Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Pharmacol., 26 August 2025

Sec. Pharmacology of Anti-Cancer Drugs

Volume 16 - 2025 | https://doi.org/10.3389/fphar.2025.1641838

This article is part of the Research TopicPhytochemical Interventions to CancerView all 7 articles

Natural products as therapeutics for malignant melanoma: preclinical evidence and mechanism

Hongjin Gao&#x;Hongjin Gao1Jianli Huang&#x;Jianli Huang1Dengfeng ZhangDengfeng Zhang1Sixuan LiSixuan Li1Shengzhi LongShengzhi Long2Yuesi Qin
Yuesi Qin1*
  • 1Chengdu Integrated TCM & Western Medicine Hospital, Chengdu, Sichuan, China
  • 2Dermatology Department of Bijie Traditional Chinese Medicine Hospital, Guiyang, Guizhou, China

Melanoma is one of the most common malignancies among fair-skinned populations. Natural products, a diverse group of bioactive compounds derived from plants and animals, have demonstrated inhibitory effects on melanoma growth, invasion, and metastasis. This review summarizes the mechanisms through which natural products inhibit melanoma progression and metastasis. These compounds are categorized based on their mechanisms of action. Many natural products have been found to induce apoptosis in melanoma cells through various signaling pathways. For instance, rhodopsin and the triazolylpeptidyl penicillin derivative TAP7f suppress the Wnt/β-catenin signaling pathway, thereby reducing melanoma cell proliferation and migration. Resveratrol and vitamin E delta-tocotrienol (δ-TT) inhibit caspase-dependent mitochondrial and endoplasmic reticulum stress pathways, inducing apoptosis in melanoma cells. Shikonin and plumbagin exert their antitumor effects through the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling pathway. In addition, natural products such as silymarin, capsaicin, and ursolic acid exhibit multi-targeted anticancer effects with high efficiency and low toxicity by modulating various signaling pathways. These findings highlight the ability of natural compounds to regulate multiple biological targets, offering new directions and potential clinical applications in melanoma therapy. Natural product–based drug development holds great promise for overcoming current limitations in cancer treatment.

1 Introduction

Random accumulation of cellular mutations or genetic defects can transform normal melanocytes into malignant melanoma. Although malignant melanoma accounts for less than 5% of all skin cancers, it is responsible for the majority of skin cancer–related deaths (Wilczak et al., 2025). The incidence of malignant melanoma varies by population, based on the current incidence rate, it is estimated that by 2040, the number of new melanoma cases will increase by 50%. According to 2020 data, the melanoma-related mortality rate is projected to rise by 68%, with the highest incidence observed in white populations (Long et al., 2023; Connor et al., 2025). Cutaneous malignant melanoma is growing faster than any other cancer and thus poses a significant health threat worldwide.

Most melanomas arise from somatic mutations that are acquired later in life. Several recently known key genetic mutations in melanoma, such as BRAF, NRAS, PTEN, MITF, CDKN2A, KIT, and TP53, have been described as crucial factors in melanoma development (Zhang et al., 2016; Palmieri et al., 2018; Castellani et al., 2023). Approximately 50% of malignant melanomas have mutations in BRAF, which promote melanin production through conformational activation of the MAPK (RAS/RAF/MEK/ERK) signaling pathway. This mediates several phenomena, including cell proliferation, differentiation, and secretion of signaling molecules associated with the appearance and progression of melanoma (Czarnecka et al., 2020; Ottaviano et al., 2021). Mitogen-activated protein kinase (MAPK) signaling pathway is one of the melanoma’s most common mutational pathways. It was estimated that 70% of melanomas contained mutations in the MAPK signaling pathway (Reddy et al., 2017; Cheng et al., 2018; Davis et al., 2019), and 50% of melanomas contained activated BRAF (mostly V600E) mutations (Scolyer et al., 2011), another 14%–20% of melanomas had NRAS mutations (Yakovian et al., 2021), and 2% had CKIT mutations (Kim and Kim, 2024), as shown in Figure 1.

Figure 1
Diagram illustrating signaling pathways in a cell, focusing on growth factor signaling and cell cycle control. Key components include GPCR, GNAQ/11, PI3K, c-KIT, RTKs, and various mutations like AKT^mut and P53^mut, showing interactions and regulatory impacts on cell functions such as proliferation and survival. The pathways lead to transcriptional changes in the nucleus.

Figure 1. The occurrence and development of melanoma are closely related to multiple signaling pathway mechanisms.

Surgical resection remains the preferred treatment for melanoma diagnosed at an early stage, before metastasis has occurred. However, once metastasis occurs, alternative therapeutic strategies are required. Chemotherapy has traditionally been used to alleviate symptoms, control tumor progression, or, in rare cases, achieve remission. It relies on cytotoxic agents to inhibit the abnormal proliferation of cancer cells or slow their overall growth rate (Long et al., 2023). Metastatic melanoma chemotherapy drugs include dacarbazine, paclitaxel, platinum compounds, and temozolomide (Megahed and Koon, 2014). Therapies can counteract the molecular defects present in melanoma, among which the most effective are BRAF inhibitors, which are used to treat metastatic and unresectable BRAF-mutated melanoma (Pelosi et al., 2024). It is highly effective in about half of patients with BRAF-mutated melanoma. Immersive checkpoint inhibitors are the most effective drugs for metastatic melanoma (Byrne and Fisher, 2017). Currently, the anti-CTLA-4 antibody (ipilimumab) and two anti-programmed cell death (PD-1) (nivolumab and pembrolizumab) have been used in clinical (Rotte et al., 2015). Although the immune checkpoint inhibitors are therapeutically effective, inevitable complications inhibit the mechanisms promoting self-cellular tolerance (Sharma and Allison, 2015). With the emergence of targeted therapy and immunotherapy over the past decade, significant progress has been made in the treatment of melanoma. However, ongoing challenges—such as limited therapeutic efficacy and disease recurrence—continue to drive the development of novel approaches and combination strategies.

Natural products and their derivatives are characterized by diverse structures, biological activities, low toxicity, and comprehensive sources. Their role in developing new anti-cancer drugs and lead drug compounds is becoming increasingly important (Agarwal et al., 2020; Kang et al., 2025; Wang et al., 2025). In addition, over the past 2 decades, many dietary and natural compounds with physiological activity—including phenols, flavonoids, alkaloids, carotenoids, gingerols, and organosulfur compounds—have been shown to inhibit both early and late stages of cancer. As a result, increasing attention has been directed toward the use of novel natural compounds in melanoma treatment, which has become an active area of research globally. For example, ginsenoside Rg3 and topotecan (a derivative of camptothecin) have already been approved for clinical use. In contrast, compounds such as luteic acid and silybin are still undergoing clinical evaluation. Their anticancer effects involve autophagy, apoptosis, and the regulation of multiple signaling pathways (Wen et al., 2021). This review focuses on natural products in the treatment of malignant melanoma and highlights their underlying molecular mechanisms. The identification of novel antitumor agents from natural products represents a promising strategy to improve long-term survival in melanoma patients and serves as a valuable source for anticancer drug discovery. Natural antitumor agents act through diverse mechanisms to inhibit cancer progression, including suppression of malignant cell proliferation, invasiveness, and neoangiogenesis, while typically exhibiting lower toxicity than conventional chemotherapeutic agents.

2 Natural products targeting melanoma-related signal pathways

By screening and summarizing the natural products that can induce melanoma cell apoptosis, we found that many in vitro and in vivo studies confirmed that natural products could induce melanoma cell apoptosis through various mechanisms. We classify these mechanisms as follows: 1) Wnt/β-Catenin signal pathway, 2) mediated endoplasmic reticulum (ER) stress signal pathway, 3) MAPK signal pathway, 4) phosphoinositol-3-kinase/protein kinase B/mammalian target protein of rapamycin (pl3k/akt/mTOR) signal pathway, and 5) others. As shown in Figure 2.

Figure 2
Illustration depicting the effects of natural products, derived from various plants, on cancer cells. It shows cancer cells targeted by compounds that activate pathways: P-PERK, JNK, LC3, and β-catenin, leading to apoptosis. Each pathway involves specific proteins and signal pathways, including ER stress, MAPK, PI3K/Akt/mTOR, and Wnt/β-catenin.

Figure 2. Natural products can induce apoptosis of melanoma cells through various mechanisms.

3 Wnt/β-catenin signal pathway

Previous studies have shown that Wnt/β-catenin signaling plays a critical role in embryonic development, cell differentiation and proliferation, and the ability of stem cells to self-renew (Riccardo et al., 2014; Li and Hou, 2018). It has now been demonstrated that the Wnt/β-catenin pathway regulates neural crest melanocyte formation and development and plays a vital role in the pathogenesis of melanoma (Umar et al., 2022; Natarelli et al., 2023). Several specific inhibitors have been developed and have been used in early clinical trials. However, there are insufficient nanotherapeutic activities for further registration trials (Voronkov and Krauss, 2013). Therefore, the Wnt/β-catenin pathway should be a priority molecular target for new drug development to expand the efficacy of clinical immunotherapy, as shown in Figure 3.

Figure 3
Diagram showing natural compounds and their effects on melanoma pathways. Emodin and Aloe-emodin influence the Wnt/β-catenin pathway. TAP7f and Shikonin affect the MAPK pathway. Plumbagin, Plyphyllin I, and Scutellarin impact the PI3K/Akt pathway. Resveratrol, δ-Tocotrienol, and 11-dehydrosinulariolide affect ER stress. Pathways lead to outcomes like apoptosis, autophagy, and anti-metastasis, with arrows indicating upregulation or downregulation of specific proteins.

Figure 3. Compounds affect changes in melanoma markers through related mechanisms.

3.1 Emodin

Emodin is also known as 1,3,8-trihydroxy-6-methylanthraquinone, its chemical structural formula as shown in Figure 4, and contains the remaining 18 natural products. It is the active ingredient in the root and rhizome of Rhubarb. Studies have shown that emodin has a wide range of pharmacological effects, including antibacterial, immunomodulation, anti-inflammatory, anti-tumor and enhanced cancer chemotherapy (Semwal et al., 2021; Zhang et al., 2022; Wang et al., 2023). It has been suggested that rhodopsin may impede the migration and invasion of melanoma B16F10 and A375 cells by inhibiting the Wnt/β-Catenin signaling pathway (Liu et al., 2021). Furthermore, rhodopsin also inhibits the growth of cancer cells by downregulating CD155 in melanoma B16F10 cells (Fang et al., 2019). Therefore, rhodopsin may be a potential drug for treating highly metastatic melanoma.

Figure 4
Chemical structures of various compounds labeled A to S. Some examples include Emodin (A), Aloe-emodin (B), TAP7f (C),Coenzyme Q0 (D), Resveratrol (E), δ-TT (F), Cudraflavone C (G), Shikonin (H), Plumbagin (I), Melittin (J), Fisetin (K), Polyphyllin I (L), Scutellarin (M), Sinomenine (N), Tanshinone IIa (O), Ursolic acid (P), Capsaicin (Q), bufalin (R) and Silymarin (S).

