Next Article in Journal
Recent Development of Organic Afterglow Probes for Diagnosis and Treatment of Cancer
Previous Article in Journal
Targeting CD3-CD16+CD56+ NK Cells and NK Cell Activity by Intralipid in the Management of Reproductive Failure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Resveratrol in Cancer Management: From Monotherapy to Combination Regimens

1
OncoPharma Research Group, Center of Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
2
CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
3
Intitute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
4
Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
*
Author to whom correspondence should be addressed.
Targets 2024, 2(4), 307-326; https://doi.org/10.3390/targets2040018
Submission received: 30 August 2024 / Revised: 9 October 2024 / Accepted: 15 October 2024 / Published: 16 October 2024

Abstract

:
Resveratrol, a naturally occurring polyphenol found in grapes, berries, and peanuts, has garnered significant attention for its potential anti-cancer properties. This review provides a comprehensive analysis of its role in cancer therapy, both as a standalone treatment and in combination with other therapeutic approaches. This review explores the molecular mechanisms underlying resveratrol’s anti-cancer effects, including its antioxidant activity, modulation of cellular signaling pathways, antiproliferative properties, anti-inflammatory effects, and epigenetic influences. This review also examines in vitro and in vivo studies that highlight resveratrol’s efficacy against various cancer types. Furthermore, the synergistic effects of resveratrol when used in conjunction with conventional treatments like chemotherapy and radiotherapy, as well as targeted therapies and immunotherapies, are discussed. Despite promising preclinical results, this review addresses the challenges and limitations faced in translating these findings into clinical practice, including issues of bioavailability and toxicity. Finally, it outlines future research directions and the potential for resveratrol to enhance existing cancer treatment regimens. This review aims to provide a thorough understanding of resveratrol’s therapeutic potential and to identify areas for further investigation in the quest for effective cancer treatments.

1. Introduction

Cancer encompasses a diverse group of diseases marked by uncontrolled cellular growth and the potential to invade other parts of the body, leading to severe health consequences and high mortality rates [1]. With over 100 different types, including breast, skin, lung, colon, and prostate cancer and lymphoma, this disease affects millions of people worldwide, posing one of the most significant challenges in modern medicine [2]. The complexity of cancer, coupled with the limitations of conventional treatments such as surgery, radiation, and chemotherapy, underscores the urgent need for the development of novel therapies [3,4]. These traditional treatments often cause significant side effects and may not be suitable for all cancer types or patients [5]. Additionally, treatment resistance and tumor heterogeneity further complicate disease management [6,7].
Given these challenges, the search for novel therapeutic agents has become imperative. One such promising agent is resveratrol, a naturally occurring polyphenolic compound found in various plants, including grapes and berries [8]. Resveratrol gained widespread attention due to its presence in red wine and its potential health benefits, particularly its connection to the ‘French Paradox’, a phenomenon where the French exhibit a low incidence of heart disease despite a diet rich in saturated fats [9,10]. Chemically, resveratrol, known as 3,5,4’-trihydroxystilbene, is classified as a stilbenoid, characterized by two phenol rings connected by a styrene double bond (Figure 1) [11].
Biologically, resveratrol exhibits a range of properties that contribute to its therapeutic potential, including antioxidant [12], anti-inflammatory [13], and anti-carcinogenic effects [14]. Due to its ability to modulate various molecular pathways, resveratrol is a promising candidate for preventing and treating a range of diseases, such as cardiovascular diseases [15], cancer [16], and neurodegenerative disorders [17,18]. Ongoing research continues to uncover the mechanisms behind resveratrol’s actions, making its potential applications in medicine and health promotion an exciting and active area of scientific investigation.
This review seeks to provide a comprehensive analysis of resveratrol’s mechanisms of action, its potential as a monotherapy in cancer treatment, and its synergistic effects when combined with other cancer therapies.

