The Molecular Actions of Myricetin and Targeting Cell Signalling Pathways in Cancer Treatment

 

Sk Mahbub Alam1, Aishwarya Mondal1, Subhajit Mukherjee1, Saikat Prodhan1, Snehasis Jana2, Sanchari Chatterjee3, Rohan Pal4*

1Department of Pharmacology, Global College of Pharmaceutical Technology, Krishnagar,

Nadia, West Bengal, 741102.

2Assistant Professor, Department of Pharmacology, Global College of Pharmaceutical Technology,

Krishnagar, Nadia, West Bengal, 741102.

3Assistant Professor, Department of Pharmaceutics, Global College of Pharmaceutical Technology,

Krishnagar, Nadia, West Bengal, 741102.

4Assistant Professor, School of Pharmacy, Sister Nivedita University, DG Block (Newtown),

Chakpachuria, West Bengal 700156.

*Corresponding Author E-mail: rohan.p@snuniv.ac.in

 

ABSTRACT:

Numerous biological pathways contribute to the formation and progression of cancer, making it a complex illness. Different molecular participants in various cell signalling cascades must be the focus of effective cancer medications. Myricetin, an isoflavone found in various fruits, vegetables, nuts, berries, and herbs, has been shown to modulate key pathways promoting the survival of cancerous cells. However, its limited bioavailability, pH, and poor water solubility make it difficult to employ clinically. Research is ongoing to develop nano-formulations to enhance bioavailability and absorption, but further research is needed to determine the most effective targeting of the cellular network. Myricetin, a natural flavonoid, has drawn interest because of its anti-inflammatory, anti-oxidant, and anti-cancer properties. Modern molecular methods have illuminated the relationship between dysregulated signal and myricetin. Pathways in the invasion, spread, and development of cancer. However, there is currently limited information available regarding myricetin nano-delivery systems in cancer. In this evaluation, we have included every item in great depth. Considering myricetin-mediated control over many cellular pathways, its consequences for preventing cancer, preclinical and clinical studies and the nano-formulations that are currently on the market for the treatment of different cancers.

 

KEYWORDS: Myricetin, Cancer, mTOR, Apoptosis, Flavonoid.

 

 


 

INTRODUCTION:

Recent pharmacological studies have shown that myricetin (MYR) possesses a variety of biological properties, such as anti-inflammatory, anticancer, antibacterial, antiviral, and anti-obesity actions, as well as protective benefits on the cardiovascular system, against neurological disorders, and damage, and shields the liver from possible harm1–10. The cellular machinery is always under stress during cancer metastasis because a vast number of interactions are occurring across a broad molecular landscape11–13. A comprehensive understanding of the sequence of concurrent molecular activities taking place in tumour cells is necessary to develop an effective medication and delivery system14,15. Due to a number of issues, the cancer treatment regimen now in use is highly hazardous to healthy cells as well as tumours16–19. According to studies, naturally occurring phytochemicals may be used therapeutically to treat long-term illnesses in people20–22. The phytochemicals target human long non-coding RNAs (lncRNAs) using RNA interference technology. The most widely distributed lncRNAs are PVTI, HI9, ROR, NEAT1.

 

These lncRNAs are influenced by a variety of phytochemicals, such as resveratrol, sulforaphane, curcumin, and epigallocatechin gallate (EGCG). Patients may also receive a combined therapy consisting of traditional chemotherapy drugs and phytochemicals to increase the effectiveness of the treatment23,24. Numerous natural sources contain the isoflavonoid MYR, which has been shown in several studies to have antioxidant and free radical scavenging qualities25,26.

 

MYR has not yet received market approval as a novel medication. Furthermore, individuals are increasingly interested in discovering natural remedies to fortify the body rather than utilizing chemical medications that are more hazardous and prone to negative effects; this has incentivized experts to advance their studies on MYR.

 

Figure 1. Structure of Myricetin

 

PATHWAYS INVOLVED IN CANCER:

PI3K/AKT and associated mTOR pathway

The PI3K/Akt-related kinase [PIKK] pathway is extended by mTOR, a serine/threonine protein kinase. mTOR is the catalytic subunit of two different protein complexes: mTOR complex 1 [Mtorc1] and Complex 227. mTOR is one of the most important regulators of mammalian metabolism and physiology. In order to control cell growth and proliferation, mTOR signalling is engaged in gene transcription and protein synthesis28. The cell signalling pathway PI3K/Akt is in charge of several processes in a cell, such as growth, survival, proliferation, and differentiation29. The cytoprotective role of MYR is reported in various studies but in different cancer. MYR interact with Akt and directly inhibits the kinase activity of Akt. Biochemically speaking, MYR tries to remove ATP in order to attach to Akt30. Furthermore, it has been documented that MYR inhibits PI3K expression in both in vitro and in vivo investigations31,32. MYR is also involved in the down-regulation of epidermal growth factor receptor (EGFR) to target the PI3K/Akt signalling pathways33. Through their interaction and inhibition of PI3K and Akt phosphorylation, MYR disrupts mTOR signalling activity34,35. MYR is also involving to control the activity of p70s6k1(effector of mTOR pathway)36. It is reported that the SIRT3 can also suppresses the action of Akt via LKB1/AMPK pathway and MYR can up-regulating the protein level of SIRT3 (negative regulator of Akt) through which MYR stop the signal transduction of Akt37,38.

 

Nuclear factor-kappa (NF-kB) beta pathway:

Nuclear factor kappa beta (NF-kB) is an essential pathway for cell survival. This is the pathway which is initiated by TNF-alpha molecules. Though this molecule has the capacity to drive Apoptosis but also TNF molecules can skip the apoptosis and drive cell survival pathways in the form of inflammation by activation of IAP proteins (inhibitors of Apoptosis protein) from there apoptosis is skipped and the cell leads towards the NF-kB pathway or cell survival pathway. The NF-kB pathway regulates the fate of cells toward growth or death and is primarily in charge of inflammatory responses. MYR either directly or indirectly modulates NF-κB signalling39. Lipopolysaccharide (LPS) is a strong inducer of inflammation. MYR suppresses the TLR4-MyD88-NF-κB signalling pathway and the AKT/IKK/NF-κB pathway in LPS-induced mastitis and lung inflammation designs, which in turn reduces the synthesis of inflammatory markers such as TNF-α, IL-6, and IL-1β. As a result, inflammation is decreased40,41. By controlling IκB/NF-Kb, MYR can also lower the levels of oxidative stress and inflammatory cytokines42,43. It reduces its activity and prevents related downstream NF-κB signals from being activated44. MYR may repair LPS-induced cardiomyocyte H9c2 injury by reducing inflammation since it enhances MALAT1 expression, inhibits NF-κB and IκBα phosphorylation, and lowers MCP-1 and IL-6 expression45. By inhibiting Akt, mTOR, IκB/NFκB, and cytokines and chemokines produced in keratinocytes, MYR prevents the activation of COX-2. Finally, MYR inhibits TNF-α and UV light-induced skin inflammation45,46.