Figure 4. Chemical structures of various compounds labeled (A–S). Some examples include Emodin (A), Aloe-emodin (B), TAP7f (C), Coenzyme Q0 (D), Resveratrol (E), δ-TT (F), Cudraflavone C (G), Shikonin (H), Plumbagin (I), Melittin (J), Fisetin (K), Polyphyllin I (L), Scutellarin (M), Sinomenine (N), Tanshinone IIa (O), Ursolic acid (P), Capsaicin (Q), bufalin (R) and Silymarin (S).

3.2 Aloe-emodin

Aloe-emodin is an anthraquinone extract from the vine with the chemical formula C15H10O5 (Bhat and Kudva, 2011). Aloe-emodin has numerous pharmacological effects, including anti-inflammatory, immunomodulatory, and wound healing (Dong et al., 2020; Luo et al., 2024). New evidence showed that aloe-emodin exhibited anti-cancer ability in various cancers by inhibiting cell proliferation, migration, and invasion (Sanders et al., 2017). It was experimentally demonstrated that the proliferation, migration, and invasion ability of A375 and SK-MEL-28 melanoma cells treated with aloe-emodin were significantly inhibited, and the growth of A375 and SK-MEL-28 melanoma cells was affected by the inactivation of the Wnt/β-catenin signaling pathway. Furthermore, aloe-emodin also significantly inhibited the growth of A375 and SK-MEL-28 cells in the mouse model of transplantation tumors (Du et al., 2021). Therefore, aloe-emodin could be a potential treatment for melanoma.

3.3 TAP7f

TAP7f have anti-proliferative activity against different tumor cell lines (Bellizzi et al., 2022). Its anti-proliferation activity against tumor cells is 30 times higher than that against normal cells. Research has demonstrated that TAP7f exerts antitumor effects by inducing cell cycle arrest and activating death receptors and mitochondria-dependent apoptotic pathways in melanoma B16-F0 cells. Moreover, the melanoma tumor growth in the mouse model treated with TAP7f was reduced by 70% (Blank et al., 2018). New research has recently revealed that TAP7f may inhibit melanoma cell proliferation, migration, and invasion by blocking the Wnt/β-catenin pathway, reducing β-catenin nuclear translocation, and decreasing β-catenin and specific downstream targets (Barrionuevo et al., 2020).

3.4 Coenzyme Q0 (CoQ0)

CoQ0 also known as 2,3 dimethoxy-5-methyl-1,4 benzoquinone, is a common ubiquinone compound with redox activity widely found in biological mitochondria. CoQ0 has been demonstrated to have proliferative inhibitory effects on various cancer cell lines (Chung et al., 2014; Wang et al., 2017). In vitro experiments have shown that CoQ0 inhibits the proliferation of melanoma B16 cells by suppressing β-catenin-induced transcriptional activation and by nuclear translocation of the β-catenin proteasome. In B16F10 xenograft mice, the expression of β-catenin, Cyclin D1, Survivin, and MMP-9 was significantly reduced in the CoQ0-treated animal model (Hseu et al., 2016). These data demonstrate that CoQ0 inhibits cell growth and apoptosis and prevents metastasis by suppressing melanoma cells’ Wnt/β-linked protein signaling pathway.

4 ER pathway

ER is a eukaryotic cell organelle responsible for protein synthesis and calcium (Ca2+) signaling (Zheng et al., 2023). Previous studies have shown ER stress is strongly associated with cancer (Oakes, 2017). ER Stress initiates the unfolded protein response (UPR) to re-establish ER homeostasis as an adaptive pathway in cancer (Urra et al., 2013). Several studies have reported the involvement of ER stress in regulating the apoptotic mechanisms leading to melanoma cell death (Selimovic et al., 2013; Wroblewski et al., 2013). Accumulating evidence has shown that ER stress-induced autophagy may be a potential pro-survival mechanism contributing to melanoma development and resistance to BRAF inhibitors (Meng et al., 2015). Recently, there has been research on a novel natural compound called kuwanon H that can induce cytotoxic ER stress, inhibit cell viability, and induce apoptosis in melanoma cells. And it can induce ER stress-induced autophagosome formation through the ATF4-DDIT3-TRIB3-AKT-MTOR axis (Hu et al., 2023). Thus, modulation of ER stress may improve existing cancer therapies and identify new targets for therapeutic intervention in melanoma.

4.1 Resveratrol

Resveratrol is a dietary product found in grapes, vegetables, and berries. Resveratrol has been reported to affect each stage of carcinogenesis. Many studies demonstrate that resveratrol can be an ideal anti-cancer molecule because it has a cytotoxic effect on cancer cells (Jang et al., 2022). Studies have shown that resveratrol may induce ROS production and ER stress, thereby hindering the antioxidant effect of resveratrol and enhancing apoptosis of melanoma A375SM cells (Heo et al., 2018), revealing the potential use of resveratrol in the treatment of melanoma. A study has found that resveratrol downregulates the protein level of anti apoptotic protein Bcl-2 and activates Bax by promoting the degradation of Bcl-2 and the release of cytochrome c. In addition, they found that PKM2 plays a crucial role in triggering cell apoptosis. Summarizing that resveratrol in melanoma cells and downregulating the Erk/PKM2/BCl-2 axis seems to be a new method for preventing or treating melanoma (Zhao et al., 2018).

4.2 δ-TT

δ-TT is found in a wide variety of natural products. Due to its powerful neuroprotective, anti-inflammatory, antioxidant, and cholesterol-lowering potential (Ahsan et al., 2014; Peh et al., 2016). It works in various chronic diseases, and δ-TT shows antitumor activity as well (Wang et al., 2022). It has been demonstrated that δ-TT triggers cell death and activates ER stress-related pathways such as PERK/p-eIF2α/ATF4/CHOP and IRE1α to induce apoptosis in melanoma cells and that δ-TT significantly inhibits tumor growth rate in A375 melanoma animal model (Montagnani et al., 2016). These studies suggest that δ-TT may also be a potential drug for the treatment of melanoma.

4.3 Cudraflavone C (Cud C)

Cudraflavone C is a natural flavonoid compound with anti proliferative activity, which was initially found to inhibit melanin production (Zheng et al., 2012; Chan et al., 2021). Tumor specific apoptosis of rectal cancer cells can be induced by targeting the PI3K/AKT pathway (Soo et al., 2018). Research proves that cudraflavone C induces apoptosis in A375.S2 melanoma cells by increasing the production of mitochondrial ROS, activating p38, ERK and JNK, and increasing the expression of apoptotic proteins (Lee et al., 2017). Thus, cudraflavone C may be considered a potential therapeutic agent for treating malignant melanoma.

4.4 11-dehydrosinulariolide

11-Dehydrosinulariolide is a cembranolide analog, with a variety of biological activities (Chen et al., 2016). It has also been shown that 11-dehydrosinulariolide induces apoptosis in human melanoma cells by up-regulating PERK/eIF2α/ATF4/CHOP and ATF6/CHOP coupled with elevated ER stress chaperones GRP78, GRP94, calcium-linked protein, calreticulin, and PDI, which impede cystein-dependent mitochondrial function and ER stress pathway in human melanoma cells A2058 (Su et al., 2012). The present findings suggest that 11-dehydrosinulariolide is an effective compound against powerful melanoma cells in vitro, facilitating the drug development of anti-melanoma drugs.

5 MAPK

The MAPK cascade regulates cell proliferation, growth, and migration, an ordinarily active pathway but overactivated in almost all melanomas (Long et al., 2014). Mutations in NRAS or BRAF genes were observed in 80% of melanoma or melanocytic nevus cases, confirming the critical role of the MAPK pathway (Huang et al., 2023). New drugs targeting the MAPK pathway have produced excellent clinical responses in melanoma treatment, from the discovery of BRAF mutations in melanoma in 2002 to the FDA’s approval of the first BRAF inhibitor, vemurafenib, for melanoma treatment in 2011, therapies targeting the MAPK pathway have proven effective in less than a decade (Cheng et al., 2013). However, sequential treatment with BRAF/MEK inhibition and immunotherapy may increase the toxicity of sepsis-like syndrome and is associated with severe side effects (Moreira et al., 2021). Consequently, there is a need to develop new drugs that target the MAPK pathway for the treatment of melanoma.

5.1 Shikonin

Shikonin is a natural product isolated from comfrey, a member of the comfrey family, and has long been used in China to treat inflammation, burns, ulcers, infections, and cancer (Sun et al., 2022). Experiments have shown that increased phospho-ERK1/2, phospho-JNK, and phospho-p38 were observed in phycocyanin-treated cells, and total-ERK1/2, total-JNK, and total-p38 tended to decrease in phycocyanin-treated cells. Identifying apoptosis in A375SM melanoma cells induced by comfrey treatment through MAPK pathway. In a mouse animal model, the tumor volume was reduced by Shikonin administration. P38, an essential protein in the MAPK pathway, was significantly increased (Liu et al., 2019). The above results demonstrate that the apoptosis induction of paclitaxel in A375SM melanoma cells seems to be mediated by the expression of ERK and JNK proteins in the MAPK pathway, especially the expression of p38 (Lee et al., 2021).

5.2 L. barbarum extracts

L. barbarum is a traditional Asian food and medicine rich in zeaxanthin. Many studies have shown that L. barbarum extract has immunomodulatory effects and anti-tumor activity (Ceccarini et al., 2016; Wawruszak et al., 2016). L. barbarum extract containing zeaxanthin has been shown to inhibit the proliferation of human melanoma A375 cells, induce the expression of MAPK factors ERK1/2, JNK, and p38 in A375 skin cells, upregulate total NF-kB, thereby inducing apoptosis and reducing tumor cell growth. Therefore, L. barbarum extract may be used as an adjuvant for standard antitumor chemotherapy (Cenariu et al., 2021).

5.3 Plumbagin (PLB)

PLB is a naphthoquinone derivative derived mainly from plants that exhibit anti-cancer potential in different cancers (Yin et al., 2020; Zhang et al., 2020). Plumbagin was shown to downregulate MAPK-related genes, including Map3k3, MAPK14, Braf, c-Myc, and MAPK1, and induce enhanced gene expression of Igfbp5 and Pten, thus promoting further activation of the MAPK pathway, and thus acting as an anti-invasive and anti-metastatic agent in melanoma B16F10 cells (Alem et al., 2020). Meanwhile, PLB can trigger cell-specific cytotoxic effects and metabolic responses in melanoma A375 cells (Zhang et al., 2021). Combining Celecoxib and Plumbagin reduces the proliferation of melanoma cells and inhibits COX-2 and STAT3-mediated tumor vascular development, thereby inducing apoptosis in human fibroblast FF2441 cells (Gowda et al., 2017). Hence, it is necessary to investigate PLB as an antitumor agent and further develop its potential clinical applications.

5.4 Melittin

Bee Venom (BV) is a natural toxin produced by honey bees (Apis mellifera) and has been widely used as a traditional medicine for many diseases (Sadek et al., 2024). Melittin is the main bioactive component of BV. Moreover, in the current study, melittin has been shown to act against melanoma A375 cells by downregulating PI3K/AKT/mTOR and MAPK signaling pathways. Moreover, the growth, migration, and invasion of melanoma B16F10, A375SM, and SK-MEL-28 cells treated with different melittin concentrations were inhibited (Lim et al., 2019). Ultrasmall lipid nanoparticles driven by mixed melittin proteolytic peptides inhibit tumor cell growth in a mouse melanoma model, with no side effects observed (Huang et al., 2013). The current findings suggest that melittin could be used as a potential targeting agent in melanoma treatment.