2. Mechanisms of Action of Resveratrol

2.1. Antioxidant Activity

Resveratrol is well known for its potent antioxidant activity, which is one of its most extensively studied biological properties, particularly in the context of cancer. As an antioxidant, it functions by neutralizing harmful free radicals (e.g., reactive oxygen species (ROS) and reactive nitrogen species (RNS))—unstable molecules that can cause cellular damage through oxidative stress [19]. Oxidative stress significantly contributes to cancer initiation and progression by damaging essential cellular components such as DNA, proteins, and lipids, thus facilitating tumor development [20,21,22]. In addition to scavenging ROS and RNS to prevent oxidative damage, resveratrol also chelates metal ions, such as iron and copper, which are known to catalyze the production of free radicals, reducing their availability for oxidative reactions [23,24]. Resveratrol also activates various signaling pathways that upregulate the expression of endogenous antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase, all of which play crucial roles in the detoxification of ROS [12]. Furthermore, resveratrol inhibits pro-oxidant enzymes such as NADPH oxidase and xanthine oxidase, which are responsible for producing ROS, thereby decreasing the overall oxidative stress burden in the tumor microenvironment [25]. Moreover, since oxidative stress is a significant factor in cancer initiation and progression, resveratrol’s ability to mitigate oxidative damage and modulate cell signaling pathways involved in cell growth and apoptosis makes it a promising candidate for cancer prevention and therapy [26].
Additionally, resveratrol’s antioxidant properties have broader implications for other diseases related to oxidative stress. In cardiovascular diseases, for instance, resveratrol helps to prevent the oxidation of low-density lipoprotein (LDL) cholesterol [15], a key step in the development of atherosclerosis, contributing to its cardioprotective effects [27]. Similarly, its ability to protect neurons from oxidative damage shows promise in neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease [28].
In addition to its potent antioxidant properties, resveratrol also exerts significant effects through the modulation of cellular signaling pathways.

2.2. Modulation of Cellular Signaling

Resveratrol’s modulation of various cellular signaling pathways underpins its broad spectrum of biological activities. This modulation is crucial to its therapeutic potential, especially in cancer prevention and treatment. Resveratrol influences multiple signaling cascades that regulate cell proliferation, apoptosis, inflammation, and oxidative stress, thereby affecting key pathways involved in cancer cell growth, survival, and metastasis.

NAD-Dependent Deacetylase Sirtuin-1 (SIRT1)

One of the primary mechanisms through which resveratrol exerts its effects is by activating the sirtuin family of proteins, particularly SIRT1 [29]. SIRT1 is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase that plays a crucial role in regulating cellular metabolism, aging, and inflammation [30,31,32]. The activation of SIRT1 by resveratrol results in the deacetylation and activation of various transcription factors and co-regulators, leading to a wide range of beneficial cellular effects. When SIRT1 is activated by resveratrol, it deacetylates the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) [33]. Deacetylation of PGC-1α enhances its activity, leading to an increase in the number and function of mitochondria [34], which are essential for energy production in cells. This upregulation of mitochondrial function enhances cellular energy metabolism and increases resistance to oxidative stress [35].
In addition to PGC-1α, SIRT1 activation also influences other important transcription factors and proteins. For instance, SIRT1 deacetylates and modulates the activity of the tumor suppressor protein p53 [36,37], which is involved in cell cycle regulation [38] and apoptosis [39]. Through the regulation of p53, resveratrol promotes cell survival under stress conditions while inducing apoptosis in damaged or tumor cells. Additionally, SIRT1 activation by resveratrol can inhibit the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway [40]. NF-κB is a transcription factor that regulates genes involved in inflammation, immune response, and cell survival [41]. By inhibiting NF-κB, SIRT1 reduces inflammation and may help prevent chronic inflammatory diseases and cancer progression.
Resveratrol also significantly affects the PI3K/Akt signaling pathway, a crucial regulator of cell growth, survival, and metabolism [42]. By inhibiting PI3K activity, resveratrol reduces the phosphorylation and activation of Akt, leading to decreased cell proliferation and enhanced apoptosis [43]. This modulation of the PI3K/Akt pathway not only hampers tumor growth and promotes cancer cell death but also contributes to resveratrol’s anti-inflammatory and metabolic regulatory effects.