 

RAF/RAS/MAPK/ERK pathway:

The Ras/Raf/MAPK/Extracellular-signal-regulated kinase (ERK) cascade is a cabalistic signalling pathway of living system. Depending on the type of signal received, this pathway entangles a variety of molecular entities and modifies the genetic expression linked to a cell's development, advancement, prevention, or induction of apoptosis47,48. Raf/MEK/ERK are directly impacted by MYR, which modifies their kinase activity. In an ATP-noncompetitive way, it binds to Raf and inhibits its function to phosphorylate its downstream molecules49. In a similar manner, MYR causes an EGF stimulus by interfering with MEK1's kinase activity and inhibiting ERK phosphorylation50. Additionally, it lowers the amounts of p38-MAPK protein, as shown by several immunoblotting tests51. The modulation of MYR in certain disorders, the Raf pathway also influences the molecules and related signalling cascades. For example, in cancers, MYR attacks the VEGF expression by the direct inhibition of MKK1 and MKK4. Using in silico docking, the investigation showed that MYR attaches to MKK4's ATP-binding location52. The ability of MYR to control MMPs has been used in research on anti-aging, immunological illness, and cancer therapy49,53,54.

 

JAK-STAT pathway:

One important mediator of cancer progression is JAK-STAT signalling, which can be activated through a variety of mechanisms, including the upregulation of cytokine expression. It can also function as a tumour intrinsic driver of cancer growth through metastasis or as a modulator of immune control55. The JAK-STAT pathway is essential for controlling immunological responses and cell inflammation, and it uses NF-κB signalling to carry out its intended function56. Numerous cancers have been linked to STAT3 activation in different studies. These include blood cancers (such as leukaemia) and solid cancers (such as bladder, breast, medulloblastoma, colon, and cervical cancers)55. MYR interacts with JAK1 inhibits it, which as a result suppresses the activity of the phosphorylated STAT3 molecule57. The attenuation of the STAT3 molecule has also been beneficial in cholangiocarcinoma58.

 

Nrf2 pathway:

In contrast to JAK-STAT, TNFα, and NF-κB signalling, the Nrf2 pathway functions to reduce oxidative stress-induced inflammation and cytotoxicity44. MYR promotes Nrf2 activity while reciprocally suppressing NF-κB function59. MYR inhibits the ubiquitination-mediated degradation of the Nrf2 pathway, which causes Nrf2 to accumulate in the cytoplasm60. MYR is also involved in the accumulation of Nrf2 translocation in nuclei, which activates ARE and increases the production of ARE-associated genes60,61. Activation of the Nrf2-associated HO-1 gene, which in turn inhibits pro-inflammatory cytokines and boosts the synthesis of the anti-inflammatory cytokine IL-1062.

 

TNF- ALPHA signalling pathway

One important cytokine implicated in inflammation, immunology, tumour growth, etc. is tumour necrosis factor alpha (TNFα)63. The main association between inflammation-based cytotoxicity and TNFα signalling in cells64. A variety of structural proteins, including lipopolysaccharides (LPS), which are bound by Toll-like receptors (TLRs) and which actively generate inflammatory cytokines, can cause this release. Toll-like receptors are involved in the development of the innate immune response. MYR directly targets TNFα and interferes with the start of the TNFα/TNFR signalling cascade, which causes the Akt, mTOR, and NF-κB pathways to be down-regulated. It also inhibits the AKT/IKK pathway, which includes the NF-κB pathway, and reduces the synthesis of these inflammatory cytokines1,40. When a cell undergoes TNF signalling pathway the TRAF-2 protein activates a special protein IKK (Inhibitor of Nuclear Factor Kappa-B-Kinase Subunit Alpha) through activation of RIP, the IKK molecule inhibits IKBA as a result NF-kB is released to cause cell survival by corporating inflammation; MYR inhibits the phosphorylation of NF-κB and IκBα, and thus it reduces inflammation through inhibition of gene expression65.

 

Therefore, MYR contributes to cytoprotection by inhibiting TNFα, which destroys signal transduction via all of these pathways. MYR also affects the TNFα adaptor protein, TRAF6. It suppresses the downstream phosphorylation cascade and encourages TRAF6 ubiquitination59.

 

ROS-Apoptosis signalling pathway:

Strong intrinsic stimuli known as reactive oxygen species (ROS) play a role in the process of stress-induced cell death. Therefore, ROS is essential for controlling the primary apoptotic pathways that are regulated by the endoplasmic reticulum, death receptors, and mitochondria66. The Tumour Suppressor Protein P53 is activated by ROS at lower concentrations. By causing cell cycle arrest, this P53 plays a critical role in controlling the regulation of cell stress and promoting DNA repair for either cell survival or apoptosis. ROS activates the P53 protein in cases of cell apoptosis or extremely severe cell damage. This P53 then triggers apoptosis by downregulating pro-survival proteins like Bcl-2, Bcl-Xl, and IAPs and upregulating pro-apoptotic genes that are crucial for controlling the intrinsic pathway of apoptosis, such as Bax, Bid, Puma, and Noxa67. MYR causes tumour cells to produce more ROS, which in turn causes the cell to undergo apoptosis. Research has shown that MYR causes oxidative stress, which raises the amount of ROS in cells, stimulates lipid peroxidation, depletes glutathione, and causes JAR and JEG-3 cells to undergo apoptosis. In order to produce anti-cancer actions, MYR causes ROS to accumulate in cancer cells68.

 

DRUG INTERACTIONS OF MYRICETIN:

MYR generally interacts with the enzymes at the hepatic level.

 

Interaction with CYP2C8:

The metabolism endogenous chemicals, such as the synthesis of epoxyeicosatrienoic acid (EETs), which are thought to be signalling molecules against cancer hallmarks, depends heavily on the enzyme CYP2C8. In an in vitro investigation, MYR demonstrated a significant suppression of CYP2C8 in human liver microsomes with the use of the CYP2C8-catalyzed amodiaquine-N-deethylation enzyme69.

 

Myricetin and cytochrome P450 (CYP):

Cytochrome P450 consist of a large heme enzyme super family, which are responsible for catalyzing the oxidative transformation of various range of organic substances mainly it is responsible for the metabolism of xenobiotics70. MYR has been found to inhibit human Cytochrome P450(CYP) enzymes and is an inhibitor of P-gp in KB/MDR cell line71.

 

Myricetin interacts with hUGTS

A class of enzymes known as UDP-glucuronosyltransferases (UGTs) aids in phase II metabolism and is helpful in the metabolism of endogenous fatty acids, including metabolites of arachidonic acid, some opioids, and antiepileptic and antiviral medications72. Now MYR contributes in food-drug interaction with UDP-glucuronosyltransferase (UGT), studies has shown MYR displayed a broad-spectrum inhibition against the human UGTs, it has shown strong inhibitory effects against UGT1A1, 1A3, 1A6, 1A7, 1A10 while moderate inhibition against and 2B773.