5.5 Fisetin

Fisetin (3,3′,4′,7-tetrahydroxyflavone) is a dietary flavonoid found in various fruits and vegetables, including strawberries, grapes, apples, onions, and cucumbers (Khan et al., 2018). In many biological activities, including neuroprotective, anti-arthritic, and anti-allergic activities (Ahmad et al., 2017; Zheng et al., 2017). Fisetin was shown to inhibit phosphorylation of MEK1/2 and ERK1/2 in the MAPK signaling pathway activation pathway and inhibit NFκB in signaling to reduce melanoma A375 cell invasion and metastasis. Fisetin reduced the invasion of melanoma cells into the dermis in 3D skin consisting of A375 cells mixed with average human keratin-forming cells embedded in a collagen-constricted fibroblast matrix (Pal et al., 2014). In 3-D melanoma constructs, fisetin inhibits the growth of human melanoma A375 cells by directly binding to p70S6K and mTOR (Syed et al., 2014). The above experimental results demonstrate that fexofenadine can be developed as a potential anti-melanoma drug.

6 PI3K/AKT/mTOR

PI3K/Akt/mTOR pathway regulates cell proliferation, growth, size, metabolism, and motility (Alzahrani, 2019). Since multiple genes of the PI3K/Akt/mTOR signaling pathway are frequently altered in human cancers, dysregulation through mutation or amplification of genes involved in the PI3K pathway, loss of the tumor suppressor PTEN or over-activation of RTK leads to tumor progression and metastasis. The genes comprising this pathway are an essential molecular therapeutic target for human cancers (Murugan et al., 2013; Arafeh and Samuels, 2019).

6.1 Polyphyllin I (PPI)

PPI is a bioactive component derived from Paris polyphylla with a wide range of biological and pharmacological activities (Yang et al., 2024; Zhang et al., 2024). It was demonstrated that the expression of p-PI3K, p-Akt, and p-mTOR was significantly reduced in A375 cells after PPI treatment, and inhibited melanoma cell proliferation and enhanced melanoma cell apoptosis by suppressing PI3K/Akt/mTOR signaling pathway, which blocked melanoma cells at G0/G1 stage, thus reducing melanoma progression. In addition, the weight and size of melanoma mice treated with PPI was significantly reduced, and apoptosis of melanoma cells was significantly enhanced (Long and Pi, 2020). Therefore, PPI may be a promising, targeted drug for melanoma treatment.

6.2 Scutellarin

Scutellarin (4′,5,6-hydroxyl-flavone-7-glucuronide) is a flavonoid derived from Calendula officinalis with a variety of functions including antioxidant, anti-inflammatory, cardioprotective, and vasodilator (Wang and Ma, 2018). Scutellarin promotes A375 cell apoptosis by upregulating bax and cleaved caspase-3 levels, downregulating bcl-2 levels, Inhibit the growth of A375 cells in G0/G1 phase, Enhance cellular autophagy by regulating the levels of Beclin 1, LC3II, and p62. And block the melanoma cell cycle by inhibiting the PI3K/Akt/mTOR signaling pathwa (Li et al., 2019). It has also been demonstrated that lanosterol inhibits the proliferation of human melanoma cells RPMI7951 melanoma cells by targeting TOPK. In addition, in vivo experiments demonstrated that SCU inhibited the growth of RPMI7951 cell xenografts and reduced the phosphorylation levels of ERK 1/2 and histone H3 in vivo (Mu et al., 2021). It suggests that scutellarin may be a potential compound for treating malignant melanoma.

6.3 Bornyl cis -4-hydroxycinnamate

Bornyl cis-4-hydroxycinnamate, an active compound isolated from Piper betle stem, could inhibit cell viability, migration, and invasion of A2058 and A375 melanoma cells in a dose-dependent manner. The expression of FAK/PI3K/Akt/mTOR signaling-related proteins, including Akt, p-Akt, PI3K, p-PI3K, mTOR, p-mTOR, and FAK, was reduced in cis-4-hydroxycinnamate treated melanoma cells, which inhibited the metastasis of human melanoma cells through the FAK/PI3K/Akt/mTOR signaling pathway (Yang et al., 2018a). Additionally, cis-4- hydroxycinnamate has been shown to mediate apoptosis in melanoma cells by activating the cystathionine cascade, inducing mitochondrial dysfunction, and causing endoplasmic reticulum stress-related stress mechanisms (Yang et al., 2018b). The above results demonstrate that bornyl cis-4-hydroxycinnamate can potentially be a chemotherapeutic agent for human melanoma development.

6.4 Sinomenine

Sinomenine (7,8-didehydro-4-hydroxy-3,7-dimethoxy-17-methylmorphinane-6-one; SIN) is the active compound of the Chinese herb Cyanidin. SIN has an antitumor effect (Gao et al., 2019), including lung cancer and breast cancer (Song et al., 2015; Jiang et al., 2016). Recent studies have shown that SIN inhibits proliferation and promotes apoptosis in melanoma B16F10 cells via PI3K/Akt/mTOR-dependent autophagic pathway. In a melanoma xenograft mouse model, tumor volume and weight were significantly reduced after SIN treatment, and the expression levels of Ki67 and PCNA were significantly reduced (Sun et al., 2018). It suggests that SIN can reduce the tumor growth of melanoma in vivo. However, SIN also has some disadvantages, for example, poor gastrointestinal response, short biological half-life, and unstable physicochemical properties (Zhang et al., 2018). Therefore, the role of SIN in the treatment of melanoma deserves further investigation.

6.5 Tanshinone IIa (TanIIA)

TanIIA, a compound isolated from Salvia miltiorrhiza, has various biological activities, including apoptosis and autophagy, anti-inflammation, oxidation, anti-thrombosis, and anti-proliferation of vascular smooth muscle cells (Luo et al., 2015; Naz et al., 2020). It was shown that the phosphorylation levels of PI3K, P-AKT, P-mTOR, and P-p7036k1 were reduced in melanoma A375, MV3, and M14 cells treated with TanIIA, which inhibited the proliferation and invasion, and migration ability of melanoma cells, and promoted the autophagosome production of A375, In addition, TanIIA inhibits the development of A375 melanoma-induced tumor weight and volume in mice (Li et al., 2017). Research has found that TanIIA photosensitization has significant toxicity to choroidal melanoma cells and can effectively induce cell apoptosis and necrosis. Increase intracellular ROS levels, decrease mitochondrial membrane potential, and cause cell arrest in G2/M phase (Juan et al., 2021). In a recent report, the first study to link Tan IIA induced ferroptosis with the STAT1/PTGS2 axis in melanoma was found. Tan IIA regulates the key marker of ferroptosis, PTGS2, and knocking down PTGS2 weakens Tan IIA induced ferroptosis in melanoma cells. In addition, they found that Tan IIA stimulated the downregulation of signal transduction and transcription factor STAT1, leading to the downregulation of PTGS2 and inhibiting ferroptosis in melanoma (Chen et al., 2025).

7 Other mitochondrial mechanisms of resistance to melanoma

7.1 Ursolic acid (UA)

UA3-(β-hydroxy-urs-12-en-28-oic acid) is a pentacyclic triterpenoid compound commonly found in fruits, foods, and medicinal plants (Castro et al., 2015; Huang et al., 2015; Yin et al., 2018). Recent experiments have shown that ursolic acid activates the proteolytic processing of caspase-3 in isolated human melanoma cells and induces apoptotic cell death (Mahmoudi et al., 2015; Alvarado et al., 2018). Nanoemulsion of ursolic acid isolated from egg flowers enhances melanoma cells’ antioxidant and cytotoxic activity. When UA and chloroquine were synergistically applied to B16F10 mouse melanoma and A375 human melanoma cells, cell viability was strongly reduced (Junco et al., 2015). The above results suggest that ursolic acid could be a new potential tool for further research as an anti-cancer agent.

7.2 Capsaicin

Capsaicin is a component of chili peppers, the active ingredient in chili peppers. Capsicum is widely used as a pungent spice in food. Previous studies have demonstrated that capsaicin has anti-inflammatory, analgesic, anesthetic, and detoxifying effects (Bernstein et al., 2011; Patel et al., 2014; Fattori et al., 2016). Capsaicin was shown to affect cancer cell viability negatively and induce apoptosis in human melanoma A375 and C8161 cells by activating cleaved caspase-3 and PARP (Chu et al., 2019). There is also evidence that capsaicin inhibits the growth of SK-MEL-28 melanoma cells and increases apoptosis by inhibiting plasma membrane NADH oxidase activity (Morré et al., 1996). Capsaicin also inhibits the migration of B16-F10 melanoma cells by suppressing the PI3K/AKT/Rac-1 pathway (Shin et al., 2008). Therefore, capsaicin administration can be an effective method for the treatment of malignant melanoma.

7.3 Toad venom

Toad venom is a traditional natural medicine that has been used for centuries in China, and a growing body of research suggests that toad venom is a source of lead compounds for the development of potential cancer therapeutics (Li et al., 2021). Na+/K+ATPase is overexpressed in a variety of cancer types, including metastatic melanoma. Recent studies have shown that toad venom’s active ingredient, toadienolactone, acts on the Na+/K+-ATPase pump to exert anti-proliferative effects on melanoma cells (Soumoy et al., 2020). Bufalin, another active ingredient, can act through extrinsic and mitochondrial-mediated signaling pathways to trigger apoptosis (Hsiao et al., 2012). Bufalin can also potentially stimulate tyrosinase activity to promote melanin synthesis (Zhang et al., 1992). It may lead to the production of toxic melanin precursors, which inhibit melanoma growth.

7.4 Silymarin

Silymarin is the main bioactive component of silybum marianum and has long been used for the prevention of allergies and liver damag (Gillessen and Schmidt, 2020). Several studies have demonstrated the chemopreventive or chemotherapeutic effects of silymarin on various cancers (Dagne et al., 2011; Ramasamy et al., 2011). It has been shown that silymarin induces cell cycle arrest and inhibits the growth of human melanoma SK-MEL-5 and SK-MEL-28 cells in the G1 phase by blocking MEK1/2-RSK2 signaling, leading to a decrease in the activation of various transcriptional regulators of proliferation genes in melanoma, such as nuclear factor-kappaB, activator protein-1, and signal transduction and transcriptional activator 3. Silymarin also attenuates the growth of melanoma xenografts in nude mice (Lee et al., 2013). Furthermore, the combination of cold atmospheric plasma and silymarin nanoemulsion inhibited the HGF/c-MET signaling pathway to promote apoptosis in G-361 human melanoma cells and reduce tumor growth in a tumor xenograft nude mouse model (Adhikari et al., 2019), as shown in Table 1.

Table 1
www.frontiersin.org

Table 1. Natural products have been proven to have an antagonistic effect on melanoma through in vitro and in vivo experiments.