2.3. Antiproliferative Effects

By inhibiting the cell cycle and inducing apoptosis, resveratrol exerts significant antiproliferative effects on cancer cells. Research shows that it induces cell cycle arrest at various stages, particularly the G1 and G2/M phases [44,45,46]. This arrest is achieved through the modulation of cyclins and cyclin-dependent kinases (CDKs), which are essential for the progression of the cell cycle [47]. By altering the expression and activity of these proteins, resveratrol halts the uncontrolled growth of cancer cells. In addition, it promotes apoptosis (programmed cell death) in cancer cells [48,49]. It activates intrinsic apoptotic pathways by upregulating pro-apoptotic factors like Bax and downregulating anti-apoptotic proteins such as Bcl-2 [50]. This shift in the balance between pro- and anti-apoptotic signals leads to mitochondrial dysfunction and the release of cytochrome c, which activates caspases and triggers cell death [51].

2.4. Anti-Inflammatory Effects

Resveratrol exerts significant anti-inflammatory effects through several key mechanisms, which contribute to its therapeutic potential in treating chronic inflammatory conditions and related diseases. Resveratrol modulates inflammation by inhibiting the NF-κB signaling pathway. NF-κB is a crucial transcription factor that regulates the expression of various pro-inflammatory cytokines, adhesion molecules, and enzymes involved in inflammation [41]. By inhibiting NF-κB activation, it effectively reduces the production of inflammatory mediators such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and cyclooxygenase-2 (COX-2), thereby mitigating the inflammatory response [52]. In addition, resveratrol directly inhibits COX-2 activity, which is critical for the synthesis of prostaglandins involved in mediating inflammation and pain [53]. Since chronic inflammation is a well-established risk factor for cancer, its anti-inflammatory effects may significantly contribute to its anticancer properties.

2.5. Epigenetic Modulation

Resveratrol significantly influences gene expression through epigenetic modulation, a process that alters gene expression without changing the DNA sequence [54]. This capability is central to its therapeutic potential across various diseases, including cancer. One of the main ways it affects epigenetic regulation is by modifying histone acetylation. Resveratrol acts as a potent activator of sirtuins, particularly SIRT1. SIRT1 deacetylates histones, leading to changes in chromatin structure and, consequently, altering gene expression [55].
Resveratrol also influences DNA methylation patterns, another crucial aspect of epigenetic control. DNA methylation involves adding methyl groups to cytosine residues within CG dinucleotides, typically leading to gene silencing [56]. Resveratrol can affect the activity of DNA methyltransferases, the enzymes responsible for DNA methylation, thereby modifying the expression of genes involved in cell growth, apoptosis and inflammation [57].
Additionally, it impacts the expression of microRNAs (miRNAs), which are small non-coding RNAs that regulate gene expression after transcription [58,59]. By altering miRNA expression, it can affect numerous cellular processes, including cell cycle regulation, apoptosis and responses to cellular stress. These changes can lead to the suppression of oncogenes and the activation of tumor suppressor genes, further enhancing resveratrol’s potential as an anti-cancer agent [60].
The following table (Table 1) summarizes the key mechanisms through which resveratrol exerts its anticancer effects. This overview highlights the primary biological activities of resveratrol, detailing the associated pathways, descriptions of their functions, and implications for cancer prevention and therapy.

3. Application of Resveratrol in Cancer Treatment

3.1. In Vitro Studies

In vitro studies have provided substantial insights into the application of resveratrol in cancer treatment, demonstrating its potential as an effective anti-cancer agent. These studies have shown that it exerts multiple anti-cancer effects on various cancer cell lines, including those of breast, prostate, colon, lung, and liver cancers.