PATHWAYS INVOLVED IN CANCER:

PI3K/AKB/PKB signalling

In response to external cues, the Akt signalling route, also known as the PI3K-Akt signalling system, is a signal transduction mechanism that enhances growth and survival. Phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) are important proteins involved. PI3K is phosphorylated and a cell surface receptor is activated upon initial stimulation by one of the growth factors. The second messenger, phosphatidylinositol (3,4,5)-trisphosphate (PIP3), is created when activated PI3K phosphorylates lipids on the plasma membrane. Through contact with these phosphoinositide docking sites, the serine/threonine kinase Akt is drawn to the membrane for complete activation shown in (figure 1)74. By phosphorylating a variety of intracellular proteins, activated Akt regulates downstream reactions such as angiogenesis, growth, proliferation, migration, and cell survival. The route is extremely conserved and found in all higher eukaryotic cells. The PI3K/AKT pathway is highly responsible for cell growth and cell proliferation. When MYR introduced it inhibit the phosphorylation PI3K, in result, PIP2 is not converted in to PIP3 and AKT/PKB protein doesn’t formed, thus MYR stop the PI3K/AKT pathway and shows its anti-cancer properties75.


 

Figure 1: PI3K/AKB/PKB signalling pathway

 


PI3K/Akt/mTOR pathway:

Numerous methods can initiate signalling through the PI3K/Akt/mTOR pathway, and each of these mechanisms leads to increased pathway activation, which is frequently observed in various cancer subtypes. mTOR, a master regulator of protein translation, is one of the many substrates that PI3K signalling can affect once it is active. Because it is the second most commonly changed pathway after p53 and because it is a convergence point for numerous stimuli, the PI3K/Akt/mTOR pathway is an appealing therapeutic target in cancer. This route regulates important biological functions such transcription, apoptosis, cell cycle progression, and translation through its downstream substrates shown in (figure 2 and 3)76. Eukaryotic cell development and metabolism are regulated by the mechanistic Target of Rapamycin (mTOR), which interacts with growth stimuli and nutrients from the environment. Many basic cell functions, including as protein synthesis and autophagy, are regulated by mTOR, according to extensive research conducted over the past 20 years. Deregulated mTOR signalling is linked to the development of diabetes and cancer as well as the aging process77.

 


 

Figure 2: PI3K/Akt/mTOR pathway in inactive state

 


Figure 3: PI3K/Akt/mTOR pathway in active state


 

Recent Perspectives of Myricetin:

The MTOR pathway is the most important pathway in the case of cancer. This pathway is responsible for the regulation of cell growth and cell proliferation. Once AKT/PKB proteins get activated its block the TSC-1 and TSC-2 protein in result RHEB proteins gets activated and RHEB further activate the MTOR protein. MTOR can inhibit Autophagy through the inhibition of ULK-1 complex and furthermore it activates the p70S6K (effector of MTOR) which inhibit the eEF2K which leads to activation of eIF4B and p70S6K also involved in the phosphorylation of S6, both S6 and eIF4B are involved in protein synthesis. Myricetin inhibits the kinase activity of Akt through direct interaction. It competes with adenosine triphosphate (ATP) for binding with Akt on a biological level78,79. Furthermore, myricetin has been shown in both in vitro and in vivo experiments to target PI3K expression. Additionally, it targets the PI3K/Akt pathway by downregulating the epidermal growth factor receptor (EGFR). Myricetin also prevents the activation of downstream molecular pathways in cancer by targeting PI3K and Akt. Through its interaction and inhibition of PI3K and Akt phosphorylation, it disrupts mTOR activation. Moreover, myricetin also affects the expression of p70s6k1, an effector of the mTOR pathway. It is demonstrated that myricetin regulates the PI3K/Akt/mTOR pathway to cause autophagy-mediated apoptosis in human colorectal cancer cells. Moreover, myricetin inhibits signal transduction via the Akt/mTOR pathway by increasing SIRT3 (Akt negative regulator) protein levels. Additionally, SIRT3 inhibits Akt through the LKB1/AMPK pathway47,80.

 

MYR has been clinically investigated to treat diabetes in conjunction with several phytochemicals. During the course of a 4-week clinical investigation, Blueberin and the dosage of MYR administered was 250mg. 300mg total, made up of 50mg of MYR and blueberry leaves. every day. This trial with a placebo found that Blueberin and MYR together effectively decreased levels of plasma sugar in people with type 2 diabetes. When blueberin was used, plasma sugar levels were lowered from between 143±5.2 and 104±5.7mg/L. Additionally, Blueberin was discovered to lower the levels of glutamyltransferase (GGT), alanine aminotransferase (ALT), and AST in the serum reduced the C-reactive protein (CRP) levels as well. which might eventually stop inflammation81. Emulin, a combination of MYR, quercetin, and chlorogenic acid, has been shown to lower blood glucose levels in people with type 2 diabetes. Forty participants in the Emulin clinical trial had blood sugar fasting (BSF) levels ranging from 126 to 249 mg/mL82. In addition to its function in diabetes, MYR has been investigated for its potential as a chemopreventive agent and as a signalling and cell proliferation inhibitor83. A meta-analysis of 5073 lung cancer patients revealed that MYR and other flavonoids could lower their risk of developing lung cancer by 30–40%84.

 

Bioavailability of Myricetin:

MYR is a weak acidic, lipophilic substance that functions best at a pH of 2.0. It has a low aqueous solubility (16.60g/ml), which renders it insoluble in the GI tract and limits its effectiveness when taken orally68. Optimal gut absorption and solubility are the two obstacles that prevent medications from being helpful in treating cancer, even with clinical advancements in human health and cancer treatment. Additionally, a drug's effectiveness depends on its concentration and mode of delivery47. Some recent researches attempted to improve the efficacy of MYR by the drug delivery designing system based on Nanotechnology85. At two oral dosages of 50 and 100mg/kg, MYR's oral bioavailability is only roughly 9.62% and 9.74%, respectively, suggesting that it has low absorption capabilities. Additionally, the gastrointestinal environment and numerous other factors can readily affect the stability of MYR86.

 

Pharmacological Effect of Myricetin in Different Cancer:

Prostate Cancer:

Prostate cancer (PCa) ranks sixth globally in terms of cancer-related mortality among men and is the second most common malignancy to be diagnosed. The bark and leaves of the bayberry are rich in the flavonoid MYR, which has been linked to a decreased risk of prostate cancer and other malignancies, according to epidemiological studies. The proviral integration site for Moloney murine leukaemia virus-1 (PIM1), a Ser/Thr protein kinase of the PIM family, is a proto-oncogene protein that is overexpressed in PCa. PIM1 plays a role in carcinogenesis, castration resistance, and PCa metastases. Additionally, PIM1 facilitates the phosphorylation and surface expression of CXCR4, supporting the CXCL12–CXCR4 axis and significantly accelerating the development and metastasis of cancer. Comparative molecular field and cell-free crystallographic investigations revealed that MYR binds to PIM1's ATP-binding pocket and further suppresses its kinase activity. In PCa cells, MYR selectively disrupted the PIM1/CXCR4 connection and inhibited PIM1, hence causing apoptosis and anti-metastasis87–89.

