121], osteoarthritis [122] and wound healing [123]. The topical and transdermal drug delivery using hydrogel is a convenient way to deliver the drugs systemically, in which the drug penetrates through the SC initially and to the epidermis and dermis for systemic absorption via dermal microcirculation [124]. Hence, it has many advantages such as increased patient compliance by reduced dosing, prevents pre-systemic metabolism for enhanced bioavailability [125], and soft swollen hydrogels [126]. In contrast to topical treatment, injectable hydrogels are generally introduced into the body via syringe or catheter. Since the blood circulation will quickly remove the normal chemotherapeutics injected inside the body, the effect of drugs will not be enough to kill the cancer cells. Therefore, engineering injectable hydrogels by physical or chemical cross-linking for sustained and controlled drug release at in situ (near cancer) upon minimal injection enables higher drug concentration at the targeted site while diminishing the systemic drug concentration and the associated site effects. Moreover, the drugs can be delivered into tissues that are difficult to access through surgery [127,128]. Hydrogel particles are transported to the intercellular matrix through plasma membrane (PM) during drug or gene delivery and PM act as a major obstacle for the efficient delivery. The small or macro molecules enter the cells by endocytosis and target specific organelles for efficient drug delivery. Hydrogels are mainly internalized through endocytosis with clathrin- and dynamin-dependent pathways [129]. The physiochemical properties such as shape, size, charge and chemistry of the surface of colloidal particles determine the cell translocation and intracellular distribution [130,131,132]. Particles with nanometer to micron sizes are generally subjected to intercellular uptake. However, particles with a diameter of around 100 nm are known to show a higher degree of uptake. For example, particles with a 100–200 nm size range avoided premature clearance by the reticuloendothelial system in cancer cells [133,134]. The smaller particles are internalized into B16F10 murine melanoma cells through clathrin-dependent pathway and microtubule dependent intracellular trafficking whereas larger particles are internalized through caveolae mechanism [135].

4. A Brief Background on the Use of Hydrogel-Based Drug Delivery System to Regulate Skin Cancer Related Genes

Over the past decade, a plethora of chemotherapeutic agents has been developed or studied to challenge skin cancers and most of these agents remained unsuccessful, especially to eradicate melanoma. Because melanoma is considered to be intrinsically resistant to chemotherapeutics, and different mechanisms such as overexpression of drug efflux proteins, alteration of enzyme activation, deregulation of apoptosis, Ras mutation, epithelial to mesenchymal transition, and deregulation of microRNAs expression are responsible for this drug resistance [136]. There will be a significant change in the recovery rate if this drug resistance is conquered. Availability of trace amounts of drugs in the tumor microenvironment is one of the factors that contribute to sensitivity of drugs. Since tumor microenvironment is very complex and dynamic, it is very difficult to deliver chemotherapeutics in sufficient concentration. Although previous studies have reported the need of an efficient advanced drug delivery system to overcome this problem, microsphere or nanomaterial-based cancer eradication developed slowly. However, initial burst release, increased drug accumulation in the tumor and rapid elimination by the reticuloendothelial system have reduced the efficacy of nanoformulations to affect the cancer cells [137,138]. Hydrogels are potential carriers for localized delivery of anti-neoplastic agents. Blood supply and morphology of blood vessel network do not affect the hydrogel-based delivery system. Therefore, hydrogels can be utilized to carry drugs that are capable of killing cancer cells as well as to regulate the genes related to skin cancer and deliver in the targeted local tumor microenvironment. Because of molecular complexity of SK, combinatorial drug therapy is gaining more importance. In opposition to single-drug therapy, multiple-agent therapy can maximize the therapeutic effects, modulate cancer related signaling pathways, and overcome drug resistance. In addition, combinatorial drug therapies may be able to trigger senescence of cancer cells and permit clearance by T cells. Hydrogels can carry and deliver more than one therapeutic agent simultaneously to the targeted site [7,139,140,141]. To decipher the gene regulations mediated by hydrogel, Zhao, Y., et al. developed an injectable hydrogel loaded with Cripto-1 receptor antibodies (2B11) for embryonic microenvironments on tumor reversion treatment using B16 tumor-bearing mice model [142] in which cancer cell morphology reversed normal melanin cells. Additional RNA-sequencing experiments in comparison with the whole gene expression show that 2B11 hydrogels could significantly stimulate apoptosis. Oncogenes (Kit, Itga4, Hapln1) were downregulated and tumor suppressors genes (Irf8, Trail, Casp1, Aim2, Irf1) were upregulated compared with the control group. The interleukin-15 (IL-15)- and cisplatin (CDDP)-loaded poly (ethylene glycol)-poly(γ-ethyl-L-glutamate) diblock copolymers (mPEG-b-PELG) thermosensitive hydrogels injected into mice bearing B16F0-RFP melanoma cells exhibited synergistic immune regulations such as Cyclin A2, CDK2 and Cdc25A expression was significantly reduced [143]. Another co-delivery of doxorubicin and curcumin peptide hydrogel increased the inhibitory effect of cell growth and improved apoptosis by differential apoptotic/anti-apoptotic gene expression profiles in head and neck squamous cell carcinoma by Karavasili, C., et al. [144]. The various studies that have been carried out on hydrogel delivery systems regulating the molecular mechanisms of skin cancers are included in Table 3 and Figure 2.

5. Conclusions and Perspectives

Skin cancers are often non-cared but persist as the most common malignancy of humans and affect millions of people every year. Even though vast research on skin cancer has enriched our understanding of the disease, 100% eradication is yet to be achieved. The expanding knowledge of molecular mechanisms and pathways involved in NMSCs, and melanoma pathogenesis could lead to identify the potential molecular players. In addition, the hydrogel-based delivery systems with drugs were mainly regulated the MAPK, PI3K–AKT–mTOR, JAK–STAT and cGAS-STING pathways. Though very minimal studies have been conducted, these pathways could be a potential target for further researchers.
In the future, the skin cancers can be treated using various therapies such as chemotherapy, immunotherapy, gene therapy as well as combinatorial therapy. In particular, the advancement in the hydrogel-based drug delivery systems can minimize the side effects of the chemotherapeutic drugs. The sequencing and genome engineering technologies could be able to identify the potential markers in the skin cancer and could help to proceed gene based targeted delivery and therapy. As the p53 gene mutations have been a critical factor for developing skin cancer, the gene therapy could induce the expression of the p53 genes which could restore the apoptosis pathway and destroy the cancer cells. In addition, as the mutation of BRAF is one of the important oncogenic mutations that act as a key regulator in the RAS–RAF–MEK–ERK MAPK pathway in melanoma, development of the targeted therapy to inhibit these point mutations also could treat skin cancers. Moreover, receptor mediated targeted therapy and microbiome replacement therapy also a potential candidate to improve the treatment options.