8 Discussion

Currently, the etiology of melanoma is generally believed to involve: 1) exposure to sunlight, 2) racial and genetic factors, 3) malignant transformation of benign pigmented nevi, 4) trauma and chronic irritants, which are also considered to be associated with melanoma development. Dacarbazine (1975), high-dose interleukin-2 (1992), and high-dose interferon α-2b (1995) were approved by the U.S. Food and Drug Administration (FDA) for the treatment of advanced melanoma. Although these advancements over 2 decades have offered hope for treating melanoma, the overall therapeutic outcomes remain unsatisfactory (Atkins et al., 1997; Kirkwood et al., 2004; Yang and Chapman, 2009). Since the early 2000s, the emergence of targeted therapies has highlighted the critical role of oncogene mutations—particularly in BRAF and NRAS—in activating cellular survival and proliferation pathways, including the MAPK and PI3K/AKT signaling cascades. The inhibition of mutated BRAF proteins by selective kinase inhibitors represents the most significant breakthrough in targeted melanoma therapy to date (Lim et al., 2017). Among the most established therapies targeting the BRAFV600E mutation site are vemurafenib (approved in 2011) and dabrafenib (approved in 2013), which serve as first-line treatments for advanced BRAF-mutant melanoma (Garbe and Eigentler, 2018; Long et al., 2024). Extensive studies have demonstrated that the immunosuppressive molecular marker PD-L1 plays a pivotal role in the mechanism of tumor immune evasion. Monoclonal antibodies such as nivolumab and pembrolizumab, which target PD-1, and atezolizumab, which targets PD-L1, have been shown to effectively block the PD-1/PD-L1 interaction (Hamanishi et al., 2016). The immune checkpoints they mediate inhibit the activity of reactivated immune cells, thereby enabling tumor cells to escape immune surveillance. Moreover, nivolumab and pembrolizumab have been approved as first-line therapies for surgically unresectable or metastatic malignant melanoma. Targeted therapy and immunotherapy have significantly altered the treatment landscape for both resectable and unresectable melanoma, representing a major advance in melanoma management.

Although progress has been made in melanoma treatment, immunosuppressive agents currently employed in clinical practice are characterized by limited specificity, and their use is constrained by toxic side effects and elevated costs. To date, the clinical efficacy of monotherapy remains insufficiently defined, and the development of individualized treatment regimens and combination approaches based on a patient’s immune status remains a priority for future investigation. Consequently, the exploration of novel natural products and their extracts as integral components of combination therapy has been widely investigated in oncology. These compounds are characterized by low toxicity, wide availability, and modest cost (Sflakidou et al., 2022). A comprehensive understanding of the relationship between chemical structure and biological activity, along with the optimization of structure–activity relationships, can help clarify the diverse structures of plant-derived compounds and facilitate the investigation of correlations between nanostructures and biological activity. This, in turn, may enhance the immunological potential of compound extracts and support the rational design of more effective drug candidates. In addition, mouse models represent the most widely utilized animal models in preclinical research. Melanoma mouse models are typically categorized into three types: inducible models (e.g., UV-induced models and chemically induced models using DMBA or TPA), transplantable models (including syngeneic and xenograft models), and transgenic models (such as Lum−/− knockout mice, TyrN-RasQ61K transgenic mice, and BrafV600E transgenic mice) (Kuzu et al., 2015; Day et al., 2017), these models have become essential tools for investigating the pathogenesis, metastasis, and therapeutic evaluation of melanoma. The models discussed in this review are broadly classified into two categories: in vivo and in vitro experimental systems. In vitro studies primarily involve melanoma cell lines such as B16F10, A375, SK-MEL-28, and A2058, whereas in vivo studies utilize animal models including BALB/c nude mice. In addition, other animal models such as C57BL/6, C3H/HeN, and CB17-SCID mice have also been utilized in various melanoma-related studies (Liu et al., 2018; Saleh, 2018). This research aims to identify susceptibility and risk factors for melanoma development, assess the efficacy of immunotherapies and tumor–immune cell interactions, elucidate the signaling pathways involved in tumor progression, clarify gene functions, and define potential therapeutic targets.

Natural product–derived compounds have been applied in clinical oncology; however, their clinical utility is often limited by inherent physicochemical and pharmacokinetic properties. For instance, poor water solubility, limited biocompatibility, low oral bioavailability, chemical instability, and suboptimal pharmacokinetics significantly hinder their clinical translation (Bitterman and Chung, 2015; Khoury et al., 2015; Cai et al., 2016; Kharat et al., 2017; Wen et al., 2017; Velavan et al., 2018; Wei et al., 2019). Therefore, there is a growing need for further studies to clarify the bioavailability and pharmacokinetic behavior of these bioactive compounds. Natural products in melanoma therapy are currently in the early stages of investigation, and their anticancer effects appear to vary in a dose-dependent manner. Although physiological effects have been observed in animal models, there are currently few clinical studies assessing the efficacy of these compounds in humans.

However, these active ingredients exhibit low bioavailability, poor stability, and limited water solubility, which restrict their clinical application and highlight the need for new strategies to enhance their absorption and therapeutic efficacy. In particular, the application and development of nanotechnology has emerged as a promising approach. In conclusion, natural product–derived compounds represent promising anticancer agents that warrant further investigation to better elucidate the mechanisms underlying their antitumor activity. Moreover, combining these compounds with conventional antitumor therapies may enhance their therapeutic potential through synergistic effects and reduced toxicity. Such combinations may offer safer and more effective strategies for clinical intervention. Natural products are expected to contribute to future breakthroughs in tumor immunotherapy.

Overall, the treatment of melanoma continues to face numerous challenges, including: 1) significant biological differences between animal and cell-based models and human physiology, such as variations in genome organization, gene regulation, cell types, and organ structures, which limit the ability to fully replicate human pathological processes; 2) uncertainty regarding how newly identified compounds can achieve optimal bioactivity in vivo to exert therapeutic effects; 3) incomplete understanding of the mechanisms of action of novel compounds; 4) the potential for immune-related toxicities associated with the widespread use of bioactive agents; 5) unclear interactions between compound-based therapies and existing targeted or immunosuppressive treatments; and 6) limitations related to sample size and study duration in experimental research. Therefore, in addition to fully understanding the advantages, disadvantages, and applicability of these compounds and animal models, it is also necessary to develop individualized diagnosis and treatment plans for different types of melanoma to provide valuable information for optimizing melanoma models and drug evaluation. Furthermore, the application of drug-specific biomarkers may enable more precise and personalized treatment approaches. The research and development of new drugs remain a long-term task that requires joint efforts from researchers.

Author contributions

HG: Writing – original draft, Formal Analysis, Investigation. JH: Conceptualization, Data curation, Writing – original draft, Writing – review and editing. DZ: Data curation, Investigation, Writing – review and editing. SiL: Data curation, Investigation, Writing – review and editing. ShL: Supervision, Visualization, Writing – review and editing. YQ: Supervision, Visualization, Writing – review and editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

PI3K, phosphoinositol-3-kinase; PDL-1, programmed death ligand 1; CDKN, cyclin dependent kinase inhibitors; ERK, extracellular signal-regulated kinase; Akt, protein kinase B; mTOR, mammalian target protein of rapamycin; TAP7f, triazolylpeptidyl penicillin derivative; JNK, c-Jun N-terminal kinase; δ-TT, vitamin E delta-tocotrienol; MAPK, mitogen-activated protein kinase; TanIIA, tanshinone IIa; ER, endoplasmic reticulum; UA, ursolic acid; CoQ0, coenzyme Q0; UPR, unfolded protein response; Cud C, cudraflavone C; PLB, plumbagin; BV, bee venom; PPI, polyphyllin I. programmed cell death ligand; PD-L1; programmed cell death, PD-1.

References

Adhikari, M., Kaushik, N., Ghimire, B., Adhikari, B., Baboota, S., Al-Khedhairy, A. A., et al. (2019). Cold atmospheric plasma and silymarin nanoemulsion synergistically inhibits human melanoma tumorigenesis via targeting HGF/c-MET downstream pathway. Cell Commun. Signal 17 (1), 52. doi:10.1186/s12964-019-0360-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Agarwal, G., Carcache, P. J. B., Addo, E. M., and Kinghorn, A. D. (2020). Current status and contemporary approaches to the discovery of antitumor agents from higher plants. Biotechnol. Adv. 38, 107337. doi:10.1016/j.biotechadv.2019.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahmad, A., Ali, T., Park, H. Y., Badshah, H., Rehman, S. U., and Kim, M. O. (2017). Neuroprotective effect of fisetin against amyloid-beta-induced cognitive/Synaptic dysfunction, neuroinflammation, and neurodegeneration in adult mice. Mol. Neurobiol. 54 (3), 2269–2285. doi:10.1007/s12035-016-9795-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahsan, H., Ahad, A., Iqbal, J., and Siddiqui, W. A. (2014). Pharmacological potential of tocotrienols: a review. Nutr. Metab. (Lond). 11 (1), 52. doi:10.1186/1743-7075-11-52

PubMed Abstract | CrossRef Full Text | Google Scholar

Alem, F. Z., Bejaoui, M., Villareal, M. O., Rhourri-Frih, B., and Isoda, H. (2020). Elucidation of the effect of plumbagin on the metastatic potential of B16F10 murine melanoma cells via MAPK signalling pathway. Exp. Dermatol 29 (4), 427–435. doi:10.1111/exd.14079

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarado, H. L., Calpena, A. C., Garduño-Ramírez, M. L., Ortiz, R., Melguizo, C., Prados, J. C., et al. (2018). Nanoemulsion strategy for ursolic and oleanic acids isolates from Plumeria Obtusa improves antioxidant and cytotoxic activity in Melanoma cells. Anticancer Agents Med. Chem. 18 (6), 847–853. doi:10.2174/1871520618666180111151846

PubMed Abstract | CrossRef Full Text | Google Scholar

Alzahrani, A. S. (2019). PI3K/Akt/mTOR inhibitors in cancer: at the bench and bedside. Semin. Cancer Biol. 59, 125–132. doi:10.1016/j.semcancer.2019.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Arafeh, R., and Samuels, Y. (2019). PIK3CA in cancer: the past 30 years. Semin. Cancer Biol. 59, 36–49. doi:10.1016/j.semcancer.2019.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Atkins, M. B., Lotze, M. T., Dutcher, J. P., Fisher, R. I., Weiss, G., Margolin, K., et al. (1997). High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J. Clin. Oncol. 17 (7), 2105–2116. doi:10.1200/JCO.1999.17.7.2105

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrionuevo, E., Cayrol, F., Cremaschi, G. A., Cornier, P. G., Boggián, D. B., Delpiccolo, C. M. L., et al. (2020). A penicillin derivative exerts an anti-metastatic activity in Melanoma cells through the downregulation of Integrin αvβ3 and Wnt/β-Catenin pathway. Front. Pharmacol. 11, 127. doi:10.3389/fphar.2020.00127