 

Hepatocellular Carcinoma:

In 2020, hepatocellular carcinoma (HCC) will rank third globally in terms of cancer-related mortality and be the sixth most prevalent type of cancer to be diagnosed. Since HCC progresses quickly and is difficult to diagnose early, most individuals are diagnosed when the cancer is intermediate or advanced. For those individuals, systemic medication and trans-arterial chemoembolization (TACE) are the primary treatment choices. Currently available chemotherapeutic medications, including sorafenib, frequently cause numerous toxicities and induce treatment resistance. For patients with advanced HCC, there is no effective systemic therapy available. Therefore, in order to treat HCC, it is imperative to investigate more effective medications with fewer side effects and low toxicity. One type of naturally occurring flavonoid found in many plants, including fruits, nuts, and vegetables, is MYR. MYR exhibits a range of biological actions, including analgesic, anti-inflammatory, and antioxidant properties. Furthermore, some data indicate that MYR may have anticancer properties against a variety of malignancies, including hepatocellular carcinoma90. In HepG2 and Huh-7 cells, MYR caused apoptosis and inhibited cell growth. MYR promoted the phosphorylation and subsequent degradation of YAP, which decreased its expression. By promoting the kinase activity of LATS1/2, MYR suppressed the expression of YAP. The phosphorylation and degradation of YAP caused by MYR were lessened by LATS1/2 knockdown expression using shRNA. Moreover, MYR inhibited YAP and its target genes in vitro and in vivo, sensitizing HCC cells to cisplatin therapy. The discovery that MYR targets the LATS1/2-YAP pathway may aid in the development of innovative preventative and therapeutic approaches for human HCC91.

 

Oesophageal Cancer:

MYR stopped apoptosis in the esophageal cancer cell lines and inhibited both invasion and proliferation. RSK2 is bound by MYR via the NH2-terminal kinase domain, it was also found. Moreover, MYR was discovered to inhibit the growth of KYSE30 and EC9706 cells via Mad1 and to induce cell death through Bad. MYR induces apoptosis and reduces the proliferative and invasive capacity of KYSE30 and EC9706 cells via RSK2. These results offer new information about the potential of MYR as an esophageal cancer preventative and therapeutic agent92.

 

Bladder Cancer:

Cell growth rates were dramatically reduced when MYR (40 or 80µM) was used. The bladder cancer cell (T24) underwent a concentration arrest of the cell cycle at the G2/M phase as a result of MYR treatments. The nuclei of the control cells were visible. The cells were exposed to either 40 or 80µM MYR showed a notable amount of nuclear fragmentation, a hallmark of apoptosis. Treatment with MYR at a daily dose of 5 mg/kg demonstrated anticancer effects. In patients with bladder cancer, MYR treatment showed a higher survival rate compared to the control group93.

 

Breast Cancer:

To assess potential modes of action, the apoptotic effect of MYR was examined in breast cancer cells (MCF-7). When MYR was applied to breast cancer cells, there was a significant increase in the expression of genes linked to apoptosis, such as caspase-3, caspase-8, and caspase-9, as well as the ratio of BAX to Bcl-2, GADD45 and p53. By inducing both intrinsic and extrinsic apoptotic pathways, MYR efficiently induces apoptosis in breast cancer cells. MYR may cause breast cancer (MCF-7) cells to undergo apoptosis by triggering the BRCA1-GADD45 pathway94–97.

 

Ovarian Cancer:

One of the most frequent cancer-related deaths among women worldwide is ovarian cancer, which also ranks second in terms of gynaecological cancer-related deaths98. The ideal concentration range for MYR's suppression of SKOV3 growth was 1×10-5-1×10-4 M for 24 hours, according to CCK-8 cell viability/cytotoxicity experiments. MYR was not harmful to IOSE-80, non-tumor cells at these concentrations, but it did have a dose-dependent inhibitory effect on SKOV-3 cells. MYR-treated SKOV3 cells had a reduced cell size, a smaller appearance, and a high rate of cell death. Moreover, MYR treatment at 10, 20, and 40 µM significantly reduced the ROS levels in these cancer cells in a dose-dependent way, as well as intracellular MDA while increasing SOD levels. In a dose-dependent manner, MYR therapy significantly reduced the amount of SKOV3 cells penetrating matrigel and moving downward in comparison to controls99,100.

 

It was shown that treating OVCAR3 and A2780 cells with MYR accelerated apoptosis. Apoptotic signal increased roughly 2.5 times in A2780 cells treated with 25 µM MYR compared to untreated cells, and nearly 4 times in OVCAR3 cells. The results support the idea that MYR-induced cytotoxicity in ovarian cancer cells involves apoptosis. Furthermore, it was shown that, in contrast to untreated cells, MYR-treated ovarian cancer cells expressed significantly more of the pro-apoptotic protein BAX (B-cell lymphoma-2-associated X-protein), and significantly less of the anti-apoptotic protein Bcl-2101.

 

Pancreatic cancer:

Pancreatic cancer is the fourth most prevalent cause of cancer-related deaths, and it has a very bad prognosis. Finding new treatments is crucial because it is resistant to both new treatment techniques and conventional chemotherapy medications. Research has demonstrated that the flavonoid MYR reduced PI3 kinase activity and caused pancreatic cancer cells to die in vitro by apoptosis. MYR therapy for orthotopic pancreatic cancers in vivo led to tumor regression and a reduction in the spread of metastases. Crucially, MYR proved non-toxic in both in vivo and in vitro settings, highlighting its potential as a pancreatic cancer treatment32.

 

Gastric Cancer:

Cell contraction, nucleus condensation, and bubble formation in the cell membrane are the hallmarks of apoptosis, a sort of controlled cell death. Apoptosis is a representative anticancer strategy since it can cause mutant cells to become malignant when apoptotic pathways are inhibited (Elmore, 2007). Phosphoinositide 3-kinase (PI3K), an upstream kinase, initiates the signalling pathway that activates protein kinase B (Akt), a serine/threonine kinase that is essential for cell survival and proliferation. When there is an adequate supply of nutrients and energy, AKt can phosphorylate and activate the mammalian target of rapamycin (mTOR) protein, which can then integrate signals from growth factors to stimulate cell development. Numerous cancers, particularly gastric cancer, are linked to an aberrant rise in the PI3K/Akt/mTOR pathway's activity102. In both in vitro and in vivo investigations, MYR has been shown to target PI3K expression. Additionally, it targets the PI3K/Akt pathway by downregulating the EGFR. Additionally, MYR alters the expression of p70s6k1, an effector of the mTOR pathway. Moreover, MYR inhibits signal transduction via the Akt/mTOR pathway by increasing SIRT3 (Akt negative regulator) protein levels. Additionally, SIRT3 inhibits Akt through the LKB1/AMPK pathway47.


 

Figure 4: Overview of pharmacological effect of myricetin in different cancer.