Author Contributions

Conceptualization, R.M. and T.T.; software, A.K.V.; validation, R.M. and T.T.; formal analysis, R.M. and A.K.V.; resources, S.C.K. and D.C.Y.; data curation R.M. and A.K.V.; writing—original draft preparation, R.M. and A.K.V.; writing—review and editing, R.M. A.K.V. and T.T.; project administration, R.M., S.C.K. and D.C.Y.; funding acquisition, R.M., S.C.K. and D.C.Y.; All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Korea Institute of Planning and Evaluation for Technology in Food, Agriculture and Forestry (IPET) through Agri-Food Export Business Model Development Program, funded by Ministry of Agriculture, Food and Rural Affairs (MAFRA) (Project No.: 320104-03) and This research was also supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (Grant No.: NRF-2020R1I1A1A01070867).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

This work was supported by Korea Institute of Planning and Evaluation for Technology in Food, Agriculture and Forestry (IPET) through Agri-Food Export Business Model Development Program, funded by Ministry of Agriculture, Food and Rural Affairs (MAFRA) (Project No.: 320104-03) and This research was also supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (Grant No.: NRF-2020R1I1A1A01070867).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. You, L.; Cho, J.Y. The regulatory role of Korean ginseng in skin cells. J. Ginseng Res. 2021, 45, 363–370. [Google Scholar] [CrossRef] [PubMed]
  2. Chaturvedi, S.; Garg, A. An insight of techniques for the assessment of permeation flux across the skin for optimization of topical and transdermal drug delivery systems. J. Drug Deliv. Sci. Technol. 2021, 62, 102355. [Google Scholar] [CrossRef]
  3. Bouwstra, J.A.; Ponec, M. The skin barrier in healthy and diseased state. Biochim. Biophys. Acta (BBA) Biomembr. 2006, 1758, 2080–2095. [Google Scholar] [CrossRef]
  4. Katz, M.; Poulsen, B.J. Absorption of drugs through the skin. In Concepts in Biochemical Pharmacology; Springer: Berlin/Heidelberg, Germany, 1971; pp. 103–174. [Google Scholar]
  5. Haque, T.; Rahman, K.M.; Thurston, D.E.; Hadgraft, J.; Lane, M.E. Topical therapies for skin cancer and actinic keratosis. Eur. J. Pharm. Sci. 2015, 77, 279–289. [Google Scholar] [CrossRef]
  6. Marzi, M.; Rostami Chijan, M.; Zarenezhad, E. Hydrogels as promising therapeutic strategy for the treatment of skin cancer. J. Mol. Struct. 2022, 1262, 133014. [Google Scholar] [CrossRef]
  7. Vishnubhakthula, S.; Elupula, R.; Durán-Lara, E.F. Recent advances in hydrogel-based drug delivery for melanoma cancer therapy: A mini review. J. Drug Deliv. 2017, 2017, 7275985. [Google Scholar] [CrossRef] [PubMed]
  8. Murphy, G.M. From precursor to cancer: Field cancerization and the opportunities for therapy. In Non-Surgical Treatment of Keratinocyte Skin Cancer; Springer: Berlin/Heidelberg, Germany, 2010; pp. 1–7. [Google Scholar]
  9. Gordon, R. Skin cancer: An overview of epidemiology and risk factors. Semin. Oncol. Nurs. 2013, 29, 160–169. [Google Scholar] [CrossRef]
  10. Maddodi, N.; Setaluri, V. Role of UV in cutaneous melanoma. Photochem. Photobiol. 2008, 84, 528–536. [Google Scholar] [CrossRef]
  11. Jerant, A.F.; Johnson, J.T.; Sheridan, C.D.; Caffrey, T.J. Early detection and treatment of skin cancer. Am. Fam. Physician 2000, 62, 357–368. [Google Scholar]
  12. Motley, R.; Kersey, P.; Lawrence, C. Multiprofessional guidelines for the management of the patient with primary cutaneous squamous cell carcinoma. Br. J. Dermatol. 2002, 146, 18–25. [Google Scholar] [CrossRef]
  13. Simões, M.F.; Sousa, J.S.; Pais, A.C. Skin cancer and new treatment perspectives: A review. Cancer Lett. 2015, 357, 8–42. [Google Scholar] [CrossRef] [PubMed]
  14. Nickoloff, B.J.; Qin, J.-Z.; Chaturvedi, V.; Bacon, P.; Panella, J.; Denning, M.F. Life and death signaling pathways contributing to skin cancer. J. Investig. Dermatol. Symp. Proc. 2002, 7, 27–35. [Google Scholar] [CrossRef] [PubMed]
  15. Cobanoglu, H.B.; Constantinides, M.; Ural, A. Nonmelanoma skin cancer of the head and neck: Molecular mechanisms. Facial Plast. Surg. Clin. 2012, 20, 437–443. [Google Scholar] [CrossRef]
  16. Uribe, P.; Gonzalez, S. Epidermal growth factor receptor (EGFR) and squamous cell carcinoma of the skin: Molecular bases for EGFR-targeted therapy. Pathol. Res. Pract. 2011, 207, 337–342. [Google Scholar] [CrossRef] [PubMed]
  17. Toll, A.; Salgado, R.; Yébenes, M.; Martín-Ezquerra, G.; Gilaberte, M.; Baró, T.; Solé, F.; Alameda, F.; Espinet, B.; Pujol, R.M. Epidermal growth factor receptor gene numerical aberrations are frequent events in actinic keratoses and invasive cutaneous squamous cell carcinomas. Exp. Dermatol. 2010, 19, 151–153. [Google Scholar] [CrossRef]
  18. Coleman, W.P.; Loria, P.R.; Reed, R.J.; Krementz, E.T. Acral lentiginous melanoma. Arch. Dermatol. 1980, 116, 773–776. [Google Scholar] [CrossRef]
  19. Abildgaard, C.; Guldberg, P. Molecular drivers of cellular metabolic reprogramming in melanoma. Trends Mol. Med. 2015, 21, 164–171. [Google Scholar] [CrossRef]
  20. Paluncic, J.; Kovacevic, Z.; Jansson, P.J.; Kalinowski, D.; Merlot, A.M.; Huang, M.L.-H.; Lok, H.C.; Sahni, S.; Lane, D.J.; Richardson, D.R. Roads to melanoma: Key pathways and emerging players in melanoma progression and oncogenic signaling. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2016, 1863, 770–784. [Google Scholar] [CrossRef]
  21. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  22. Veer, L.J.V.T.; Burgering, B.M.; Versteeg, R.; Boot, A.J.; Ruiter, D.J.; Osanto, S.; Schrier, P.I.; Bos, J.L. N-ras mutations in human cutaneous melanoma from sun-exposed body sites. Mol. Cell. Biol. 1989, 9, 3114–3116. [Google Scholar] [CrossRef]
  23. Curtin, J.A.; Busam, K.; Pinkel, D.; Bastian, B.C. Somatic Activation of KIT in Distinct Subtypes of Melanoma. J. Clin. Oncol. 2006, 24, 4340–4346. [Google Scholar] [CrossRef] [PubMed]
  24. Dahl, C.; Abildgaard, C.; Riber-Hansen, R.; Steiniche, T.; Lade-Keller, J.; Guldberg, P. KIT Is a Frequent Target for Epigenetic Silencing in Cutaneous Melanoma. J. Investig. Dermatol. 2015, 135, 516–524. [Google Scholar] [CrossRef]
  25. Van Raamsdonk, C.D.; Bezrookove, V.; Green, G.; Bauer, J.; Gaugler, L.; O’Brien, J.M.; Simpson, E.M.; Barsh, G.S.; Bastian, B.C. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature 2009, 457, 599–602. [Google Scholar] [CrossRef] [PubMed]