3.1.1. Lung Cancer

In studies focusing on non-small lung cancer (NSCLC) cell lines such as A549 and H460, resveratrol has demonstrated potential as a treatment option. Resveratrol treatment suppressed cell growth in these NSCLC cell lines, indicating its ability to inhibit proliferation. Additionally, it increased the expression of microtubule-associated protein 1 light chain 3 (LC3), which plays a crucial role in autophagy—a cellular process that degrades and recycles damaged or unnecessary cellular components [61]. The treatment of A549 cells with resveratrol has been found to induce apoptosis and cause G1 cell cycle arrest, halting cell cycle progression at the G1 phase. This effect is accompanied by the upregulation of p53 and p21 [62]. Ebi et al. [63] studied resveratrol’s effects on three human NSCLC cell lines: A427, NCI-H23, and A549. The treatment of these cancer cells resulted in the inhibition of the PI3K pathway, a critical signaling pathway regulating cell proliferation, growth, survival, metabolism, and protein synthesis. Additionally, treatment with resveratrol decreased mTOR phosphorylation, indicating reduced mTOR activity. Since mTOR is vital for regulating cell growth and protein synthesis, its inhibition by resveratrol can hinder cancer cell proliferation and survival [64]. In H460 cells, resveratrol decreased cell viability and proliferation while inducing significant apoptosis. This apoptosis was associated with increased hydrogen peroxide (H2O2) production, the activation of Bid, PARP, and caspase 8, and the downregulation of pEGFR, pAkt, c-FLIP, and NFkB protein expression [65]. Li et al. [66] used the small cell lung cancer (SCLC) cell line H446 to investigate the antitumor mechanism of resveratrol. Their results showed that resveratrol inhibited H446 cell viability, induced apoptosis, increased cytochrome c expression, inhibited the PI3K/Akt/c-Myc pathway, and promoted AIF translocation from the cytoplasm to the nucleus. This study therefore suggests that in SCLC H446 cells, resveratrol may reduce cell viability and induce apoptosis by affecting the PI3K/Akt/c-Myc pathway, with oxidative stress and mitochondrial membrane depolarization contributing to these effects.

3.1.2. Prostate Cancer

Resveratrol exhibits varying effects on growth, cell cycle arrest, and apoptosis induction in human prostate cancer cell lines such as PC-3 and C42B [67,68]. Substantial evidence indicates that its anti-prostate cancer effects are primarily mediated through its targeting of the androgen receptor (AR) and its regulated genes [69,70,71]. A study conducted by Farhan demonstrated that resveratrol reduces cell growth and induces apoptosis-like cell death in the PC-3 and C42B cell lines. These effects were significantly inhibited by copper chelators and ROS. This suggests that intracellular copper reacts with resveratrol to generate ROS, resulting in DNA damage [24].

3.1.3. Breast Cancer

Resveratrol has been shown to inhibit the proliferation of MCF-7 breast cancer cells in a dose- and time-dependent manner. Significant cytotoxic effects were observed even at a concentration of 100 μM after 24 h of treatment. Compared to untreated cells, it reduced the viability of MCF-7 cells to approximately 57.5%, yielding a half-maximal inhibitory concentration (IC50) of 51.18 μM. Additionally, treatment with resveratrol led to the induction of apoptosis in this cell line, as indicated by elevated levels of apoptosis markers [72]. In another study, resveratrol moderately inhibited MCF-7 cell viability. Specifically, it increased the number of early apoptotic cells in MCF-7 [49]. Kim et al. [73] demonstrated that resveratrol reduces the expression of YAP target genes and inhibits breast cancer cell invasion. Resveratrol accomplishes this by activating Lats1, resulting in the inactivation of YAP. Notably, it also inactivates RhoA, which enhances Lats1 activation and promotes YAP phosphorylation.

3.1.4. Colorectal Cancer

Resveratrol reduces the proliferation and invasion of colorectal cancer (CRC) cells by activating SIRT1, decreasing the NF-κB-mediated inflammatory pathway, and inhibiting the focal adhesion kinase (FAK) activity. This results in a reduction in focal adhesion molecules and an increase in apoptosis. Furthermore, it inhibits the invasion, colony formation, and cell growth of CRC cells, as well as the expression of β1-Integrin and the activation of FAK in an alginate cancer microenvironment [74]. It has also been shown to suppress the Wnt signaling pathway, which is crucial in the development of several serious diseases, including cancer [75]. Resveratrol reduces this pathway in a dose-dependent manner, reducing the expression of key target genes such as cyclin D1 and c-Myc, thereby inhibiting the growth of Wnt-induced and Wnt-driven colorectal cancer (CRC) cells [76]. Additionally, it inhibits the activity of several enzymes involved in DNA replication and cell proliferation [77]. These combined effects likely contribute to its ability to modulate cell proliferation.