 


CONCLUSION:

Since cancer is a complex illness, the formation and progression of the disease are influenced at the same time by a number of cellular pathways that are essential for cell growth, survival, and proliferation. Cancer treatments that are effective must be able to target several molecular actors involved in various cell signalling cascades. An isofavonoid called MYR is typically found as a glycoside in a variety of berries, vegetables, fruits, nuts, and plants. Additionally, it has been discovered to be a significant component of wine, tea, and a few therapeutic plants. This paper discusses the modulation of cellular pathways by MYR in humans, including PI3K/Akt, nrf signalling, mTOR, Ras/Raf, and JAK/STAT pathways, highlighting its potential as a cancer therapeutic solution. However, its low bioavailability, pH and poor water solubility hinder its clinical use. Research is underway to develop nano-formulations of MYR to improve its bioavailability and absorption. However, clinical trials and human use require significant efforts. Understanding MYR's interaction with non-coding RNAs could lead to smart cancer cell targeting. Nano formulations have shown promise in enhancing bioavailability and efficacy. One possible and potentially effective treatment approach is the addition of MYR to nano formulations. MYR nano formulation's dosage safety will be further investigated, which will advance the drug's clinical use.

 

CONFLICT OF INTEREST:

The authors have no conflicts of interest regarding this investigation.

 

ACKNOWLEDGMENTS:

The authors would like to thank Dr. N. N. Bala and Dr. Arin Bhattacharjee for his valuable resources and opinions.

 

REFERENCES:

1.   Hou W, Hu S, Su Z, et al. Myricetin attenuates LPS-induced inflammation in RAW 264.7 macrophages and mouse models. Future Med Chem. 2018; 10(19): 2253-2264. doi:10.4155/FMC-2018-0172,

2.   Jiang M, Zhu M, Wang L, Yu S. Anti-tumor effects and associated molecular mechanisms of myricetin. Biomedicine and Pharmacotherapy. 2019; 120. doi: 10.1016/j.biopha.2019.109506

3.   Stoll S, Bitencourt S, Laufer S, Inęs Goettert M. Myricetin inhibits panel of kinases implicated in tumorigenesis. Basic Clin Pharmacol Toxicol. 2019; 125(1): 3-7. doi:10.1111/BCPT.13201; Ctype: String: Journal

4.   Jiang S, Tang X, Chen M, et al. Design, synthesis and antibacterial activities against Xanthomonas oryzae pv. oryzae, Xanthomonas axonopodis pv. Citri and Ralstonia solanacearum of novel myricetin derivatives containing sulfonamide moiety. Pest Manag Sci. 2020; 76(3): 853-860. doi:10.1002/PS.5587

5.   Ortega JT, Suárez AI, Serrano ML, Baptista J, Pujol FH, Rangel HR. The role of the glycosyl moiety of myricetin derivatives in anti-HIV-1 activity in vitro. AIDS Res Ther. 2017; 14(1). doi:10.1186/S12981-017-0183-6

6.   Ren R, Yin S, Lai B, et al. Myricetin antagonizes semen-derived enhancer of viral infection (SEVI) formation and influences its infection-enhancing activity. Retrovirology. 2018; 15(1). doi:10.1186/S12977-018-0432-3

7.   Hu T, Yuan X, Wei G, Luo H, Lee HJ, Jin W. Myricetin-induced brown adipose tissue activation prevents obesity and insulin resistance in db/db mice. Eur J Nutr. 2018; 57(1): 391-403. doi:10.1007/S00394-017-1433-Z

8.   Wang L, Wu H, Yang F, Dong W. The protective effects of myricetin against cardiovascular disease. J Nutr Sci Vitaminol (Tokyo). 2019;65(6):470-476. doi:10.3177/JNSV.65.470

9.   Ahmed S, Khan H, Aschner M, Hasan MM, Hassan STS. Therapeutic potential of naringin in neurological disorders. Food and Chemical Toxicology. 2019; 132. doi: 10.1016/j.fct.2019.110646

10. Guo C, Xue G, Pan B, et al. Myricetin Ameliorates Ethanol-Induced Lipid Accumulation in Liver Cells by Reducing Fatty Acid Biosynthesis. Mol Nutr Food Res. 2019; 63(14). doi:10.1002/MNFR.201801393,

11. Sharma R, Abbasi-Kangevari M, Abd-Rabu R, et al. Global, regional, and national burden of colorectal cancer and its risk factors, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Gastroenterol Hepatol. 2022; 7(7):627-647. doi:10.1016/S2468-1253(22)00044-9

12. Mitrut P, Docea A, Kamal A, Mitrut R, Calina D. Colorectal cancer and inflammatory bowel disease. Published online 2016. Accessed May 1, 2025.

13. Yadav AR, Mohite SK. Cancer-A silent killer: An overview. Asian Journal of Pharmaceutical Research. 2020; 10(3): 213. doi:10.5958/2231-5691.2020.00036.2

14. Sharifi-Rad J, Quispe C, Butnariu M, et al. Chitosan nanoparticles as a promising tool in nanomedicine with particular emphasis on oncological treatment. Cancer Cell Int. 2021; 21(1). doi:10.1186/S12935-021-02025-4

15. Sharifi-Rad J, Quispe C, Patra JK, et al. Paclitaxel: Application in Modern Oncology and Nanomedicine-Based Cancer Therapy. Oxid Med Cell Longev. 2021; 2021(1): 3687700. doi:10.1155/2021/3687700

16. Cano A, Espina M, García ML. Recent Advances on Antitumor Agents-loaded Polymeric and Lipid-based Nanocarriers for the Treatment of Brain Cancer. Curr Pharm Des. 2020; 26(12): 1316-1330. doi:10.2174/1381612826666200116142922

17. Kamble DD, Maner SS, Jadhav PP. Conventional and Modern Techniques to Treat Cancer. Asian Journal of Pharmaceutical Research. Published online September 20, 2024: 303-308. doi:10.52711/2231-5691.2024.00047

18. Jisha K, Venkateswaramurthy N, Sambathkumar R. The Influence of Pharmacogenetics in Cancer Chemotherapy. Research Journal of Pharmacology and Pharmacodynamics. 2020; 12(1): 29-33. doi:10.5958/2321-5836.2020.00007.5

19. Grace CSrP, Tomy AT, Benny A, et al. Knowledge of Adolescents regarding Cancer Prevention and Factors Contributing to Cancer. Asian Journal of Nursing Education and Research. 2019; 9(4): 515. doi:10.5958/2349-2996.2019.00109.5

20. Calina D, Buga AM, Mitroi M, et al. The treatment of cognitive, behavioural and motor impairments from brain injury and neurodegenerative diseases through cannabinoid system modulation—evidence from in vivo studies. J Clin Med. 2020; 9(8): 1-28. doi:10.3390/JCM9082395

21. Salehi B, Quispe C, Chamkhi I, et al. Pharmacological Properties of Chalcones: A Review of Preclinical Including Molecular Mechanisms and Clinical Evidence. Front Pharmacol. 2021; 11: 592654. doi:10.3389/FPHAR.2020.592654

22. Sharifi-Rad J, Quispe C, Shaheen S, et al. Flavonoids as potential anti-platelet aggregation agents: from biochemistry to health promoting abilities. Crit Rev Food Sci Nutr. 2022;62(29):8045-8058. doi:10.1080/10408398.2021.1924612