  26. Van Raamsdonk, C.D.; Griewank, K.G.; Crosby, M.B.; Garrido, M.C.; Vemula, S.; Wiesner, T.; Obenauf, A.C.; Wackernagel, W.; Green, G.; Bouvier, N.; et al. Mutations in GNA11 in Uveal Melanoma. N. Engl. J. Med. 2010, 363, 2191–2199. [Google Scholar] [CrossRef] [PubMed]
  27. Freedberg, D.E.; Rigas, S.H.; Russak, J.; Gai, W.; Kaplow, M.; Osman, I.; Turner, F.; Randerson-Moor, J.A.; Houghton, A.; Busam, K.; et al. Frequent p16-Independent Inactivation of p14ARF in Human Melanoma. JNCI J. Natl. Cancer Inst. 2008, 100, 784–795. [Google Scholar] [CrossRef] [PubMed]
  28. Jönsson, G.; Dahl, C.; Staaf, J.; Sandberg, T.; Bendahl, P.O.; Ringnér, M.; Guldberg, P.; Borg, Å. Genomic profiling of malignant melanoma using tiling-resolution arrayCGH. Oncogene 2007, 26, 4738–4748. [Google Scholar] [CrossRef] [PubMed]
  29. Bartkova, J.; Lukas, J.; Guldberg, P.; Alsner, J.; Kirkin, A.F.; Zeuthen, J.; Bartek, J. The p16-cyclin D/Cdk4-pRb Pathway as a Functional Unit Frequently Altered in Melanoma Pathogenesis1. Cancer Res. 1996, 56, 5475–5483. [Google Scholar]
  30. Cully, M.; You, H.; Levine, A.J.; Mak, T.W. Beyond PTEN mutations: The PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat. Rev. Cancer 2006, 6, 184–192. [Google Scholar] [CrossRef]
  31. Guldberg, P.; Straten, P.T.; Ahrenkiel, V.; Seremet, T.; Kirkin, A.F.; Zeuthen, J. Somatic mutation of the Peutz-Jeghers syndrome gene, LKB1/STK11, in malignant melanoma. Oncogene 1999, 18, 1777–1780. [Google Scholar] [CrossRef]
  32. Rowan, A.; Tomlinson, I.; Bataille, V.; MacKie, R.; Healy, E.; Bicknell, D.; Bodmer, W. Somatic Mutations in the Peutz-Jegners (LKB1/STKII) Gene in Sporadic Malignant Melanomas. J. Investig. Dermatol. 1999, 112, 509–511. [Google Scholar] [CrossRef]
  33. Bertolotto, C.; Lesueur, F.; Giuliano, S.; Strub, T.; de Lichy, M.; Bille, K.; Dessen, P.; d’Hayer, B.; Mohamdi, H.; Remenieras, A.; et al. A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma. Nature 2011, 480, 94–98. [Google Scholar] [CrossRef] [PubMed]
  34. Shtivelman, E.; Davies, M.Q.A.; Hwu, P.; Yang, J.; Lotem, M.; Oren, M.; Flaherty, K.T.; Fisher, D.E. Pathways and therapeutic targets in melanoma. Oncotarget 2014, 5, 1701–1752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Maertens, O.; Johnson, B.; Hollstein, P.; Frederick, D.T.; Cooper, Z.A.; Messiaen, L.; Bronson, R.T.; McMahon, M.; Granter, S.; Flaherty, K.; et al. Elucidating Distinct Roles for NF1 in Melanomagenesis. Cancer Discov. 2013, 3, 338–349. [Google Scholar] [CrossRef]
  36. Kraehn, G.M.; Utikal, J.; Udart, M.; Greulich, K.M.; Bezold, G.; Kaskel, P.; Leiter, U.; Peter, R.U. Extra c-myc oncogene copies in high risk cutaneous malignant melanoma and melanoma metastases. Br. J. Cancer 2001, 84, 72–79. [Google Scholar] [CrossRef] [PubMed]
  37. Polsky, D.; Cordon-Cardo, C. Oncogenes in melanoma. Oncogene 2003, 22, 3087–3091. [Google Scholar] [CrossRef]
  38. Sauter, E.R.; Yeo, U.-C.; von Stemm, A.; Zhu, W.; Litwin, S.; Tichansky, D.S.; Pistritto, G.; Nesbit, M.; Pinkel, D.; Herlyn, M. Cyclin D1 is a candidate oncogene in cutaneous melanoma. Cancer Res. 2002, 62, 3200–3206. [Google Scholar]
  39. Muthusamy, V.; Hobbs, C.; Nogueira, C.; Cordon-Cardo, C.; McKee, P.H.; Chin, L.; Bosenberg, M.W. Amplification of CDK4 and MDM2 in malignant melanoma. Genes Chromosomes Cancer 2006, 45, 447–454. [Google Scholar] [CrossRef]
  40. Polsky, D.; Melzer, K.; Hazan, C.; Panageas, K.S.; Busam, K.; Drobnjak, M.; Kamino, H.; Spira, J.G.; Kopf, A.W.; Houghton, A.; et al. HDM2 Protein Overexpression and Prognosis in Primary Malignant Melanoma. JNCI J. Natl. Cancer Inst. 2002, 94, 1803–1806. [Google Scholar] [CrossRef]
  41. Omholt, K.; Kröckel, D.; Ringborg, U.; Hansson, J. Mutations of PIK3CA are rare in cutaneous melanoma. Melanoma Res. 2006, 16, 197–200. [Google Scholar] [CrossRef]
  42. Davies, M.A.; Stemke-Hale, K.; Tellez, C.; Calderone, T.L.; Deng, W.; Prieto, V.G.; Lazar, A.J.F.; Gershenwald, J.E.; Mills, G.B. A novel AKT3 mutation in melanoma tumours and cell lines. Br. J. Cancer 2008, 99, 1265–1268. [Google Scholar] [CrossRef]
  43. Boone, B.; Jacobs, K.; Ferdinande, L.; Taildeman, J.; Lambert, J.; Peeters, M.; Bracke, M.; Pauwels, P.; Brochez, L. EGFR in melanoma: Clinical significance and potential therapeutic target. J. Cutan Pathol. 2011, 38, 492–502. [Google Scholar] [CrossRef] [PubMed]
  44. Yadav, V.; Zhang, X.; Liu, J.; Estrem, S.; Li, S.; Gong, X.-Q.; Buchanan, S.; Henry, J.R.; Starling, J.J.; Peng, S.-B. Reactivation of mitogen-activated protein kinase (MAPK) pathway by FGF receptor 3 (FGFR3)/Ras mediates resistance to vemurafenib in human B-RAF V600E mutant melanoma. J. Biol. Chem. 2012, 287, 28087–28098. [Google Scholar] [CrossRef]
  45. Easty, D.J.; Gray, S.G.; O’Byrne, K.J.; O’Donnell, D.; Bennett, D.C. Receptor tyrosine kinases and their activation in melanoma. Pigment Cell Melanoma Res. 2011, 24, 446–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.-M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef] [PubMed]
  47. Guo, W.; Wang, H.; Li, C. Signal pathways of melanoma and targeted therapy. Signal Transduct. Target. Ther. 2021, 6, 424. [Google Scholar] [CrossRef] [PubMed]
  48. Garraway, L.A.; Widlund, H.R.; Rubin, M.A.; Getz, G.; Berger, A.J.; Ramaswamy, S.; Beroukhim, R.; Milner, D.A.; Granter, S.R.; Du, J.; et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 2005, 436, 117–122. [Google Scholar] [CrossRef] [PubMed]
  49. Soufir, N.; Avril, M.-F.; Chompret, A.; Demenais, F.; Bombled, J.; Spatz, A.; Stoppa-Lyonnet, D.; Group, F.F.M.S.; Bénard, J.; de Paillerets, B.B. Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in France. Hum. Mol. Genet. 1998, 7, 209–216. [Google Scholar] [CrossRef]
  50. Zuo, L.; Weger, J.; Yang, Q.; Goldstein, A.M.; Tucker, M.A.; Walker, G.J.; Hayward, N.; Dracopoli, N.C. Germline mutations in the p16INK4a binding domain of CDK4 in familial melanoma. Nat. Genet. 1996, 12, 97–99. [Google Scholar] [CrossRef]
  51. Krishnan, V.; Mitragotri, S. Nanoparticles for topical drug delivery: Potential for skin cancer treatment. Adv. Drug Deliv. Rev. 2020, 153, 87–108. [Google Scholar] [CrossRef]
  52. Ishizuki, S.; Nakamura, Y. Evidence from Clinical Studies Related to Dermatologic Surgeries for Skin Cancer. Cancers 2022, 14, 3835. [Google Scholar] [CrossRef]
  53. Nanamori, H.; Sawada, Y. Epigenetic Modification of PD-1/PD-L1-Mediated Cancer Immunotherapy against Melanoma. Int. J. Mol. Sci. 2022, 23, 1119. [Google Scholar]