3.1.5. Liver Cancer

Resveratrol has been shown to inhibit the proliferation of HepG2 hepatocellular carcinoma cells in a dose- and time-dependent manner. Significant cytotoxic effects were observed even at a concentration of 100 μM after 24 h of treatment. Compared to untreated cells, resveratrol reduced the viability of HepG2 cells by about 56.2%, with an IC50 of 57.4 μM [72]. Dai et al. [78] found that it induces apoptosis and inhibits the proliferation, migration, and invasion of the HepG2 and Hep3B hepatocellular carcinoma cell lines. Resveratrol reduced the expression of MARCH1 and phospho-protein kinase B (p-AKT), while increasing the expression of the tumor suppressor PTEN in a dose-dependent manner. Another study demonstrated that it significantly impedes the respiratory metabolism of hepatocellular carcinoma cells, thereby inhibiting their proliferation, migration and invasion, while also promoting apoptosis [79]. Resveratrol inhibited both anchorage-dependent and anchorage-independent growth of hepatocellular carcinoma cells in a dose-dependent manner. Short-term exposure to resveratrol significantly reduced the activation of the HGF-induced c-Met signaling pathway, while long-term exposure markedly decreased c-Met expression on the cell membrane. Additionally, it suppressed HGF-induced cell invasion [80].

3.1.6. Pancreatic Cancer

Ratajczak et al. [81] investigated the effects of resveratrol on three human pancreatic cancer cell lines, EPP85-181P, EPP85-181RNOV (mitoxantrone-resistant), and AsPC-1, as well as the normal pancreatic cell line H6c7. Their findings suggest that it exerts anticarcinogenic effects by inhibiting the proliferation of pancreatic cancer cells and modulating levels of pro- and anti-apoptotic Bcl-2 proteins. Notably, its activity was cell line-specific, with the most significant effects observed in the mitoxantrone-resistant cells. Another study showed that it reduced tumor growth, triggered apoptosis, and increased Bax expression in Capan-2 cells [82]. Qin and his team revealed that nutrient-deprivation autophagy factor-1 (NAF-1) is present in pancreatic cancer tissue and is associated with disease progression. Additionally, they discovered that inhibiting NAF-1 significantly reduces the stem cell properties and invasion and migration capabilities of pancreatic cancer cells [83]. Another study demonstrated that treatment with resveratrol (ranging from 0 to 200 µM) significantly increased the proportion of pancreatic cancer cells (ASPC-1, BXPC-3, Panc-1 cell lines) in the G0/G1 phase of the cell cycle compared to the control group [84,85]. Li et al. [86] showed that resveratrol, applied at concentrations ranging from 0 to 50 µM for 24 h, inhibited the phosphorylation of AKT and NF-κB in BxPC-3 and Panc-1 cells in a dose-dependent manner.

3.2. In Vivo Studies

In vivo studies are essential for comprehending the physiological and therapeutic effects of resveratrol within complex biological systems. These studies provide insights into how it interacts with various pathways and influences disease progression in living organisms, effectively bridging the gap between in vitro research and clinical applications. Multiple animal models, including murine and xenograft models, have been utilized to assess the anticancer effects of resveratrol. Table 2 highlights the diverse effects of resveratrol observed in vivo, such as its capacity to reduce oxidative stress, inhibit tumor growth, and improve cardiovascular health. These effects are attributed to its interaction with key signaling pathways and its ability to modulate gene expression and inflammatory responses.
In a subcutaneous xenograft model of pancreatic cancer in nude mice, resveratrol was administered at a dose of 50 mg/kg daily for six weeks. This treatment resulted in the suppression of NAF-1 expression, which correlated with a significant reduction in tumor growth. These findings suggest that it acts as an effective anti-tumor agent, potentially targeting NAF-1 as a therapeutic approach [83]. Another study investigated its impact and nanoformulation (NP-RSV) on tumor growth and vascularity in colon cancer models using both xenograft and orthotopic mice. Mice treated with resveratrol at 100 mg/kg and NP-RSV at an equivalent dose showed significant reductions in tumor growth and hemoglobin content within the tumor mass, indicating decreased vascularity and improved therapeutic efficacy of NP-RSV compared to resveratrol alone [110]. These studies collectively highlight its potential as an adjunctive therapy in cancer treatment. The observed anti-tumor effects, particularly when combined with nanoformulation techniques, underscore the importance of further exploring its mechanisms of action and its application in clinical settings. Future research should focus on optimizing dosage regimens, understanding long-term effects, and exploring synergistic combinations with other therapeutic agents.
A search for “resveratrol and cancer” on Clinicalrials.gov reveals only 18 studies, with some focusing primarily on resveratrol’s pharmacokinetics rather than its therapeutic effects (Table 3). This limited number of clinical trials raises concerns about the practicality of resveratrol as a cancer treatment.
While the clinical trials summarized in Table 2 indicate the potential of resveratrol as an adjunct therapy in cancer treatment, it is crucial to acknowledge certain limitations that may impact its clinical application. Despite the promising preclinical data, challenges such as small-scale trials and variability in study design complicate our understanding of its therapeutic potential. These factors highlight the need for larger, more comprehensive studies to fully explore its efficacy in conjunction with established therapeutic agents.