23. Mishra S, Verma SS, Rai V, et al. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell Mol Life Sci. 2019; 76(10): 1947. doi:10.1007/S00018-019-03053-0

24. Newman DJ, Cragg GM. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J Nat Prod. 2020; 83(3): 770-803. doi: 10.1021/ACS.JNATPROD.9B01285

25. Bahaloo M, Rezvani ME, Yazd EF, et al. Effect of myricetin on the gene expressions of NOX3, TGF-β1, prestin, and HSP-70 and anti-oxidant activity in the cochlea of noise-exposed rats. Iran J Basic Med Sci. 2020; 23(5): 594. doi:10.22038/IJBMS.2020.41007.9693

26. Zhang Q, Zhao Y, Zhang M, Zhang Y, Ji H, Shen L. Recent advances in research on vine tea, a potential and functional herbal tea with dihydromyricetin and myricetin as major bioactive compounds. J Pharm Anal. 2020; 11(5): 555. doi: 10.1016/J.JPHA.2020.10.002

27. Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell. 2017; 168(6): 960. doi: 10.1016/J.CELL.2017.02.004

28. Zou Z, Tao T, Li H, Zhu X. MTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 2020; 10(1): 1-11. doi:10.1186/S13578-020-00396-1/TABLES/2

29. Ah Kang K, Wang ZH, Zhang R, et al. Myricetin Protects Cells against Oxidative Stress-Induced Apoptosis via Regulation of PI3K/Akt and MAPK Signaling Pathways. Int J Mol Sci. 2010; 11(11):4348. doi:10.3390/IJMS11114348

30. Kumamoto T, Fujii M, Hou DX. Akt is a direct target for myricetin to inhibit cell transformation. Mol Cell Biochem. 2009;332(1-2):33-41. doi:10.1007/S11010-009-0171-9

31. Jung SK, Lee KW, Byun S, et al. Myricetin inhibits UVB-induced angiogenesis by regulating PI-3 kinase in vivo. Carcinogenesis. 2009; 31(5): 911-917. doi:10.1093/CARCIN/BGP221

32. Phillips PA, Sangwan V, Borja-Cacho D, Dudeja V, Vickers SM, Saluja AK. Myricetin Induces Pancreatic Cancer Cell Death via the Induction of Apoptosis and Inhibition of the Phosphatidylinositol 3-Kinase (PI3K) Signaling Pathway. Cancer Lett. 2011; 308(2): 181. doi:10.1016/J.CANLET.2011.05.002

33. Li W, Xu C, Hao C, et al. Inhibition of herpes simplex virus by myricetin through targeting viral gD protein and cellular EGFR/PI3K/Akt pathway. Antiviral Res. 2020; 177: 104714. doi:10.1016/J.ANTIVIRAL.2020.104714

34. Cao J, Chen H, Lu W, et al. Myricetin Induces Protective Autophagy by Inhibiting the Phosphorylation of mTOR in HepG2 Cells. Anatomical Record. 2018; 301(5): 786-795. doi:10.1002/AR.23754

35. Kim GD. Myricetin Inhibits Angiogenesis by Inducing Apoptosis and Suppressing PI3K/Akt/mTOR Signaling in Endothelial Cells. J Cancer Prev. 2017; 22(4): 219. doi:10.15430/JCP.2017.22.4.219

36. Zhang XH, Chen SY, Tang L, et al. Myricetin Induces Apoptosis in HepG2 Cells Through Akt/p70S6K/Bad Signaling and Mitochondrial Apoptotic Pathway. Anticancer Agents Med Chem. 2013; 13(10): 1575-1581. doi:10.2174/1871520613666131125123059

37. Quan Y, Wang N, Chen Q, et al. SIRT3 inhibits prostate cancer by destabilizing oncoprotein c-MYC through regulation of the PI3K/Akt pathway. Oncotarget. 2015; 6(28): 26494-26507. doi:10.18632/ONCOTARGET.4764

38. Wang G, Wang JJ, Wang YZ, Feng S, Jing G, Fu XL. Myricetin nanoliposomes induced SIRT3-mediated glycolytic metabolism leading to glioblastoma cell death. Artif Cells Nanomed Biotechnol. 2018; 46(sup3): S180-S191. doi:10.1080/21691401.2018.1489825

39. Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017; 2(1):1-9. doi:10.1038/SIGTRANS.2017.23

40. Zhu M mao, Wang L, Yang D, et al. Wedelolactone alleviates doxorubicin-induced inflammation and oxidative stress damage of podocytes by IκK/IκB/NF-κB pathway. Biomedicine and Pharmacotherapy. 2019; 117. doi:10.1016/j.biopha.2019.109088

41. Kan X, Liu B, Guo W, et al. Myricetin relieves LPS-induced mastitis by inhibiting inflammatory response and repairing the blood–milk barrier. J Cell Physiol. 2019; 234(9): 16252-16262. doi:10.1002/JCP.28288

42. Qiu Y, Cong N, Liang M, Wang Y, Wang J. Systems Pharmacology Dissection of the Protective Effect of Myricetin Against Acute Ischemia/Reperfusion-Induced Myocardial Injury in Isolated Rat Heart. Cardiovasc Toxicol. 2017; 17(3): 277-286. doi:10.1007/S12012-016-9382-Y

43. Zhang N, Feng H, Liao HH, et al. Myricetin attenuated LPS induced cardiac injury in vivo and in vitro. Phytotherapy Research. 2018; 32(3): 459-470. doi:10.1002/PTR.5989

44. Ahmed SMU, Luo L, Namani A, Wang XJ, Tang X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim Biophys Acta Mol Basis Dis. 2017; 1863(2): 585-597. doi:10.1016/J.BBADIS.2016.11.005

45. Shih YW, Wu PF, Lee YC, Shi M Der, Chiang TA. Myricetin suppresses invasion and migration of human lung adenocarcinoma a549 cells: Possible mediation by blocking the erk signaling pathway. J Agric Food Chem. 2009; 57(9): 3490-3499. doi:10.1021/JF900124R

46. Xie J, Zheng Y. Myricetin protects keratinocyte damage induced by UV through IκB/NFκb signaling pathway. J Cosmet Dermatol. 2017;16(4):444-449. doi:10.1111/JOCD.12399

47. Javed Z, Khan K, Herrera-Bravo J, et al. Myricetin: targeting signaling networks in cancer and its implication in chemotherapy. Cancer Cell International 2022 22:1. 2022; 22(1): 1-13. doi:10.1186/S12935-022-02663-2

48. Chang F, Steelman LS, Lee JT, et al. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: Potential targeting for therapeutic intervention. Leukemia. 2003; 17(7): 1263-1293. doi:10.1038/SJ.LEU.2402945

49. Jung SK, Lee KW, Kim HY, et al. Myricetin suppresses UVB-induced wrinkle formation and MMP-9 expression by inhibiting Raf. Biochem Pharmacol. 2010; 79(10): 1455-1461. doi:10.1016/J.BCP.2010.01.004

50. Lee KW, Kang NJ, Rogozin EA, et al. Myricetin is a novel natural inhibitor of neoplastic cell transformation and MEK1. Carcinogenesis. 2007; 28(9): 1918-1927. doi:10.1093/CARCIN/BGM110