  54. Chinembiri, T.N.; Du Plessis, L.H.; Gerber, M.; Hamman, J.H.; Du Plessis, J. Review of natural compounds for potential skin cancer treatment. Molecules 2014, 19, 11679–11721. [Google Scholar] [PubMed]
  55. Fahradyan, A.; Howell, A.C.; Wolfswinkel, E.M.; Tsuha, M.; Sheth, P.; Wong, A.K. Updates on the management of non-melanoma skin cancer (NMSC). Healthcare 2017, 5, 82. [Google Scholar]
  56. Skok, K.; Zidarič, T.; Orthaber, K.; Pristovnik, M.; Kostevšek, N.; Rožman, K.Ž.; Šturm, S.; Gradišnik, L.; Maver, U.; Maver, T. Novel Methacrylate-Based Multilayer Nanofilms with Incorporated FePt-Based Nanoparticles and the Anticancer Drug 5-Fluorouracil for Skin Cancer Treatment. Pharmaceutics 2022, 14, 689. [Google Scholar] [PubMed]
  57. Yu, N.; Luo, X.; Wei, T.; Zhang, Q.; Yu, J.; Wu, L.; Su, J.; Chen, M.; Huang, K.; Li, F. Dermabrasion combined with photodynamic therapy: A new option for the treatment of non-melanoma skin cancer. Lasers Med. Sci. 2022, 37, 1255–1263. [Google Scholar] [PubMed]
  58. Tang, X.; Chen, X.; Zhang, S.; Gu, X.; Wu, R.; Huang, T.; Zhou, Z.; Sun, C.; Ling, J.; Liu, M. Silk-Inspired In Situ Hydrogel with Anti-Tumor Immunity Enhanced Photodynamic Therapy for Melanoma and Infected Wound Healing. Adv. Funct. Mater. 2021, 31, 2101320. [Google Scholar]
  59. Lucena, S.R.; Salazar, N.; Gracia-Cazaña, T.; Zamarrón, A.; González, S.; Juarranz, Á.; Gilaberte, Y. Combined treatments with photodynamic therapy for non-melanoma skin cancer. Int. J. Mol. Sci. 2015, 16, 25912–25933. [Google Scholar]
  60. Kanekura, T.; Higashi, Y.; Kanzaki, T. Inhibitory effects of 9-cis-retinoic acid and pyrrolidinedithiocarbamate on cyclooxygenase (COX)-2 expression and cell growth in human skin squamous carcinoma cells. Cancer Lett. 2000, 161, 177–183. [Google Scholar] [CrossRef]
  61. Buckman, S.; Gresham, A.; Hale, P.; Hruza, G.; Anast, J.; Masferrer, J.; Pentland, A.P. COX-2 expression is induced by UVB exposure in human skin: Implications for the development of skin cancer. Carcinogenesis 1998, 19, 723–729. [Google Scholar]
  62. Fecker, L.F.; Stockfleth, E.; Braun, F.K.; Rodust, P.M.; Schwarz, C.; Köhler, A.; Leverkus, M.; Eberle, J. Enhanced death ligand-induced apoptosis in cutaneous SCC cells by treatment with diclofenac/hyaluronic acid correlates with downregulation of c-FLIP. J. Investig. Dermatol. 2010, 130, 2098–2109. [Google Scholar]
  63. Sligh, J.E., Jr. New therapeutic options for actinic keratosis and basal cell carcinoma. Semin. Cutan Med. Surg. 2014, 33, S76–S80. [Google Scholar] [CrossRef]
  64. Rosen, R.H.; Gupta, A.K.; Tyring, S.K. Dual mechanism of action of ingenol mebutate gel for topical treatment of actinic keratoses: Rapid lesion necrosis followed by lesion-specific immune response. J. Am. Acad. Dermatol. 2012, 66, 486–493. [Google Scholar] [PubMed]
  65. Kedei, N.; Lundberg, D.J.; Toth, A.; Welburn, P.; Garfield, S.H.; Blumberg, P.M. Characterization of the interaction of ingenol 3-angelate with protein kinase C. Cancer Res. 2004, 64, 3243–3255. [Google Scholar] [PubMed]
  66. Skeel, R.T.; Khleif, S.N. Handbook of Cancer Chemotherapy; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2011. [Google Scholar]
  67. Luke, J.J.; Schwartz, G.K. Chemotherapy in the management of advanced cutaneous malignant melanoma. Clin. Dermatol. 2013, 31, 290–297. [Google Scholar] [PubMed]
  68. Landi, M.M.; Ahmadi, S.A.; Akhgar, M.R.; Ghazanfari, D. Theoretical study of the tautomerization of Carmustine in a biological media as an anti-cancer drug. J. Part. Sci. Technol. 2021, 7, 51–57. [Google Scholar]
  69. Larkin, J.M.G.; Hughes, S.A.; Beirne, D.A.; Patel, P.M.; Gibbens, I.M.; Bate, S.C.; Thomas, K.; Eisen, T.G.; Gore, M.E. A Phase I/II study of lomustine and temozolomide in patients with cerebral metastases from malignant melanoma. Br. J. Cancer 2007, 96, 44–48. [Google Scholar] [CrossRef]
  70. Livshits, Z.; Rao, R.B.; Smith, S.W. An approach to chemotherapy-associated toxicity. Emerg. Med. Clin. 2014, 32, 167–203. [Google Scholar]
  71. Rowinsky, E.K.; Donehower, R.C.; Jones, R.J.; Tucker, R.W. Microtubule changes and cytotoxicity in leukemic cell lines treated with taxol. Cancer Res. 1988, 48, 4093–4100. [Google Scholar]
  72. Manfredi, J.J.; Horwitz, S.B. Taxol: An antimitotic agent with a new mechanism of action. Pharmacol. Ther. 1984, 25, 83–125. [Google Scholar]
  73. Mhaidat, N.M.; Wang, Y.; Kiejda, K.A.; Zhang, X.D.; Hersey, P. Docetaxel-induced apoptosis in melanoma cells is dependent on activation of caspase-2. Mol. Cancer Ther. 2007, 6, 752–761. [Google Scholar]
  74. Meyer, T.; Surber, C.; French, L.E.; Stockfleth, E. Resiquimod, a topical drug for viral skin lesions and skin cancer. Expert Opin. Investig. Drugs 2013, 22, 149–159. [Google Scholar] [CrossRef] [PubMed]
  75. Liner, K.; Brown, C.; McGirt, L.Y. Clinical potential of mechlorethamine gel for the topical treatment of mycosis fungoides-type cutaneous T-cell lymphoma: A review on current efficacy and safety data. Drug Des. Dev. Ther. 2018, 12, 241–254. [Google Scholar] [CrossRef]
  76. Minchinton, A.I.; Tannock, I.F. Drug penetration in solid tumours. Nat. Rev. Cancer 2006, 6, 583–592. [Google Scholar]
  77. Fujimura, T.; Kambayashi, Y.; Fujisawa, Y.; Hidaka, T.; Aiba, S. Tumor-associated macrophages: Therapeutic targets for skin cancer. Front. Oncol. 2018, 8, 3. [Google Scholar] [CrossRef] [PubMed]
  78. Georgescu, S.R.; Tampa, M.; Mitran, C.I.; Mitran, M.I.; Caruntu, C.; Caruntu, A.; Lupu, M.; Matei, C.; Constantin, C.; Neagu, M. Tumour microenvironment in skin carcinogenesis. Tumor Microenviron. Organs 2020, 1226, 123–142. [Google Scholar]
  79. Castano, A.P.; Mroz, P.; Hamblin, M.R. Photodynamic therapy and anti-tumour immunity. Nat. Rev. Cancer 2006, 6, 535–545. [Google Scholar] [PubMed] [Green Version]
  80. Ghafouri-Fard, S.; Ghafouri-Fard, S. Immunotherapy in nonmelanoma skin cancer. Immunotherapy 2012, 4, 499–510. [Google Scholar] [CrossRef]
  81. Thambi, T.; Li, Y.; Lee, D.S. Injectable hydrogels for sustained release of therapeutic agents. J. Control. Release 2017, 267, 57–66. [Google Scholar] [CrossRef]
  82. Phan, V.H.G.; Le, T.M.D.; Janarthanan, G.; Ngo, P.-K.T.; Lee, D.S.; Thambi, T. Development of bioresorbable smart injectable hydrogels based on thermo-responsive copolymer integrated bovine serum albumin bioconjugates for accelerated healing of excisional wounds. J. Ind. Eng. Chem. 2021, 96, 345–355. [Google Scholar] [CrossRef]