PubMed Abstract | CrossRef Full Text | Google Scholar

Bellizzi, Y., Anselmi Relats, J. M., Cornier, P. G., Delpiccolo, C. M. L., Mata, E. G., Cayrol, F., et al. (2022). Contribution of endoplasmic reticulum stress, MAPK and PI3K/Akt pathways to the apoptotic death induced by a penicillin derivative in melanoma cells. Apoptosis 27 (1-2), 34–48. doi:10.1007/s10495-021-01697-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Bernstein, J. A., Davis, B. P., Picard, J. K., Cooper, J. P., Zheng, S., and Levin, L. S. (2011). A randomized, double-blind, parallel trial comparing capsaicin nasal spray with placebo in subjects with a significant component of nonallergic rhinitis. Ann. Allergy Asthma Immunol. 107 (2), 171–178. doi:10.1016/j.anai.2011.05.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhat, G., Kudva, P., and Dodwad, V. (2011). Aloe vera: nature's soothing healer to periodontal disease. J. Indian Soc. Periodontol. 15 (3), 205–209. doi:10.4103/0972-124X.85661

PubMed Abstract | CrossRef Full Text | Google Scholar

Bitterman, J. L., and Chung, J. H. (2015). Metabolic effects of resveratrol: addressing the controversies. Cell Mol. Life Sci. 72 (8), 1473–1488. doi:10.1007/s00018-014-1808-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Blank, V., Bellizzi, Y., Zotta, E., Cornier, P. G., Delpiccolo, C. M. L., Boggián, D. B., et al. (2018). A novel penicillin derivative induces antitumor effect in melanoma cells. Anticancer Drugs 29 (5), 416–428. doi:10.1097/CAD.0000000000000611

PubMed Abstract | CrossRef Full Text | Google Scholar

Byrne, E. H., and Fisher, D. E. (2017). Immune and molecular correlates in melanoma treated with immune checkpoint blockade. Cancer 123 (S11), 2143–2153. doi:10.1002/cncr.30444

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, Y., Zhang, W., Chen, Z., Shi, Z., He, C., and Chen, M. (2016). Recent insights into the biological activities and drug delivery systems of tanshinones. Int. J. Nanomedicine 11, 121–130. doi:10.2147/IJN.S84035

PubMed Abstract | CrossRef Full Text | Google Scholar

Castellani, G., Buccarelli, M., Arasi, M. B., Rossi, S., Pisanu, M. E., Bellenghi, M., et al. (2023). BRAF mutations in Melanoma: biological aspects, therapeutic implications, and circulating biomarkers. Cancers (Basel) 15 (16), 4026. doi:10.3390/cancers15164026

PubMed Abstract | CrossRef Full Text | Google Scholar

Castro, A. J., Frederico, M. J., Cazarolli, L. H., Mendes, C. P., Bretanha, L. C., Schmidt, É. C., et al. (2015). The mechanism of action of ursolic acid as insulin secretagogue and insulinomimetic is mediated by cross-talk between calcium and kinases to regulate glucose balance. Biochim. Biophys. Acta 1850 (1), 51–61. doi:10.1016/j.bbagen.2014.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Ceccarini, M. R., Vannini, S., Cataldi, S., Moretti, M., Villarini, M., Fioretti, B., et al. (2016). In vitro protective effects of Lycium barbarum berries cultivated in Umbria (Italy) on Human hepatocellular carcinoma cells. Biomed. Res. Int. 2016, 7529521. doi:10.1155/2016/7529521

PubMed Abstract | CrossRef Full Text | Google Scholar

Cenariu, D., Fischer-Fodor, E., Tigu, A. B., Bunea, A., Virág, P., Perde-Schrepler, M., et al. (2021). Zeaxanthin-Rich extract from Superfood Lycium barbarum selectively modulates the cellular adhesion and MAPK signaling in Melanoma versus normal skin cells in vitro. Molecules 26 (2), 333. doi:10.3390/molecules26020333

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, D., Meister, M. L., Madhani, C. R., Elfakhani, M., Yount, S. T., Ji, X., et al. (2021). Synergistic impact of Xanthorrhizol and d-δ-Tocotrienol on the proliferation of murine B16 melanoma cells and human DU145 prostate carcinoma cells. Nutr. Cancer 73 (9), 1746–1757. doi:10.1080/01635581.2020.1807573

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, C. H., Chen, N. F., Feng, C. W., Cheng, S. Y., Hung, H. C., Tsui, K. H., et al. (2016). A coral-derived compound improves functional recovery after spinal cord injury through its antiapoptotic and anti-inflammatory effects. Mar. Drugs 14 (9), 160. doi:10.3390/md14090160

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, S., Li, P., Shi, K., Tang, S., Zhang, W., Peng, C., et al. (2025). Tanshinone IIA promotes ferroptosis in cutaneous melanoma via STAT1-mediated upregulation of PTGS2 expression. Phytomedicine 141, 156702. doi:10.1016/j.phymed.2025.156702

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, Y., Zhang, G., and Li, G. (2013). Targeting MAPK pathway in melanoma therapy. Cancer Metastasis Rev. 32 (3-4), 567–584. doi:10.1007/s10555-013-9433-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, L., Lopez-Beltran, A., Massari, F., MacLennan, G. T., and Montironi, R. (2018). Molecular testing for BRAF mutations to inform melanoma treatment decisions: a move toward precision medicine. Mod. Pathol. 31, 24–38. doi:10.1038/modpathol.2017.104

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, H., Li, M., and Wang, X. (2019). Capsaicin induces apoptosis and autophagy in human melanoma cells. Oncol. Lett. 17 (6), 4827–4834. doi:10.3892/ol.2019.10206

PubMed Abstract | CrossRef Full Text | Google Scholar

Chung, C. H., Yeh, S. C., Chen, C. J., and Lee, K. T. (2014). Coenzyme Q0 from Antrodia cinnamomea in submerged cultures induces reactive oxygen species-mediated apoptosis in A549 human lung cancer cells. Evid. Based Complement. Altern. Med. 2014, 246748. doi:10.1155/2014/246748

PubMed Abstract | CrossRef Full Text | Google Scholar

Connor, L., Carr, Q. L., Sweazy, A., McMasters, K., and Hao, H. (2025). Clinical approaches for the management of skin cancer: a review of Current progress in diagnosis, treatment, and prognosis for patients with Melanoma. Cancers (Basel) 17 (4), 707. doi:10.3390/cancers17040707

PubMed Abstract | CrossRef Full Text | Google Scholar

Czarnecka, A. M., Bartnik, E., Fiedorowicz, M., and Rutkowski, P. (2020). Targeted therapy in Melanoma and mechanisms of resistance. Int. J. Mol. Sci. 21, 4576. doi:10.3390/ijms21134576

PubMed Abstract | CrossRef Full Text | Google Scholar

Dagne, A., Melkamu, T., Schutten, M. M., Qian, X., Upadhyaya, P., Luo, X., et al. (2011). Enhanced inhibition of lung adenocarcinoma by combinatorial treatment with indole-3-carbinol and silibinin in A/J mice. Carcinogenesis 32 (4), 561–567. doi:10.1093/carcin/bgr010

PubMed Abstract | CrossRef Full Text | Google Scholar

Davis, L. E., Shalin, S. C., and Tackett, A. J. (2019). Current state of melanoma diagnosis and treatment. Cancer Biol. Ther. 20 (11), 1366–1379. doi:10.1080/15384047.2019.1640032

PubMed Abstract | CrossRef Full Text | Google Scholar

Day, C. P., Marchalik, R., Merlino, G., and Michael, H. (2017). Mouse models of UV-induced melanoma: genetics, pathology, and clinical relevance. Lab. Invest 97 (6), 698–705. doi:10.1038/labinvest.2016.155

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, X., Zeng, Y., Liu, Y., You, L., Yin, X., Fu, J., et al. (2020). Aloe-emodin: a review of its pharmacology, toxicity, and pharmacokinetics. Phytother. Res. 34 (2), 270–281. doi:10.1002/ptr.6532

PubMed Abstract | CrossRef Full Text | Google Scholar

Du, M., Shen, P., Tan, R., Wu, D., and Tu, S. (2021). Aloe-emodin inhibits the proliferation, migration, and invasion of melanoma cells via inactivation of the Wnt/beta-catenin signaling pathway. Ann. Transl. Med. 9 (23), 1722. doi:10.21037/atm-21-5437

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, L., Zhao, F., Iwanowycz, S., Wang, J., Yin, S., Wang, Y., et al. (2019). Anticancer activity of emodin is associated with downregulation of CD155. Int. Immunopharmacol. 75, 105763. doi:10.1016/j.intimp.2019.105763

PubMed Abstract | CrossRef Full Text | Google Scholar

Fattori, V., Hohmann, M. S., Rossaneis, A. C., Pinho-Ribeiro, F. A., and Verri, W. A. (2016). Capsaicin: current understanding of its mechanisms and therapy of pain and other pre-clinical and clinical uses. Molecules 21 (7), 844. doi:10.3390/molecules21070844

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, L. N., Zhong, B., and Wang, Y. (2019). Mechanism underlying antitumor effects of sinomenine. Chin. J. Integr. Med. 25 (11), 873–878. doi:10.1007/s11655-019-3151-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Garbe, C., and Eigentler, T. K. (2018). Vemurafenib. Cancer Res. 211, 77–89. doi:10.1007/978-3-319-91442-8_6

PubMed Abstract | CrossRef Full Text | Google Scholar

Gillessen, A., and Schmidt, H. H. (2020). Silymarin as supportive treatment in liver diseases: a narrative review. Adv. Ther. 37 (4), 1279–1301. doi:10.1007/s12325-020-01251-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Gowda, R., Sharma, A., and Robertson, G. P. (2017). Synergistic inhibitory effects of Celecoxib and Plumbagin on melanoma tumor growth. Cancer Lett. 385, 243–250. doi:10.1016/j.canlet.2016.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamanishi, J., Mandai, M., Matsumura, N., Abiko, K., Baba, T., and Konishi, I. (2016). PD-1/PD-L1 blockade in cancer treatment: perspectives and issues. Int. J. Clin. Oncol. 21 (3), 462–473. doi:10.1007/s10147-016-0959-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Heo, J. R., Kim, S. M., Hwang, K. A., Kang, J. H., and Choi, K. C. (2018). Resveratrol induced reactive oxygen species and endoplasmic reticulum stress-mediated apoptosis, and cell cycle arrest in the A375SM malignant melanoma cell line. Int. J. Mol. Med. 42 (3), 1427–1435. doi:10.3892/ijmm.2018.3732

PubMed Abstract | CrossRef Full Text | Google Scholar

Hseu, Y. C., Thiyagarajan, V., Tsou, H. T., Lin, K. Y., Chen, H. J., Lin, C. M., et al. (2016). In vitro and in vivo anti-tumor activity of CoQ0 against melanoma cells: inhibition of metastasis and induction of cell-cycle arrest and apoptosis through modulation of Wnt/β-catenin signaling pathways. Oncotarget 7 (16), 22409–22426. doi:10.18632/oncotarget.7983