51. Zhou Z, Mao W, Li Y, Qi C, He Y. Myricetin Inhibits Breast Tumor Growth and Angiogenesis by Regulating VEGF/VEGFR2 and p38MAPK Signaling Pathways. Anatomical Record. 2019; 302(12): 2186-2192. doi:10.1002/AR.24222

52. Javed Z, Khan K, Herrera-Bravo J, et al. Myricetin: targeting signaling networks in cancer and its implication in chemotherapy. Cancer Cell Int. 2022; 22(1): 239. doi:10.1186/S12935-022-02663-2

53. Ci Y, Zhang Y, Liu Y, et al. Myricetin suppresses breast cancer metastasis through down-regulating the activity of matrix metalloproteinase (MMP)-2/9. Phytotherapy Research. 2018; 32(7): 1373-1381. doi:10.1002/PTR.6071

54. Ko SY. Myricetin suppresses LPS-induced MMP expression in human gingival fibroblasts and inhibits osteoclastogenesis by downregulating NFATc1 in RANKL-induced RAW 264.7 cells. Arch Oral Biol. 2012; 57(12): 1623-1632. doi:10.1016/J.ARCHORALBIO.2012.06.012

55. Brooks AJ, Putoczki T. JAK-STAT Signalling Pathway in Cancer. Cancers (Basel). 2020; 12(7): 1971. doi:10.3390/CANCERS12071971

56. Seif F, Khoshmirsafa M, Aazami H, Mohsenzadegan M, Sedighi G, Bahar M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Communication and Signaling.  2017; 15(1):1-13. doi:10.1186/S12964-017-0177-Y

57. Kumamoto T, Fujii M, Hou DX. Myricetin directly targets JAK1 to inhibit cell transformation. Cancer Lett. 2009; 275(1): 17-26. doi:10.1016/J.CANLET.2008.09.027

58. Tuponchai P, Kukongviriyapan V, Prawan A, Kongpetch S, Senggunprai L. Myricetin ameliorates cytokine-induced migration and invasion of cholangiocarcinoma cells via suppression of STAT3 pathway. J Cancer Res Ther. 2019; 15(1): 157-163. doi:10.4103/JCRT.JCRT_287_17

59. Liao HH, Zhang N, Meng YY, et al. Myricetin Alleviates Pathological Cardiac Hypertrophy via TRAF6/TAK1/MAPK and Nrf2 Signaling Pathway. Oxid Med Cell Longev. 2019; 2019. doi:10.1155/2019/6304058

60. Qin S, Chen J, Tanigawa S, Hou DX. Microarray and pathway analysis highlight Nrf2/ARE-mediated expression profiling by polyphenolic myricetin. Mol Nutr Food Res. 2013; 57(3): 435-446. doi:10.1002/MNFR.201200563

61. Nunes C, Teixeira N, Serra D, Freitas V, Almeida L, Laranjinha J. Red wine polyphenol extract efficiently protects intestinal epithelial cells from inflammation via opposite modulation of JAK/STAT and Nrf2 pathways. Toxicol Res (Camb). 2015; 5(1): 53. doi:10.1039/C5TX00214A

62. D’Ambrosio M, Bigagli E, Cinci L, et al. Ethyl acetate extract from Cistus x incanus L. leaves enriched in myricetin and quercetin derivatives, inhibits inflammatory mediators and activates Nrf2/HO-1 pathway in LPS-stimulated RAW 264.7 macrophages. Zeitschrift fur Naturforschung - Section C Journal of Biosciences. 2021; 76(1-2): 79-86. doi:10.1515/ZNC-2020-0053

63. Wu Y, Zhou BP. TNF-α/NF-κB/Snail pathway in cancer cell migration and invasion. Br J Cancer. 2010; 102(4): 639. doi:10.1038/SJ.BJC.6605530

64. Zhou XL, Yang J, Qu XJ, Meng J, Miao RR, Cui SX. M10, a Myricetin-3-O-b-D-Lactose Sodium Salt, Prevents Ulcerative Colitis Through Inhibiting Necroptosis in Mice. Front Pharmacol. 2020;11. doi:10.3389/FPHAR.2020.557312

65. Sun J, Sun J, Zhou X. Protective functions of myricetin in LPS-induced cardiomyocytes H9c2 cells injury by regulation of MALAT1. Eur J Med Res. 2019; 24(1). doi:10.1186/S40001-019-0378-5

66. Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta Mol Cell Res. 2016; 1863(12): 2977-2992. doi:10.1016/J.BBAMCR.2016.09.012

67. Yoshida K, Miki Y. The cell death machinery governed by the p53 tumor suppressor in response to DNA damage. Cancer Sci. 2010; 101(4): 831-835. doi:10.1111/J.1349-7006.2010.01488.X

68. Song X, Tan L, Wang M, et al. Myricetin: A review of the most recent research. Biomedicine and Pharmacotherapy. 2021; 134. doi:10.1016/J.BIOPHA.2020.111017

69. Bhatt S, Manhas D, Kumar V, et al. Effect of Myricetin on CYP2C8 Inhibition to Assess the Likelihood of Drug Interaction Using In Silico, In Vitro, and In Vivo Approaches. ACS Omega. 2022; 7(15): 13260-13269. doi:10.1021/ACSOMEGA.2C00726/SUPPL_FILE/AO2C00726_SI_001.PDF

70. Munro AW, McLean KJ, Grant JL, Makris TM. Structure and function of the cytochrome P450 peroxygenase enzymes. Biochem Soc Trans. 2018; 46(1):183-196. doi:10.1042/BST20170218,

71. Huang Y, Zheng SL, Zhu HY, Xu ZS, Xu RA. Effects of aescin on cytochrome P450 enzymes in rats. J Ethnopharmacol. 2014; 151(1): 583-590. doi:10.1016/J.JEP.2013.11.016

72. Jarrar Y, Lee SJ. The Functionality of UDP-Glucuronosyltransferase Genetic Variants and their Association with Drug Responses and Human Diseases. J Pers Med. 2021; 11(6): 554. doi:10.3390/JPM11060554

73. Li X, Wang C, Chen J, et al. Potential interactions among myricetin and dietary flavonols through the inhibition of human UDP-glucuronosyltransferase in vitro. Toxicol Lett. 2022; 358: 40-47. doi: 10.1016/J.TOXLET.2022.01.007

74. He Y, Sun MM, Zhang GG, et al. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduction and Targeted Therapy. 2021; 6(1):1-17. doi:10.1038/s41392-021-00828-5

75. Fresno Vara JÁ, Casado E, de Castro J, Cejas P, Belda-Iniesta C, González-Barón M. P13K/Akt signalling pathway and cancer. Cancer Treat Rev. 2004; 30(2): 193-204. doi: 10.1016/j.ctrv.2003.07.007

76. Peng Y, Wang Y, Zhou C, Mei W, Zeng C. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? Front Oncol. 2022; 12: 819128. doi:10.3389/FONC.2022.819128/XML/NLM