  83. Duong, H.T.T.; Thambi, T.; Yin, Y.; Kim, S.H.; Nguyen, T.L.; Phan, V.H.G.; Kim, J.; Jeong, J.H.; Lee, D.S. Degradation-regulated architecture of injectable smart hydrogels enhances humoral immune response and potentiates antitumor activity in human lung carcinoma. Biomaterials 2020, 230, 119599. [Google Scholar] [CrossRef]
  84. Giang Phan, V.H.; Duong, H.T.T.; Thambi, T.; Nguyen, T.L.; Turabee, M.H.; Yin, Y.; Kim, S.H.; Kim, J.; Jeong, J.H.; Lee, D.S. Modularly engineered injectable hybrid hydrogels based on protein-polymer network as potent immunologic adjuvant in vivo. Biomaterials 2019, 195, 100–110. [Google Scholar] [CrossRef] [PubMed]
  85. Luu, C.H.; Nguyen, G.; Le, T.-T.; Nguyen, T.-M.N.; Giang Phan, V.H.; Murugesan, M.; Mathiyalagan, R.; Jing, L.; Janarthanan, G.; Yang, D.C.; et al. Graphene Oxide-Reinforced Alginate Hydrogel for Controlled Release of Local Anesthetics: Synthesis, Characterization, and Release Studies. Gels 2022, 8, 246. [Google Scholar] [CrossRef] [PubMed]
  86. Buwalda, S.J.; Boere, K.W.M.; Dijkstra, P.J.; Feijen, J.; Vermonden, T.; Hennink, W.E. Hydrogels in a historical perspective: From simple networks to smart materials. J. Control. Release 2014, 190, 254–273. [Google Scholar] [CrossRef] [PubMed]
  87. 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]
  88. Thambi, T.; Giang Phan, V.H.; Kim, S.H.; Duy Le, T.M.; Duong, H.T.T.; Lee, D.S. Smart injectable biogels based on hyaluronic acid bioconjugates finely substituted with poly(β-amino ester urethane) for cancer therapy. Biomater. Sci. 2019, 7, 5424–5437. [Google Scholar] [CrossRef]
  89. Kim, S.H.; Thambi, T.; Giang Phan, V.H.; Lee, D.S. Modularly engineered alginate bioconjugate hydrogel as biocompatible injectable scaffold for in situ biomineralization. Carbohydr. Polym. 2020, 233, 115832. [Google Scholar] [CrossRef]
  90. Zhu, J.; Marchant, R.E. Design properties of hydrogel tissue-engineering scaffolds. Expert Rev. Med. Devices 2011, 8, 607–626. [Google Scholar] [CrossRef]
  91. Sun, T.L.; Kurokawa, T.; Kuroda, S.; Ihsan, A.B.; Akasaki, T.; Sato, K.; Haque, M.; Nakajima, T.; Gong, J.P. Physical hydrogels composed of polyampholytes demonstrate high toughness and viscoelasticity. Nat. Mater. 2013, 12, 932–937. [Google Scholar] [CrossRef]
  92. Pang, X.; Wu, J.; Chu, C.-C.; Chen, X. Development of an arginine-based cationic hydrogel platform: Synthesis, characterization and biomedical applications. Acta Biomater. 2014, 10, 3098–3107. [Google Scholar] [CrossRef]
  93. Gil, M.S.; Cho, J.; Thambi, T.; Giang Phan, V.H.; Kwon, I.; Lee, D.S. Bioengineered robust hybrid hydrogels enrich the stability and efficacy of biological drugs. J. Control. Release 2017, 267, 119–132. [Google Scholar] [CrossRef]
  94. Anwary, M.; Kumar, P.; du Toit, L.C.; Choonara, Y.E.; Pillay, V. Polymeric, injectable, intravitreal hydrogel devices for posterior segment applications and interventions. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1074–1081. [Google Scholar] [CrossRef] [PubMed]
  95. Phan, V.H.G.; Thambi, T.; Kim, B.S.; Huynh, D.P.; Lee, D.S. Engineering highly swellable dual-responsive protein-based injectable hydrogels: The effects of molecular structure and composition in vivo. Biomater. Sci. 2017, 5, 2285–2294. [Google Scholar] [CrossRef] [PubMed]
  96. Cipriano, B.H.; Banik, S.J.; Sharma, R.; Rumore, D.; Hwang, W.; Briber, R.M.; Raghavan, S.R. Superabsorbent hydrogels that are robust and highly stretchable. Macromolecules 2014, 47, 4445–4452. [Google Scholar] [CrossRef]
  97. Omidian, H.; Rocca, J.G.; Park, K. Advances in superporous hydrogels. J. Control. Release 2005, 102, 3–12. [Google Scholar] [CrossRef]
  98. Lee, J.H. Injectable hydrogels delivering therapeutic agents for disease treatment and tissue engineering. Biomater. Res. 2018, 22, 27. [Google Scholar] [CrossRef]
  99. Turabee, M.H.; Thambi, T.; Duong, H.T.T.; Jeong, J.H.; Lee, D.S. A pH- and temperature-responsive bioresorbable injectable hydrogel based on polypeptide block copolymers for the sustained delivery of proteins in vivo. Biomater. Sci. 2018, 6, 661–671. [Google Scholar] [CrossRef]
  100. Turabee, M.H.; Thambi, T.; Lym, J.S.; Lee, D.S. Bioresorbable polypeptide-based comb-polymers efficiently improves the stability and pharmacokinetics of proteins in vivo. Biomater. Sci. 2017, 5, 837–848. [Google Scholar] [CrossRef]
  101. Harrison, I.P.; Spada, F. Hydrogels for Atopic Dermatitis and Wound Management: A Superior Drug Delivery Vehicle. Pharmaceutics 2018, 10, 71. [Google Scholar] [CrossRef]
  102. Kim, S.H.; Thambi, T.; Lym, J.S.; Giang Phan, V.H.; Lee, D.S. Tunable Engineering of Heparinized Injectable Hydrogels for Affinity-Based Sustained Delivery of Bioactive Factors. Macromol. Mater. Eng. 2019, 304, 1900279. [Google Scholar] [CrossRef]
  103. Thambi, T.; Phan, V.H.G.; Lee, D.S. Stimuli-Sensitive Injectable Hydrogels Based on Polysaccharides and Their Biomedical Applications. Macromol. Rapid Commun. 2016, 37, 1881–1896. [Google Scholar] [CrossRef]
  104. 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]
  105. Thambi, T.; Giang Phan, V.H.; Kim, S.H.; Duy Le, T.M.; Lee, D.S. Hyaluronic acid decorated pH- and temperature-induced injectable bioconjugates for sustained delivery of bioactive factors and highly efficient wound regeneration. New J. Chem. 2019, 43, 18979–18982. [Google Scholar] [CrossRef]
  106. Phan, V.H.G.; Murugesan, M.; Manivasagan, P.; Nguyen, T.L.; Phan, T.-H.; Luu, C.H.; Ho, D.-K.; Li, Y.; Kim, J.; Lee, D.S.; et al. Injectable Hydrogel Based on Protein-Polyester Microporous Network as an Implantable Niche for Active Cell Recruitment. Pharmaceutics 2022, 14, 709. [Google Scholar] [CrossRef] [PubMed]
  107. Phan, V.H.G.; Lee, E.; Maeng, J.H.; Thambi, T.; Kim, B.S.; Lee, D.; Lee, D.S. Pancreatic cancer therapy using an injectable nanobiohybrid hydrogel. RSC Adv. 2016, 6, 41644–41655. [Google Scholar] [CrossRef]
  108. Peppas, N.A.; Van Blarcom, D.S. Hydrogel-based biosensors and sensing devices for drug delivery. J. Control. Release 2016, 240, 142–150. [Google Scholar] [CrossRef]
  109. Calvert, P. Hydrogels for soft machines. Adv. Mater. 2009, 21, 743–756. [Google Scholar] [CrossRef]
  110. Kirschner, C.M.; Anseth, K.S. Hydrogels in healthcare: From static to dynamic material microenvironments. Acta Mater. 2013, 61, 931–944. [Google Scholar] [CrossRef]
  111. Chittasupho, C.; Angklomklew, J.; Thongnopkoon, T.; Senavongse, W.; Jantrawut, P.; Ruksiriwanich, W. Biopolymer hydrogel scaffolds containing doxorubicin as a localized drug delivery system for inhibiting lung cancer cell proliferation. Polymers 2021, 13, 3580. [Google Scholar] [CrossRef]