PubMed Abstract | CrossRef Full Text | Google Scholar

Hsiao, Y. P., Yu, C. S., Yu, C. C., Yang, J. S., Chiang, J. H., Lu, C. C., et al. (2012). Triggering apoptotic death of human malignant melanoma a375.s2 cells by bufalin: involvement of caspase cascade-dependent and independent mitochondrial signaling pathways. Evid. Based Complement. Altern. Med. 2012, 591241. doi:10.1155/2012/591241

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, X., Pan, G., Luo, J., Gao, X., Mu, Y., Wang, Z., et al. (2023). Kuwanon H inhibits melanoma growth through cytotoxic endoplasmic reticulum stress and impaired autophagy flux. J. Agric. Food Chem. 71 (37), 13768–13782. doi:10.1021/acs.jafc.3c02257

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, C., Jin, H., Qian, Y., Qi, S., Luo, H., Luo, Q., et al. (2013). Hybrid melittin cytolytic peptide-driven ultrasmall lipid nanoparticles block melanoma growth in vivo. ACS Nano 7 (7), 5791–5800. doi:10.1021/nn400683s

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, L., Luo, H., Li, Q., Wang, D., Zhang, J., Hao, X., et al. (2015). Pentacyclic triterpene derivatives possessing polyhydroxyl ring A inhibit gram-positive bacteria growth by regulating metabolism and virulence genes expression. Eur. J. Med. Chem. 95, 64–75. doi:10.1016/j.ejmech.2015.01.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, F., Cai, F., Dahabieh, M. S., Gunawardena, K., Talebi, A., Dehairs, J., et al. (2023). Peroxisome disruption alters lipid metabolism and potentiates antitumor response with MAPK-targeted therapy in melanoma. J. Clin. investigation 133 (20), e166644. doi:10.1172/JCI166644

PubMed Abstract | CrossRef Full Text | Google Scholar

Jang, J. Y., Im, E., and Kim, N. D. (2022). Mechanism of resveratrol-induced programmed cell death and new drug discovery against cancer: a review. Int. J. Mol. Sci. 23 (22), 13689. doi:10.3390/ijms232213689

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, S., Gao, Y., Hou, W., Liu, R., Qi, X., Xu, X., et al. (2016). Sinomenine inhibits A549 human lung cancer cell invasion by mediating the STAT3 signaling pathway. Oncol. Lett. 12 (2), 1380–1386. doi:10.3892/ol.2016.4768

PubMed Abstract | CrossRef Full Text | Google Scholar

Juan, L., Diandian, W., Jianfeng, W., Ning, L., Yuchen, F., Na, L., et al. (2021). Efficient anticancer effect on choroidal melanoma cells induced by tanshinone IIA photosensitization. Photochem Photobiol. 97 (4), 841–850. doi:10.1111/php.13399

PubMed Abstract | CrossRef Full Text | Google Scholar

Junco, J. J., Mancha-Ramirez, A., Malik, G., Wei, S. J., Kim, D. J., Liang, H., et al. (2015). Ursolic acid and resveratrol synergize with chloroquine to reduce melanoma cell viability. Melanoma Res. 25 (2), 103–112. doi:10.1097/CMR.0000000000000137

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, H. S., Lim, H. K., Jang, W. Y., and Cho, J. Y. (2025). Anti-Colorectal cancer activity of panax and its active components, ginsenosides: a review. Int. J. Mol. Sci. 26 (6), 2593. doi:10.3390/ijms26062593

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, H., Marya, A. S., Kamal, M. A., and Patel, S. (2018). Flavonoids as acetylcholinesterase inhibitors: current therapeutic standing and future prospects. Biomed. Pharmacother. 101, 860–870. doi:10.1016/j.biopha.2018.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Kharat, M., Du, Z., Zhang, G., and McClements, D. J. (2017). Physical and chemical stability of curcumin in aqueous solutions and emulsions: impact of pH, temperature, and molecular environment. J. Agric. Food Chem. 65 (8), 1525–1532. doi:10.1021/acs.jafc.6b04815

PubMed Abstract | CrossRef Full Text | Google Scholar

Khoury, T., Rmeileh, A. A., Yosha, L., Benson, A. A., Daher, S., and Mizrahi, M. (2015). Drug induced liver injury: review with a focus on genetic factors, tissue diagnosis, and treatment options. J. Clin. Transl. Hepatol. 3 (2), 99–108. doi:10.14218/JCTH.2015.00007

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, H. J., and Kim, Y. H. (2024). Molecular frontiers in melanoma: pathogenesis, diagnosis, and therapeutic advances. Int. J. Mol. Sci. 25 (5), 2984. doi:10.3390/ijms25052984

PubMed Abstract | CrossRef Full Text | Google Scholar

Kirkwood, J. M., Manola, J., Ibrahim, J., Sondak, V., Ernstoff, M. S., Rao, U., et al. (2004). A pooled analysis of eastern cooperative oncology group and intergroup trials of adjuvant high-dose interferon for melanoma. Clin. Cancer Res. 10 (5), 1670–1677. doi:10.1158/1078-0432.ccr-1103-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuzu, O. F., Nguyen, F. D., Noory, M. A., and Sharma, A. (2015). Current State of animal (Mouse) modeling in Melanoma research. Cancer Growth Metastasis 8 (Suppl. 1), 81–94. doi:10.4137/CGM.S21214

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, M. H., Huang, Z., Kim, D. J., Kim, S. H., Kim, M. O., Lee, S. Y., et al. (2013). Direct targeting of MEK1/2 and RSK2 by silybin induces cell-cycle arrest and inhibits melanoma cell growth. Cancer Prev. Res. (Phila) 6 (5), 455–465. doi:10.1158/1940-6207.CAPR-12-0425

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, C. W., Yen, F. L., Ko, H. H., Li, S. Y., Chiang, Y. C., Lee, M. H., et al. (2017). Cudraflavone C induces apoptosis of A375.S2 melanoma cells through mitochondrial ROS production and MAPK activation. Int. J. Mol. Sci. 18 (7), 1508. doi:10.3390/ijms18071508

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, J. H., Han, S. H., Kim, Y. M., Kim, S. H., Yoo, E. S., Woo, J. S., et al. (2021). Shikonin inhibits proliferation of melanoma cells by MAPK pathway-mediated induction of apoptosis. Biosci. Rep. 41 (1), BSR20203834. doi:10.1042/BSR20203834

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., and Hou, L. (2018). Regulation of melanocyte stem cell behavior by the niche microenvironment. Pigment. Cell Melanoma Res. 31 (5), 556–569. doi:10.1111/pcmr.12701

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Li, Z., Li, X., Liu, B., and Liu, Z. (2017). Mechanisms of Tanshinone II a inhibits malignant melanoma development through blocking autophagy signal transduction in A375 cell. BMC Cancer 17 (1), 357. doi:10.1186/s12885-017-3329-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C. Y., Wang, Q., Wang, X., Li, G., Shen, S., and Wei, X. (2019). Scutellarin inhibits the invasive potential of malignant melanoma cells through the suppression epithelial-mesenchymal transition and angiogenesis via the PI3K/Akt/mTOR signaling pathway. Eur. J. Pharmacol. 858, 172463. doi:10.1016/j.ejphar.2019.172463

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, F. J., Hu, J. H., Ren, X., Zhou, C. M., Liu, Q., and Zhang, Y. Q. (2021). Toad venom: a comprehensive review of chemical constituents, anticancer activities, and mechanisms. Arch. Pharm. Weinh. 354 (7), e2100060. doi:10.1002/ardp.202100060

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, S. Y., Menzies, A. M., and Rizos, H. (2017). Mechanisms and strategies to overcome resistance to molecularly targeted therapy for melanoma. Cancer 123 (S11), 2118–2129. doi:10.1002/cncr.30435

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, H. N., Baek, S. B., and Jung, H. J. (2019). Bee venom and its peptide component melittin suppress growth and migration of Melanoma cells via inhibition of PI3K/AKT/mTOR and MAPK pathways. Molecules 24 (5), 929. doi:10.3390/molecules24050929

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Q., Das, M., Liu, Y., and Huang, L. (2018). Targeted drug delivery to melanoma. Adv. Drug Deliv. Rev. 127, 208–221. doi:10.1016/j.addr.2017.09.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Kang, X., Niu, G., He, S., Zhang, T., Bai, Y., et al. (2019). Shikonin induces apoptosis and prosurvival autophagy in human melanoma A375 cells via ROS-mediated ER stress and p38 pathways. Artif. Cells Nanomed Biotechnol. 47 (1), 626–635. doi:10.1080/21691401.2019.1575229

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, C., Chen, L., Wang, W., Qin, D., Jia, C., Yuan, M., et al. (2021). Emodin suppresses the migration and invasion of Melanoma cells. Biol. Pharm. Bull. 44 (6), 771–779. doi:10.1248/bpb.b20-00807

PubMed Abstract | CrossRef Full Text | Google Scholar

Long, J., and Pi, X. (2020). Polyphyllin I promoted Melanoma cells autophagy and apoptosis via PI3K/Akt/mTOR signaling pathway. Biomed. Res. Int. 2020, 5149417. doi:10.1155/2020/5149417

PubMed Abstract | CrossRef Full Text | Google Scholar

Long, G. V., Stroyakovskiy, D., Gogas, H., Levchenko, E., de Braud, F., Larkin, J., et al. (2014). Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N. Engl. J. Med. 371 (20), 1877–1888. doi:10.1056/NEJMoa1406037

PubMed Abstract | CrossRef Full Text | Google Scholar

Long, G. V., Swetter, S. M., Menzies, A. M., Gershenwald, J. E., and Scolyer, R. A. (2023). Cutaneous melanoma. Lancet 402 (10400), 485–502. doi:10.1016/S0140-6736(23)00821-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Long, G. V., Hauschild, A., Santinami, M., Kirkwood, J. M., Atkinson, V., Mandala, M., et al. (2024). Final results for adjuvant dabrafenib plus trametinib in stage III melanoma. N. Engl. J. Med. 391 (18), 1709–1720. doi:10.1056/NEJMoa2404139

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, J., Song, W., Yang, G., Xu, H., and Chen, K. (2015). Compound Danshen (Salvia miltiorrhiza) dripping pill for coronary heart disease: an overview of systematic reviews. Am. J. Chin. Med. 43 (1), 25–43. doi:10.1142/S0192415X15500020

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, H., Ji, X., Zhang, M., Ren, Y., Tan, R., Jiang, H., et al. (2024). Aloe-emodin: progress in pharmacological activity, safety, and pharmaceutical formulation applications. Mini Rev. Med. Chem. 24 (19), 1784–1798. doi:10.2174/0113895575298364240409064833

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahmoudi, M., Rabe, S. Z., Balali-Mood, M., Karimi, G., Tabasi, N., and Riahi-Zanjani, B. (2015). Ursolic acid induced apoptotic cell death following activation of caspases in isolated human melanoma cells. Cell Biol. Int. 39 (2), 230–236. doi:10.1002/cbin.10376

PubMed Abstract | CrossRef Full Text | Google Scholar

Megahed, A. I., and Koon, H. B. (2014). What is the role of chemotherapy in the treatment of melanoma? Curr. Treat. Options Oncol. 15 (2), 321–335. doi:10.1007/s11864-014-0277-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng, X. X., Yao, M., Zhang, X. D., Xu, H. X., and Dong, Q. (2015). ER stress-induced autophagy in melanoma. Clin. Exp. Pharmacol. Physiol. 42 (8), 811–816. doi:10.1111/1440-1681.12436