77. Porta C, Paglino C, Mosca A. Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 2014; 4 APR: 67531. doi:10.3389/FONC.2014.00064/BIBTEX

78. Pópulo H, Lopes JM, Soares P. The mTOR Signalling Pathway in Human Cancer. Int J Mol Sci. 2012; 13(2): 1886. doi:10.3390/IJMS13021886

79. Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell. 2017; 168(6): 960. doi: 10.1016/J.CELL.2017.02.004

80. Song X, Tan L, Wang M, et al. Myricetin: A review of the most recent research. Biomedicine and Pharmacotherapy. 2021; 134: 111017. doi: 10.1016/J.BIOPHA.2020.111017

81. Schwartz SL, Ratner RE, Kim DD, et al. Effect of exenatide on 24-hour blood glucose profile compared with placebo in patients with type 2 diabetes: A randomized, double-blind, two-arm, parallel-group, placebo-controlled, 2-week study. Clin Ther. 2008; 30(5): 858-867. doi: 10.1016/J.CLINTHERA.2008.05.004

82. Knickle A, Fernando W, Greenshields AL, Rupasinghe HPV, Hoskin DW. Myricetin-induced apoptosis of triple-negative breast cancer cells is mediated by the iron-dependent generation of reactive oxygen species from hydrogen peroxide. Food and Chemical Toxicology. 2018; 118: 154-167. doi:10.1016/J.FCT.2018.05.005

83. Devi KP, Rajavel T, Habtemariam S, Nabavi SF, Nabavi SM. Molecular mechanisms underlying anticancer effects of myricetin. Life Sci. 2015; 142: 19-25. doi:10.1016/j.lfs.2015.10.004

84. Tang NP, Zhou B, Wang B, Yu R Bin, Ma J. Flavonoids intake and risk of lung cancer: A meta-analysis. Jpn J Clin Oncol. 2009; 39(6): 352-359. doi:10.1093/JJCO/HYP028

85. Xia W, Zheng B, Li T, Lian F, Lin Y, Liu R. Fabrication, characterization and evaluation of myricetin adsorption onto starch nanoparticles. Carbohydr Polym. 2020; 250. doi: 10.1016/j.carbpol.2020.116848

86. Li J, Xiang H, Huang C, Lu J. Pharmacological Actions of Myricetin in the Nervous System: A Comprehensive Review of Preclinical Studies in Animals and Cell Models. Front Pharmacol. 2021; 12. doi:10.3389/FPHAR.2021.797298

87. Ye C, Zhang C, Huang H, et al. The Natural Compound Myricetin Effectively Represses the Malignant Progression of Prostate Cancer by Inhibiting PIM1 and Disrupting the PIM1/CXCR4 Interaction. Cellular Physiology and Biochemistry. 2018; 48(3): 1230-1244. doi:10.1159/000492009,

88. Kulchenko NG, Kostin AA, Chibisov SM, et al. Modern Principles of Early Diagnosis of Prostate Cancer. Res J Pharm Technol. 2017; 10(3): 696-698. doi:10.5958/0974-360X.2017.00130.5

89. P. P, V.C. Y, Singh KK, Kala KS, L. AK, P.K. S. The Cytological Diagnosis of Prostate Cancer. Research Journal of Pharmacology and Pharmacodynamics. 2012;4(4). Accessed June 28, 2025. https://rjppd.org/AbstractView.aspx?PID=2012-4-4-25

90. Yang W, Su J, Li M, et al. Myricetin Induces Autophagy and Cell Cycle Arrest of HCC by Inhibiting MARCH1-Regulated Stat3 and p38 MAPK Signaling Pathways. Front Pharmacol. 2021; 12. doi:10.3389/FPHAR.2021.709526

91. Li M, Chen J, Yu X, et al. Myricetin suppresses the propagation of hepatocellular carcinoma via down-regulating expression of YAP. Cells. 2019; 8(4). doi:10.3390/CELLS8040358

92. Zang W, Wang T, Wang Y, et al. Myricetin exerts anti-proliferative, anti-invasive, and pro-apoptotic effects on esophageal carcinoma EC9706 and KYSE30 cells via RSK2. Tumor Biology. 2014; 35(12): 12583-12592. doi:10.1007/S13277-014-2579-4

93. Sun F, Zheng XY, Ye J, Wu TT, Wang JL, Chen W. Potential anticancer activity of myricetin in human T24 bladder cancer cells both in vitro and in vivo. Nutr Cancer. 2012; 64(4): 599-606. doi:10.1080/01635581.2012.665564

94. Sajedi N, Homayoun M, Mohammadi F, Soleimani M. Myricetin Exerts its Apoptotic Effects on MCF-7 Breast Cancer Cells through Evoking the BRCA1-GADD45 Pathway. Asian Pacific Journal of Cancer Prevention. 2020; 21(12): 3461-3468. doi:10.31557/APJCP.2020.21.12.3461

95. Karale PA, Karale MA, Utikar MC. Advanced Molecular Targeted Therapy in Breast Cancer. Research Journal of Pharmacology and Pharmacodynamics. 2018; 10(1): 29-37. doi:10.5958/2321-5836.2018. 00006.X

96. Bhagya SS, Shanmuga RP, Ezhilarasi R. Role of Nursing in Lymphoedema with Breast Cancer Patients. Asian Journal of Nursing Education and Research. 2015; 5(4): 542. doi:10.5958/2349-2996.2015.00111.1

97. Srivastava S, Kumar A. Breast Cancer Survivorship among Indian Women: An Overview. Asian Journal of Nursing Education and Research. Published online August 1, 2022: 262-266. doi:10.52711/2349-2996.2022.00056

98. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68(6): 394-424. doi:10.3322/CAAC.21492

99. Li Q, Tan Q, Ma Y, Gu Z, Chen S. Myricetin Suppresses Ovarian Cancer In Vitro by Activating the p38/Sapla Signaling Pathway and Suppressing Intracellular Oxidative Stress. Front Oncol. 2022; 12. doi:10.3389/FONC.2022.903394

100. Tippegowda SB. A Case Study on Ovarian Cancer-Palliative Perspective. Asian Journal of Nursing Education and Research. 2015; 5(3): 446. doi:10.5958/2349-2996.2015.00090.7

101. Zheng AW, Chen YQ, Zhao LQ, Feng JG. Myricetin induces apoptosis and enhances chemosensitivity in ovarian cancer cells. Oncol Lett. 2017; 13(6): 4974. doi:10.3892/OL.2017.6031

102. Han SH, Lee JH, Woo JS, et al. Myricetin induces apoptosis and autophagy in human gastric cancer cells through inhibition of the PI3K/Akt/mTOR pathway. Heliyon. 2022; 8(5). doi: 10.1016/J.HELIYON. 2022.E09309

 

 

 

 

Received on 04.05.2025      Revised on 28.06.2025

Accepted on 07.08.2025      Published on 11.10.2025

Available online from October 25, 2025

Res.J. Pharmacology and Pharmacodynamics.2025;17(4):275-285.

DOI: 10.52711/2321-5836.2025.00044

©A and V Publications All right reserved

 

This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License.