  112. Javanbakht, S.; Namazi, H. Doxorubicin loaded carboxymethyl cellulose/graphene quantum dot nanocomposite hydrogel films as a potential anticancer drug delivery system. Mater. Sci. Eng. C 2018, 87, 50–59. [Google Scholar] [CrossRef]
  113. Zheng, Y.; Wang, W.; Zhao, J.; Wu, C.; Ye, C.; Huang, M.; Wang, S. Preparation of injectable temperature-sensitive chitosan-based hydrogel for combined hyperthermia and chemotherapy of colon cancer. Carbohydr. Polym. 2019, 222, 115039. [Google Scholar] [CrossRef]
  114. Xin, H.; Naficy, S. Drug delivery based on stimuli-responsive injectable hydrogels for breast cancer therapy: A review. Gels 2022, 8, 45. [Google Scholar] [CrossRef] [PubMed]
  115. Hwang, J.; Jin, J.-O. Attachable hydrogel containing indocyanine green for selective photothermal therapy against melanoma. Biomolecules 2020, 10, 1124. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, J.; Xu, W.; Zhao, Z.; Ling, G.; Zhang, P. Intelligent nanocomposite hydrogels with simultaneous photothermal antitumor and antibacterial efficacy for cutaneous melanoma treatment. Compos. Part B Eng. 2022, 243, 110130. [Google Scholar] [CrossRef]
  117. Jung, J.M.; Kim, S.H.; Giang Phan, V.H.; Thambi, T.; Lee, D.S. Therapeutic effects of boronate ester cross-linked injectable hydrogels for the treatment of hepatocellular carcinoma. Biomater. Sci. 2021, 9, 7275–7286. [Google Scholar] [CrossRef]
  118. Narayanaswamy, R.; Torchilin, V.P. Hydrogels and Their Applications in Targeted Drug Delivery. Molecules 2019, 24, 603. [Google Scholar] [CrossRef] [PubMed]
  119. Prasannan, A.; Tsai, H.-C.; Hsiue, G.-H. Formulation and evaluation of epinephrine-loaded poly (acrylic acid-co-N-isopropylacrylamide) gel for sustained ophthalmic drug delivery. React. Funct. Polym. 2018, 124, 40–47. [Google Scholar] [CrossRef]
  120. Carthew, J.; Donderwinkel, I.; Shrestha, S.; Truong, V.X.; Forsythe, J.S.; Frith, J.E. In situ miRNA delivery from a hydrogel promotes osteogenesis of encapsulated mesenchymal stromal cells. Acta Biomater. 2020, 101, 249–261. [Google Scholar] [CrossRef]
  121. Xu, L.; Bai, X.; Yang, J.; Li, J.; Xing, J.; Yuan, H.; Xie, J.; Li, J. Preparation and characterisation of a gellan gum-based hydrogel enabling osteogenesis and inhibiting Enterococcus faecalis. Int. J. Biol. Macromol. 2020, 165, 2964–2973. [Google Scholar] [CrossRef]
  122. Zhang, Z.; Wei, X.; Gao, J.; Zhao, Y.; Zhao, Y.; Guo, L.; Chen, C.; Duan, Z.; Li, P.; Wei, L. Intra-articular injection of cross-linked hyaluronic acid-dexamethasone hydrogel attenuates osteoarthritis: An experimental study in a rat model of osteoarthritis. Int. J. Mol. Sci. 2016, 17, 411. [Google Scholar] [CrossRef]
  123. Diniz, F.R.; Maia, R.C.A.; Rannier Andrade, L.; Andrade, L.N.; Vinicius Chaud, M.; da Silva, C.F.; Corrêa, C.B.; de Albuquerque Junior, R.L.C.; Pereira da Costa, L.; Shin, S.R. Silver nanoparticles-composing alginate/gelatine hydrogel improves wound healing in vivo. Nanomaterials 2020, 10, 390. [Google Scholar] [CrossRef]
  124. Donnelly, R.F.; Singh, T.R.R.; Morrow, D.I.; Woolfson, A.D. Microneedle-Mediated Transdermal and Intradermal Drug Delivery; John and Wiley and Sons: Hoboken, NJ, USA, 2012. [Google Scholar]
  125. Brown, M.; Jones, S. Hyaluronic acid: A unique topical vehicle for the localized delivery of drugs to the skin. J. Eur. Acad. Dermatol. Venereol. 2005, 19, 308–318. [Google Scholar] [CrossRef] [PubMed]
  126. Sim, S.; Figueiras, A.; Veiga, F. Modular Hydrogels for Drug Delivery. J. Biomater. Nanobiotechnol. 2012, 3, 18938. [Google Scholar]
  127. Langer, R. New methods of drug delivery. Science 1990, 249, 1527–1533. [Google Scholar] [CrossRef]
  128. Overstreet, D.J.; Dutta, D.; Stabenfeldt, S.E.; Vernon, B.L. Injectable hydrogels. J. Polym. Sci. Part B Polym. Phys. 2012, 50, 881–903. [Google Scholar] [CrossRef]
  129. Harush-Frenkel, O.; Rozentur, E.; Benita, S.; Altschuler, Y. Surface Charge of Nanoparticles Determines Their Endocytic and Transcytotic Pathway in Polarized MDCK Cells. Biomacromolecules 2008, 9, 435–443. [Google Scholar] [CrossRef] [PubMed]
  130. Liu, W.; Zhou, X.; Mao, Z.; Yu, D.; Wang, B.; Gao, C. Uptake of hydrogel particles with different stiffness and its influence on HepG2 cell functions. Soft Matter 2012, 8, 9235–9245. [Google Scholar] [CrossRef]
  131. Thambi, T.; Deepagan, V.G.; Yoon, H.Y.; Han, H.S.; Kim, S.-H.; Son, S.; Jo, D.-G.; Ahn, C.-H.; Suh, Y.D.; Kim, K.; et al. Hypoxia-responsive polymeric nanoparticles for tumor-targeted drug delivery. Biomaterials 2014, 35, 1735–1743. [Google Scholar] [CrossRef]
  132. Thambi, T.; Yoon, H.Y.; Kim, K.; Kwon, I.C.; Yoo, C.K.; Park, J.H. Bioreducible Block Copolymers Based on Poly(Ethylene Glycol) and Poly(γ-Benzyl l-Glutamate) for Intracellular Delivery of Camptothecin. Bioconjugate Chem. 2011, 22, 1924–1931. [Google Scholar] [CrossRef]
  133. Lei, T.; Srinivasan, S.; Tang, Y.; Manchanda, R.; Nagesetti, A.; Fernandez-Fernandez, A.; McGoron, A.J. Comparing cellular uptake and cytotoxicity of targeted drug carriers in cancer cell lines with different drug resistance mechanisms. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 324–332. [Google Scholar] [CrossRef]
  134. Jang, J.Y.; Le, T.M.D.; Ko, J.H.; Ko, Y.-J.; Lee, S.M.; Kim, H.J.; Jeong, J.H.; Thambi, T.; Lee, D.S.; Son, S.U. Triple-, Double-, and Single-Shelled Hollow Spheres of Sulfonated Microporous Organic Network as Drug Delivery Materials. Chem. Mater. 2019, 31, 300–304. [Google Scholar] [CrossRef]
  135. Banquy, X.; Suarez, F.; Argaw, A.; Rabanel, J.-M.; Grutter, P.; Bouchard, J.-F.; Hildgen, P.; Giasson, S. Effect of mechanical properties of hydrogel nanoparticles on macrophage cell uptake. Soft Matter 2009, 5, 3984–3991. [Google Scholar] [CrossRef]
  136. Kalal, B.S.; Upadhya, D.; Pai, V.R. Chemotherapy resistance mechanisms in advanced skin cancer. Oncol. Rev. 2017, 11, 326. [Google Scholar] [CrossRef]
  137. Sepantafar, M.; Maheronnaghsh, R.; Mohammadi, H.; Radmanesh, F.; Hasani-Sadrabadi, M.M.; Ebrahimi, M.; Baharvand, H. Engineered hydrogels in cancer therapy and diagnosis. Trends Biotechnol. 2017, 35, 1074–1087. [Google Scholar] [CrossRef] [PubMed]
  138. Manivasagan, P.; Joe, A.; Han, H.-W.; Thambi, T.; Selvaraj, M.; Chidambaram, K.; Kim, J.; Jang, E.-S. Recent advances in multifunctional nanomaterials for photothermal-enhanced Fenton-based chemodynamic tumor therapy. Mater. Today Bio 2022, 13, 100197. [Google Scholar] [CrossRef]