PubMed Abstract | CrossRef Full Text | Google Scholar

Montagnani, M. M., Marzagalli, M., Moretti, R. M., Beretta, G., Casati, L., Comitato, R., et al. (2016). Vitamin E δ-tocotrienol triggers endoplasmic reticulum stress-mediated apoptosis in human melanoma cells. Sci. Rep. 6, 30502. doi:10.1038/srep30502

PubMed Abstract | CrossRef Full Text | Google Scholar

Moreira, A., Lebbé, C., and Heinzerling, L. (2021). MAPK blockade, toxicities, pathogenesis and management. Curr. Opin. tant MelanomasOncol 33 (2), 139–145. doi:10.1097/CCO.0000000000000710

PubMed Abstract | CrossRef Full Text | Google Scholar

Morré, D. J., Sun, E., Geilen, C., Wu, L. Y., de Cabo, R., Krasagakis, K., et al. (1996). Capsaicin inhibits plasma membrane NADH oxidase and growth of human and mouse melanoma lines. Eur. J. Cancer 32A (11), 1995–2003. doi:10.1016/0959-8049(96)00234-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Mu, X., Wang, L., Zhang, Z., Ge, R., Zhang, J., Liu, W., et al. (2021). Scutellarin suppresses RPMI7951 Melanoma cell proliferation by targeting TOPK. Anticancer Agents Med. Chem. 21 (5), 640–648. doi:10.2174/1871520620666200811112156

PubMed Abstract | CrossRef Full Text | Google Scholar

Murugan, A. K., Munirajan, A. K., and Tsuchida, N. (2013). Genetic deregulation of the PIK3CA oncogene in oral cancer. Cancer Lett. 338 (2), 193–203. doi:10.1016/j.canlet.2013.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Natarelli, N., Boby, A., Aflatooni, S., Tran, J. T., Diaz, M. J., Taneja, K., et al. (2023). Regulatory miRNAs and lncRNAs in skin cancer: a narrative review. Life (Basel) 13 (8), 1696. doi:10.3390/life13081696

PubMed Abstract | CrossRef Full Text | Google Scholar

Naz, I., Merarchi, M., Ramchandani, S., Khan, M. R., Malik, M. N., Sarwar, S., et al. (2020). An overview of the anti-cancer actions of Tanshinones, derived from Salvia miltiorrhiza (Danshen). Explor Target Antitumor Ther. 1 (3), 153–170. doi:10.37349/etat.2020.00010

PubMed Abstract | CrossRef Full Text | Google Scholar

Oakes, S. A. (2017). Endoplasmic reticulum proteostasis: a key checkpoint in cancer. Am. J. Physiol. Cell Physiol. 312 (2), C93-C102–C102. doi:10.1152/ajpcell.00266.2016

PubMed Abstract | CrossRef Full Text | Google Scholar

Ottaviano, M., Giunta, E. F., Tortora, M., Curvietto, M., Attademo, L., Bosso, D., et al. (2021). BRAF gene and melanoma: back to the future. Int. J. Mol. Sci. 22, 3474. doi:10.3390/ijms22073474

PubMed Abstract | CrossRef Full Text | Google Scholar

Pal, H. C., Sharma, S., Strickland, L. R., Katiyar, S. K., Ballestas, M. E., Athar, M., et al. (2014). Fisetin inhibits human melanoma cell invasion through promotion of mesenchymal to epithelial transition and by targeting MAPK and NFκB signaling pathways. PLoS One 9 (1), e86338. doi:10.1371/journal.pone.0086338

PubMed Abstract | CrossRef Full Text | Google Scholar

Palmieri, G., Colombino, M., Casula, M., Manca, A., Mandalà, M., Cossu, A., et al. (2018). Molecular pathways in melanomagenesis: what we learned from next-generation sequencing approaches. Curr. Oncol. Rep. 20, 86. doi:10.1007/s11912-018-0733-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel, S., Trueman, D., Bentley, A., Poole, C., and Chambers, C. (2014). Cost-Effectiveness of Capsaicin 8% patch (Qutenza(tm)) compared with Pregabalin for the treatment of patients with peripheral neuropathic pain (Pnp) in Scotland. Value Health 17 (7), A531. doi:10.1016/j.jval.2014.08.1686

PubMed Abstract | CrossRef Full Text | Google Scholar

Peh, H. Y., Tan, W. S., Liao, W., and Wong, W. S. (2016). Vitamin E therapy beyond cancer: tocopherol versus tocotrienol. Pharmacol. Ther. 162, 152–169. doi:10.1016/j.pharmthera.2015.12.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Pelosi, E., Castelli, G., and Testa, U. (2024). Braf-Mutant melanomas: biology and therapy. Curr. Oncol. 31 (12), 7711–7737. doi:10.3390/curroncol31120568

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramasamy, K., Dwyer-Nield, L. D., Serkova, N. J., Hasebroock, K. M., Tyagi, A., Raina, K., et al. (2011). Silibinin prevents lung tumorigenesis in wild-type but not in iNOS-/- mice: potential of real-time micro-CT in lung cancer chemoprevention studies. Clin. Cancer Res. 17 (4), 753–761. doi:10.1158/1078-0432.CCR-10-2290

PubMed Abstract | CrossRef Full Text | Google Scholar

Reddy, B. Y., Miller, D. M., and Tsao, H. (2017). Somatic driver mutations in melanoma. Cancer 123, 2104–2117. doi:10.1002/cncr.30593

PubMed Abstract | CrossRef Full Text | Google Scholar

Riccardo, F., Iussich, S., Maniscalco, L., Lorda, M. S., La Rosa, G., Arigoni, M., et al. (2014). CSPG4-specific immunity and survival prolongation in dogs with oral malignant melanoma immunized with human CSPG4 DNA. Clin. Cancer Res. 20 (14), 3753–3762. doi:10.1158/1078-0432.CCR-13-3042

PubMed Abstract | CrossRef Full Text | Google Scholar

Rotte, A., Bhandaru, M., Zhou, Y., and McElwee, K. J. (2015). Immunotherapy of melanoma: present options and future promises. Cancer Metastasis Rev. 34, 115–128. doi:10.1007/s10555-014-9542-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Sadek, K. M., Shib, N. A., Taher, E. S., Rashed, F., Shukry, M., Atia, G. A., et al. (2024). Harnessing the power of bee venom for therapeutic and regenerative medical applications: an updated review. Front. Pharmacol. 15, 1412245. doi:10.3389/fphar.2024.1412245

PubMed Abstract | CrossRef Full Text | Google Scholar

Saleh, J. (2018). Murine models of melanoma. Pathol. Res. Pract. 214 (9), 1235–1238. doi:10.1016/j.prp.2018.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanders, B., Ray, A. M., Goldberg, S., Clark, T., McDaniel, H. R., Atlas, S. E., et al. (2017). Anti-cancer effects of aloe-emodin: a systematic review. J. Clin. Transl. Res. 3 (3), 283–296. doi:10.18053/jctres.03.201703.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Scolyer, R. A., Long, G. V., and Thompson, J. F. (2011). Evolving concepts in melanoma classification and their relevance to multidisciplinary melanoma patient care. Mol. Oncol. 5 (2), 124–136. doi:10.1016/j.molonc.2011.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Selimovic, D., Porzig, B. B., El-Khattouti, A., Badura, H. E., Ahmad, M., Ghanjati, F., et al. (2013). Bortezomib/proteasome inhibitor triggers both apoptosis and autophagy-dependent pathways in melanoma cells. Cell Signal 25 (1), 308–318. doi:10.1016/j.cellsig.2012.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Semwal, R. B., Semwal, D. K., Combrinck, S., and Viljoen, A. (2021). Emodin - a natural anthraquinone derivative with diverse pharmacological activities. Phytochemistry 190, 112854. doi:10.1016/j.phytochem.2021.112854

PubMed Abstract | CrossRef Full Text | Google Scholar

Sflakidou, E., Leonidis, G., Foroglou, E., Siokatas, C., and Sarli, V. (2022). Recent advances in natural product-based hybrids as anti-cancer agents. Molecules 27 (19), 6632. doi:10.3390/molecules27196632

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, P., and Allison, J. P. (2015). Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161 (2), 205–214. doi:10.1016/j.cell.2015.03.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Shin, D. H., Kim, O. H., Jun, H. S., and Kang, M. K. (2008). Inhibitory effect of capsaicin on B16-F10 melanoma cell migration via the phosphatidylinositol 3-kinase/Akt/Rac1 signal pathway. Exp. Mol. Med. 240 (5), 486–494. doi:10.3858/emm.2008.40.5.486

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, L., Liu, D., Zhao, Y., He, J., Kang, H., Dai, Z., et al. (2015). Sinomenine inhibits breast cancer cell invasion and migration by suppressing NF-κB activation mediated by IL-4/miR-324-5p/CUEDC2 axis. Biochem. Biophys. Res. Commun. 464 (3), 705–710. doi:10.1016/j.bbrc.2015.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Soo, H. C., Chung, F. F., Lim, K. H., Yap, V. A., Bradshaw, T. D., Hii, L. W., et al. (2018). Cudraflavone C induces tumor-specific apoptosis in colorectal cancer cells through inhibition of the phosphoinositide 3-Kinase (PI3K)-AKT pathway. PLoS One 12 (1), e0170551. doi:10.1371/journal.pone.0170551

PubMed Abstract | CrossRef Full Text | Google Scholar

Soumoy, L., Wells, M., Najem, A., Krayem, M., Ghanem, G., Hambye, S., et al. (2020). Toad venom antiproliferative activities on metastatic melanoma: Bio-Guided fractionation and screening of the compounds of two different venoms. Biol. (Basel). 9 (8), 218. doi:10.3390/biology9080218

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, T. R., Tsai, F. J., Lin, J. J., Huang, H. H., Chiu, C. C., Su, J. H., et al. (2012). Induction of apoptosis by 11-dehydrosinulariolide via mitochondrial dysregulation and ER stress pathways in human melanoma cells. Mar. Drugs 10 (8), 1883–1898. doi:10.3390/md10081883

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Z., Zheng, L., Liu, X., Xing, W., and Liu, X. (2018). Sinomenine inhibits the growth of melanoma by enhancement of autophagy via PI3K/AKT/mTOR inhibition. Drug Des. Devel. Ther. 12, 2413–2421. doi:10.2147/DDDT.S155798

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Q., Gong, T., Liu, M., Ren, S., Yang, H., Zeng, S., et al. (2022). Shikonin, a naphthalene ingredient: therapeutic actions, pharmacokinetics, toxicology, clinical trials and pharmaceutical researches. Phytomedicine Int. J. Phytotherapy Phytopharm. 94, 153805. doi:10.1016/j.phymed.2021.153805