  139. Gil, M.S.; Thambi, T.; Phan, V.H.G.; Kim, S.H.; Lee, D.S. Injectable hydrogel-incorporated cancer cell-specific cisplatin releasing nanogels for targeted drug delivery. J. Mater. Chem. B 2017, 5, 7140–7152. [Google Scholar] [CrossRef]
  140. Nguyen, D.T.; Phan, V.H.G.; Lee, D.S.; Thambi, T.; Huynh, D.P. Bioresorbable pH- and temperature-responsive injectable hydrogels-incorporating electrosprayed particles for the sustained release of insulin. Polym. Degrad. Stab. 2019, 162, 36–46. [Google Scholar] [CrossRef]
  141. Phan, V.H.G.; Thambi, T.; Duong, H.T.T.; Lee, D.S. Poly(amino carbonate urethane)-based biodegradable, temperature and pH-sensitive injectable hydrogels for sustained human growth hormone delivery. Sci. Rep. 2016, 6, 29978. [Google Scholar] [CrossRef] [Green Version]
  142. Zhao, Y.; Yan, H.; Qiao, S.; Zhang, L.; Wang, T.; Meng, Q.; Chen, X.; Lin, F.-H.; Guo, K.; Li, C. Hydrogels bearing bioengineered mimetic embryonic microenvironments for tumor reversion. J. Mater. Chem. B 2016, 4, 6183–6191. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, Z.; Deng, J.; Cao, J.; Wu, H.; Feng, G.; Zhang, R.; Ran, B.; Hu, K.; Cao, H.; Zhu, X. Nano-hydroxyapatite-evoked immune response synchronized with controllable immune adjuvant release for strengthening melanoma-specific growth inhibition. Acta Biomater. 2022, 145, 159–171. [Google Scholar] [CrossRef]
  144. Karavasili, C.; Andreadis, D.A.; Katsamenis, O.L.; Panteris, E.; Anastasiadou, P.; Kakazanis, Z.; Zoumpourlis, V.; Markopoulou, C.K.; Koutsopoulos, S.; Vizirianakis, I.S. Synergistic antitumor potency of a self-assembling peptide hydrogel for the local co-delivery of doxorubicin and curcumin in the treatment of head and neck cancer. Mol. Pharm. 2019, 16, 2326–2341. [Google Scholar] [CrossRef]
  145. Wu, X.; Wu, Y.; Ye, H.; Yu, S.; He, C.; Chen, X. Interleukin-15 and cisplatin co-encapsulated thermosensitive polypeptide hydrogels for combined immuno-chemotherapy. J. Control. Release 2017, 255, 81–93. [Google Scholar] [CrossRef] [PubMed]
  146. Lv, Q.; He, C.; Quan, F.; Yu, S.; Chen, X. DOX/IL-2/IFN-γ co-loaded thermo-sensitive polypeptide hydrogel for efficient melanoma treatment. Bioact. Mater. 2018, 3, 118–128. [Google Scholar] [CrossRef] [PubMed]
  147. Fujii, H.; Shin-Ya, M.; Takeda, S.; Hashimoto, Y.; Mukai, S.A.; Sawada, S.I.; Adachi, T.; Akiyoshi, K.; Miki, T.; Mazda, O. Cycloamylose-nanogel drug delivery system-mediated intratumor silencing of the vascular endothelial growth factor regulates neovascularization in tumor microenvironment. Cancer Sci. 2014, 105, 1616–1625. [Google Scholar] [CrossRef] [PubMed]
  148. Li, Y.; Fang, M.; Zhang, J.; Wang, J.; Song, Y.; Shi, J.; Li, W.; Wu, G.; Ren, J.; Wang, Z. Hydrogel dual delivered celecoxib and anti-PD-1 synergistically improve antitumor immunity. Oncoimmunology 2016, 5, e1074374. [Google Scholar] [CrossRef] [PubMed]
  149. Zhou, Y.; Ye, T.; Ye, C.; Wan, C.; Yuan, S.; Liu, Y.; Li, T.; Jiang, F.; Lovell, J.F.; Jin, H. Secretions from hypochlorous acid-treated tumor cells delivered in a melittin hydrogel potentiate cancer immunotherapy. Bioact. Mater. 2022, 9, 541–553. [Google Scholar] [CrossRef]
  150. Sun, R.; Chen, Y.; Yang, Q.; Zhang, W.; Guo, L.; Feng, M. Polysaccharide hydrogels regulate macrophage polarization and enhance the anti-tumor efficacy of melanoma. Int. J. Pharm. 2022, 613, 121390. [Google Scholar] [CrossRef]
  151. Zhang, R.; Zhang, D.; Sun, X.; Song, X.; Yan, K.C.; Liang, H. Polyvinyl alcohol/gelatin hydrogels regulate cell adhesion and chromatin accessibility. Int. J. Biol. Macromol. 2022, 219, 672–684. [Google Scholar] [CrossRef]
Figure 1. Different layers of the skin and types of skin cancer.
Figure 1. Different layers of the skin and types of skin cancer.
Gels 08 00560 g001
Figure 2. Gene regulations on hydrogel mediated drug delivery treatment for skin cancers, based on the recent reports. The upregulation of Caspase 3, Bim, BAD, CXCL9, KLF14, p53, p21 and downregulation of Bcl-2, CDK2, CDC25A were mainly reported after the hydrogel-based drug delivery for skin cancers as apoptosis and cell proliferation regulators.
Figure 2. Gene regulations on hydrogel mediated drug delivery treatment for skin cancers, based on the recent reports. The upregulation of Caspase 3, Bim, BAD, CXCL9, KLF14, p53, p21 and downregulation of Bcl-2, CDK2, CDC25A were mainly reported after the hydrogel-based drug delivery for skin cancers as apoptosis and cell proliferation regulators.
Gels 08 00560 g002
Table 1. Genes and pathways involved in melanoma.
Table 1. Genes and pathways involved in melanoma.
GenePathwayRegulationReferences
BRAFRAS–RAF–MEK–ERK
mitogen-activated protein kinase (MAPK) pathway
Somatic missense mutation-valine-to-glutamic acid substitution at position 599 (V599E)[21]
NRASRAS–RAF–MEK–ERK
MAPK pathway
Substitution mutations[22]
KITmitogen-activated protein (MAP) kinase and phosphatidylinositol 3 (PI3) kinase pathways, PI3K–AKT–mTOR, JAK–STATSomatic mutations- on exon 11 (L576P or exon 13 (K642E)[23,24]
GNAQMAPK pathwaysSomatic mutations—glutamine at position 209 (Q209) is mutated to either leucine or proline[25]
GNA11MAPK pathwaySomatic mutations—glutamine at position 209 (Q209) is mutated to either leucine or proline[26]
CDKN2A (p14ARF) MDM2–p53Deletion[27,28]
CDKN2A (p16INK4A)p16INK4A–cyclin D/CDK4–RB checkpoint‘G’ TO ‘A’ transition at the first nucleotide of the splice donor site of intron 2[28,29]
PTENoncogenic phosphatidylinositol-3-kinase (PI3K) signaling pathwayDeletion or mutation leads to constitutive activation of this pathway[30]
LKB1LKB1–AMPK‘C’ to ‘T’ transition, resulting in the substitution of the normal glutamine codon (CAG) with a premature termination codon (TAG)[31,32]
MITFMITF–PGC1a
Transcription, lineage, cell cycle
Amplification/germline missense substitution[33,34]
NF1/neurofibrominPI3K and MAPK pathwaysMutation [35]
MYC Amplification[36]
Cyclin D1RAS/MAPK pathwaysAmplification[37,38]
CDK4Cell cycle, G1/S cyclin-dependent kinaseAmplification or Point mutation[39]
HDM2P53Amplification[40]
PIK3CAPI3K–AKT–mTORMissense mutations[41]
AKT1,AKT2, AKT3PI3K–AKT–mTOROncogenic mutation[42]
ERBB4Receptor tyrosine kinasesAmplification[43]
fibroblast growth factor receptor 3 (FGFR3)Ras/MAPKAmplification, gain-of-function mutations[44,45]
METPI3K, MAPKAmplification/single-nucleotide variations[46]
Table 2. Current drugs used for skin cancer.
Table 2. Current drugs used for skin cancer.
DrugOriginMolecular Weight (g/mol)StructureRoute of AdministrationEffectReferences
AlitretinoinSynthetic/Natural300.4Gels 08 00560 i001TopicalInhibit cyclooxygenase (COX)-2 expression, suppress cell growth[5,60]
Diclofenac sodiumSynthetic318.1Gels 08 00560 i002TopicalCycloxygenase-2 enzyme overexpression & increase apoptosis[61,62]
FluorouracilSynthetic130.1Gels 08 00560 i003TopicalEnzyme responsible for synthesis of thymidine, one of the pyrimidine nucleosides of DNA has been inhibited[5]
ImiquimodSynthetic240.3Gels 08 00560 i004TopicalThrough toll-like receptor 7 (TLR7) an immune response which upregulates cytokines and consequently leads to apoptosis has been triggered [5,63]
Ingenol mebutateSynthetic/Natural430.5Gels 08 00560 i005TopicalUpregulate rapid lesion necro sis and neutrophil-mediated cellular cytotoxicity, Modulation of protein kinase C isoforms[64,65]
CisplatinSynthetic301.1Gels 08 00560 i006Intravenous DNA cross linking agents[66]
DacarbazineSynthetic182.18Gels 08 00560 i007IntravenousAlkylating agents that damage DNA by introducing alkyl groups to guanine bases, eventually causing cell death via apoptosis and other cell death mechanisms[7,67]
TemozolomideSynthetic194.151Gels 08 00560 i008intravenous infusionAlkylating agents that damage DNA by introducing alkyl groups to guanine bases, eventually causing cell death via apoptosis and other cell death mechanisms[67]
CarmustineSynthetic214.05Gels 08 00560 i009intravenous infusionDNA binding and DNA alkylation by a nitrogenous base within a duplex[68]
LomustineSynthetic233.69Gels 08 00560 i010oralAlkylation of the O6 position of guanine-containing bases in DNA and the enzyme O6 -alkylguanine transferase mediates[69]
VincristineNatural824.958Gels 08 00560 i011intravenous infusionInhibition of microtubule polymerization; microtubule destabilization[70]
VinblastineNatural810.974Gels 08 00560 i012intravenous infusionInhibition of microtubule polymerization; microtubule destabilization[70]
CarboplatinSynthetic371.249Gels 08 00560 i013intravenous infusionDNA alkylation: formation of DNA crosslinks and adducts; inhibition of DNA synthesis[7,70]
Taxol/PaclitaxelNatural853.906Gels 08 00560 i014intravenous infusionInteracts with microtubules causing polymerization and stabilization of the tubulin polymer which prevents successful completion of cell division[71,72]
DocetaxelNatural807.879Gels 08 00560 i015intravenous infusionBinds to β-tubulins in the cell and causes their polymerization, induces caspase-2 dependent apoptosis[73]
ResiquimodSynthetic314.4Gels 08 00560 i016TopicalStimulates immune responses through toll-like receptors (TLR) 7 and 8 dependent pathway activation[74]
MechlorethamineSynthetic156.05Gels 08 00560 i017TopicalCancer cell growth postulated by DNA cross-linking and depurination, or abnormal base paring. [75]
Table 3. Recent studies on the gene regulations upon hydrogel-based drug delivery system for skin cancers.
Table 3. Recent studies on the gene regulations upon hydrogel-based drug delivery system for skin cancers.
HydrogelGenePathwayEffectIn Vivo/In VitroCancerReference
Hyaluronic acid scaffold loaded with Nodal Signaling Crypto 1 receptor antibodies (2B11)Kit, Itga4, Hapln1, Irf8, Trail, Casp1, Aim2, and Irf1Apoptosis pathways, MAPK pathways and PI3K–AKT–mTOR, JAK–STATUpregulation of tumor suppressor genes and the downregulation of oncogenesIn vivo and in vitro Melanoma[142]
mPEG-b-PELG hydrogel encapsulating f interleukin-15 (IL-15) and cisplatinCyclin A2, CDK2, and Cdc25A Cell cycleSignificant decrease in the expressions of genes and cell cycle arrestEx vivoMelanoma[145]
Poly Lactic-co-Glycolic Acid (PLGA)-polyethylene glycol-PLGA hydrogel encapsulating, nano-hydroxyapatite and granulocyte-macrophage colony-stimulating factorE2Fs family genes and PLK1, KLF14, KLF11Cell cycleCell cycle arrested in the G2/M phase, apoptosisIn vitroMelanoma[143]
PELG-PEG-PELG loaded with DOX, IL-2, and IFN-gBcl-2
Caspase 3
Janus kinase, JAK/STAT and mitochondrial signal pathwaysInduces apoptosis In vitroMelanoma[146]
Olesterol-bearing cycloamylose with spermine group nanogel carrying VEGF-specific short interfering RNAVascular endothelial growth factor (VEGF)Angiogenesis In vitro [147]
peptide hydrogel (ac-(RADA)4-CONH2) loaded with curcumin and doxorubicinp53, p21, BAX, BAD, Cdk2, Bcl-2, c-myc and CyclD1Apoptosis pathways,High rate of apoptosisIn vitroHead and neck squamous cell carcinoma[144]
Alginate hydrogel bearing loaded with anti-PD-1 monoclonal antibody and celecoxibIL-1b, IL-6, CXCL9 and CXCL10Programmed death 1 (PD-1) signaling pathwayIncreases the expression of two anti-angiogenic chemokines and suppresses the intra tumoral production of interleukin (IL)-1, IL-6, and cycloxygenase-2 (COX2)In vitro Melanoma[148]
RADA24-melittin fusion peptide hydrogel loaded with cell-derived secretions from cells exposed to HOClIFN-α, IFN-β, IL-6cGAS-STING pathway and PD-1 signaling pathwayIncreased tumor cell death, cytotoxic T lymphocyte infiltration, and tumor-associated macrophage reprogramming towards an M1 phenotypeIn vitro Melanoma [149]
N-succinyl chitosan and oxidized dextran hydrogel loaded with doxorubicinCD206, Arginase-1, TNF-α and Inosp65 NF-kappaB and P53 pathwayInduced macrophages to produce anti-tumorigenic cytokines such as TNF-α, iNOS, IL-6 and IL-1βIn vivo and in vitroMelanoma [150]
Polyvinyl alcohol/gelatinmechanotransduction related genes and transposase-accessible chromatinMAPK pathway and MKL1/SRF pathwayPoor cell adhesion and increased chromatin accessibilityin vitromelanoma[151]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mathiyalagan, R.; Kariyarath Valappil, A.; Yang, D.C.; Kang, S.C.; Thambi, T. Gene Regulations upon Hydrogel-Mediated Drug Delivery Systems in Skin Cancers—An Overview. Gels 2022, 8, 560. https://doi.org/10.3390/gels8090560

AMA Style

Mathiyalagan R, Kariyarath Valappil A, Yang DC, Kang SC, Thambi T. Gene Regulations upon Hydrogel-Mediated Drug Delivery Systems in Skin Cancers—An Overview. Gels. 2022; 8(9):560. https://doi.org/10.3390/gels8090560

Chicago/Turabian Style

Mathiyalagan, Ramya, Anjali Kariyarath Valappil, Deok Chun Yang, Se Chan Kang, and Thavasyappan Thambi. 2022. "Gene Regulations upon Hydrogel-Mediated Drug Delivery Systems in Skin Cancers—An Overview" Gels 8, no. 9: 560. https://doi.org/10.3390/gels8090560

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