PubMed Abstract | CrossRef Full Text | Google Scholar

Syed, D. N., Chamcheu, J. C., Khan, M. I., Sechi, M., Lall, R. K., Adhami, V. M., et al. (2014). Fisetin inhibits human melanoma cell growth through direct binding to p70S6K and mTOR: findings from 3-D melanoma skin equivalents and computational modeling. Biochem. Pharmacol. 89 (3), 349–360. doi:10.1016/j.bcp.2014.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Umar, S. A., Dong, B., Nihal, M., and Chang, H. (2022). Frizzled receptors in melanomagenesis: from molecular interactions to target identification. Front. Oncol. 12, 1096134. doi:10.3389/fonc.2022.1096134

PubMed Abstract | CrossRef Full Text | Google Scholar

Urra, H., Dufey, E., Lisbona, F., Rojas-Rivera, D., and Hetz, C. (2013). When ER stress reaches a dead end. Biochim. Biophys. Acta 1833 (12), 3507–3517. doi:10.1016/j.bbamcr.2013.07.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Velavan, B., Divya, T., Sureshkumar, A., and Sudhandiran, G. (2018). Nano-chemotherapeutic efficacy of (-) -epigallocatechin 3-gallate mediating apoptosis in A549 cells: involvement of reactive oxygen species mediated Nrf2/Keap1signaling. Biochem. Biophys. Res. Commun. 503 (3), 1723–1731. doi:10.1016/j.bbrc.2018.07.105

PubMed Abstract | CrossRef Full Text | Google Scholar

Voronkov, A., and Krauss, S. (2013). Wnt/beta-catenin signaling and small molecule inhibitors. Curr. Pharm. Des. 19 (4), 634–664. doi:10.2174/138161213804581837

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., and Ma, Q. (2018). Clinical benefits and pharmacology of scutellarin: a comprehensive review. Pharmacol. Ther. 190, 105–127. doi:10.1016/j.pharmthera.2018.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H. M., Yang, H. L., Thiyagarajan, V., Huang, T. H., Huang, P. J., Chen, S. C., et al. (2017). Coenzyme Q0 enhances ultraviolet B-Induced apoptosis in human Estrogen receptor-positive breast (MCF-7) cancer cells. Integr. Cancer Ther. 16 (3), 385–396. doi:10.1177/1534735416673907

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., Yan, W., Sun, Y., and Yang, C. S. (2022). δ-Tocotrienol is the Most potent vitamin E form in inhibiting prostate cancer cell growth and inhibits prostate carcinogenesis in Ptenp-/- mice. Cancer Prev. Res. (Phila) 15 (4), 233–245. doi:10.1158/1940-6207.capr-21-0508

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Yang, S., Li, Y., Jin, X., Lu, J., and Wu, M. (2023). Role of emodin in atherosclerosis and other cardiovascular diseases: pharmacological effects, mechanisms, and potential therapeutic target as a phytochemical. Biomed. and Pharmacother. 161, 114539. doi:10.1016/j.biopha.2023.114539

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, M., Zhang, Y., Ni, S., Sun, M., Wu, Q., Wu, X., et al. (2025). The anti-cancer activity of Dioscin: an update and future perspective. Med. Oncol. 42 (3), 63. doi:10.1007/s12032-024-02572-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Wawruszak, A., Czerwonka, A., Okła, K., and Rzeski, W. (2016). Anticancer effect of ethanol Lycium barbarum (Goji berry) extract on human breast cancer T47D cell line. Nat. Prod. Res. 30 (17), 1993–1996. doi:10.1080/14786419.2015.1101691

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, J., Yan, Y., Chen, X., Qian, L., Zeng, S., Li, Z., et al. (2019). The roles of plant-derived triptolide on non-small cell lung cancer. Oncol. Res. 27 (7), 849–858. doi:10.3727/096504018X15447833065047

PubMed Abstract | CrossRef Full Text | Google Scholar

Wen, Y., Wang, Y., Liu, X., Zhang, W., Xiong, X., Han, Z., et al. (2017). Camptothecin-based nanodrug delivery systems. Cancer Biol. Med. 14 (4), 363–370. doi:10.20892/j.issn.2095-3941.2017.0099

PubMed Abstract | CrossRef Full Text | Google Scholar

Wen, T., Song, L., and Hua, S. (2021). Perspectives and controversies regarding the use of natural products for the treatment of lung cancer. Cancer Med. 10 (7), 2396–2422. doi:10.1002/cam4.3660

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilczak, M., Surman, M., and PrzybyłO, M. (2025). Melanoma-derived extracellular vesicles transfer proangiogenic factors. Oncol. Res. 33 (2), 245–262. doi:10.32604/or.2024.055449

PubMed Abstract | CrossRef Full Text | Google Scholar

Wroblewski, D., Jiang, C. C., Croft, A., Farrelly, M. L., Zhang, X. D., and Hersey, P. (2013). OBATOCLAX and ABT-737 induce ER stress responses in human melanoma cells that limit induction of apoptosis. PLoS One 8 (12), e84073. doi:10.1371/journal.pone.0084073

PubMed Abstract | CrossRef Full Text | Google Scholar

Yakovian, O., Sajman, J., Arafeh, R., Neve-Oz, Y., Alon, M., Samuels, Y., et al. (2021). MEK inhibition reverses aberrant signaling in Melanoma cells through reorganization of NRas and BRAF in self nanoclusters. Cancer Res. 81 (5), 1279–1292. doi:10.1158/0008-5472.CAN-20-1205

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, A. S., and Chapman, P. B. (2009). The history and future of chemotherapy for melanoma. Hematol. Oncol. Clin. North Am. 23 (3), 583–597. doi:10.1016/j.hoc.2009.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, T. Y., Wu, M. L., Chang, C. I., Liu, C. I., Cheng, T. C., and Wu, Y. J. (2018a). Bornyl cis-4-Hydroxycinnamate suppresses cell metastasis of melanoma through FAK/PI3K/Akt/mTOR and MAPK signaling pathways and inhibition of the epithelial-to-mesenchymal transition. Int. J. Mol. Sci. 19 (8), 2152. doi:10.3390/ijms19082152

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, T. Y., Wu, Y. J., Chang, C. I., Chiu, C. C., and Wu, M. L. (2018b). The effect of bornyl cis-4-Hydroxycinnamate on Melanoma cell apoptosis is associated with mitochondrial dysfunction and endoplasmic reticulum stress. Int. J. Mol. Sci. 19 (5), 1370. doi:10.3390/ijms19051370

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, R., Gao, W., Wang, Z., Jian, H., Peng, L., Yu, X., et al. (2024). Polyphyllin I induced ferroptosis to suppress the progression of hepatocellular carcinoma through activation of the mitochondrial dysfunction via Nrf2/HO-1/GPX4 axis. Phytomedicine 122, 155135. doi:10.1016/j.phymed.2023.155135

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, R., Li, T., Tian, J. X., Xi, P., and Liu, R. H. (2018). Ursolic acid, a potential anticancer compound for breast cancer therapy. Crit. Rev. Food Sci. Nutr. 58 (4), 568–574. doi:10.1080/10408398.2016.1203755

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, Z., Zhang, J., Chen, L., Guo, Q., Yang, B., Zhang, W., et al. (2020). Anticancer effects and mechanisms of action of plumbagin: review of research advances. Biomed. Res. Int. 2020, 6940953. doi:10.1155/2020/6940953

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Yoshida, T., and Kuroiwa, Y. (1992). Stimulation of melanin synthesis of B16-F10 mouse melanoma cells by bufalin. Life Sci. 51 (1), 17–24. doi:10.1016/0024-3205(92)90213-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, T., Dutton-Regester, K., Brown, K. M., and Hayward, N. K. (2016). The genomic landscape of cutaneous melanoma. Pigment. Cell Melanoma Res. 29, 266–283. doi:10.1111/pcmr.12459

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y. S., Han, J. Y., Iqbal, O., and Liang, A. H. (2018). Research advances and prospects on mechanism of sinomenin on histamine release and the binding to histamine receptors. Int. J. Mol. Sci. 20 (1), 70. doi:10.3390/ijms20010070

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, R., Wang, Z., You, W., Zhou, F., Guo, Z., Qian, K., et al. (2020). Suppressive effects of plumbagin on the growth of human bladder cancer cells via PI3K/AKT/mTOR signaling pathways and EMT. Cancer Cell Int. 20, 520. doi:10.1186/s12935-020-01607-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Zhang, A., Gupte, A. A., and Hamilton, D. J. (2021). Plumbagin elicits cell-specific cytotoxic effects and metabolic responses in Melanoma cells. Pharmaceutics 13 (5), 706. doi:10.3390/pharmaceutics13050706

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Q., Chen, W. W., Sun, X., Qian, D., Tang, D. D., Zhang, L. L., et al. (2022). The versatile emodin: a natural easily acquired anthraquinone possesses promising anticancer properties against a variety of cancers. Int. J. Biol. Sci. 18 (8), 3498–3527. doi:10.7150/ijbs.70447

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, D., Tian, X., Wang, Y., Liu, F., Zhang, J., Wang, H., et al. (2024). Polyphyllin I ameliorates gefitinib resistance and inhibits the VEGF/VEGFR2/p38 pathway by targeting HIF-1a in lung adenocarcinoma. Phytomedicine 129, 155690. doi:10.1016/j.phymed.2024.155690

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, H., Han, L., Jian, Y., Ma, Y., Yan, W., Chen, X., et al. (2018). Resveratrol induces apoptosis in human melanoma cell through negatively regulating Erk/PKM2/Bcl-2 axis. Onco Targets Ther. 11, 8995–9006. doi:10.2147/OTT.S186247

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, Z. P., Tan, H. Y., and Wang, M. (2012). Tyrosinase inhibition constituents from the roots of Morus australis. Fitoterapia 83 (6), 1008–1013. doi:10.1016/j.fitote.2012.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, W., Feng, Z., You, S., Zhang, H., Tao, Z., Wang, Q., et al. (2017). Fisetin inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes through activating SIRT1 and attenuates the progression of osteoarthritis in mice. Int. Immunopharmacol. 45, 135–147. doi:10.1016/j.intimp.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, S., Wang, X., Zhao, D., Liu, H., and Hu, Y. (2023). Calcium homeostasis and cancer: insights from endoplasmic reticulum-centered organelle communications. Trends Cell Biol. 33 (4), 312–323. doi:10.1016/j.tcb.2022.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: melanoma, natural products, signaling pathway, apoptosis, biological activity

Citation: Gao H, Huang J, Zhang D, Li S, Long S and Qin Y (2025) Natural products as therapeutics for malignant melanoma: preclinical evidence and mechanism . Front. Pharmacol. 16:1641838. doi: 10.3389/fphar.2025.1641838

Received: 05 June 2025; Accepted: 05 August 2025;
Published: 26 August 2025.

Edited by:

Shashanka Prasad, JSS Academy of Higher Education and Research, India

Reviewed by:

Jayaprakash N. Kolla, Institute of Molecular Genetics (ASCR), Czechia
Wenfu Ma, Beijing University of Chinese Medicine, China

Copyright © 2025 Gao, Huang, Zhang, Li, Long and Qin. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yuesi Qin, cWlueXVlc2lAY2R1dGNtLmVkdS5jbg==

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.