4. Resveratrol in Combination with Other Treatments

4.1. Chemotherapy

Resveratrol has attracted considerable interest as a potential adjunct in cancer therapy, especially when paired with conventional chemotherapy. This interest is driven by its ability to both enhance the effectiveness of chemotherapeutic agents and mitigate their common side effects, which can limit the broad application of these treatments in clinical settings.
A major concern in cancer treatment is cardiotoxicity, a common side effect of many chemotherapeutic agents [111]. Commonly used anthracyclines like doxorubicin often result in severe cardiotoxic effects, including myocarditis, arrhythmias, dilated cardiomyopathy, and congestive heart failure [112]. These adverse effects are linked to the production of free radicals and oxidative stress, as doxorubicin increases ROS formation in heart tissues [113,114]. A study by Sheu and his team examined the antioxidant effects of four different compounds, including resveratrol, and found that co-treatment with doxorubicin significantly reduced intracellular ROS levels while increasing SOD levels, which are involved in eliminating ROS. Among the antioxidants tested, resveratrol was the most effective at neutralizing ROS, suggesting its potential as a supplement to mitigate doxorubicin-induced cardiotoxicity [115]. Additionally, Ito et al. [116] found that resveratrol and its analogs can bind to carbonyl reductase 1 (CBR1), inhibiting its activity and thereby reducing doxorubicin’s cardiotoxicity. Arsenic trioxide (As2O3), another traditional anticancer drug primarily used for treating acute promyelocytic leukemia (APL), also causes adverse effects such as cardiotoxicity. Like anthracyclines, the primary pathogenic mechanism of As2O3 is associated with oxidative stress [117,118]. Zhao et al. [119] reported that pre-treatment with resveratrol (3 mg/kg) one hour before As2O3 administration could protect against As2O3-induced cardiotoxicity and reduce myocardial injury, oxidative damage, and DNA fragmentation.
In addition to cardiotoxicity, chemotherapy often leads to gastrointestinal side effects including anorexia, nausea, vomiting, constipation, and diarrhea [120]. Ko et al. [121] demonstrated that pretreating Wistar rats with whole grape juice, which contains high levels of resveratrol could promote gastric emptying and improve tubular dilation and vacuolization in renal tubules, thereby helping to prevent cisplatin-induced gastrointestinal disorders.
In addition to mitigating adverse effects, resveratrol shows promise in enhancing the efficacy of various chemotherapeutic agents. For example, it has shown synergistic effects with 5-fluorouracil (5-FU) and doxorubicin, enhancing their antitumor efficacy in colorectal [122] and oral [123] cancer types. Hu et al. [122] highlighted the therapeutic potential of resveratrol, particularly when combined with ginkgetin and 5-FU. This combination suppressed the expression of COX-2 and inflammatory cytokines, alleviating the inflammatory response induced by 5-FU. Additionally, ginkgetin and resveratrol inhibit VEGF-induced endothelial cell proliferation, migration, invasion, and angiogenesis. Another study by Kong et al. [124] found that combining 5 μg/mL or 10 μg/mL resveratrol with 5 μg/mL paclitaxel resulted in greater growth inhibition and apoptosis of the A549 NSCLC cell line compared to treatment with each agent alone. This combination treatment notably downregulated mRNA and protein levels of several key molecules associated with cancer progression, such as COX-2, VEGF, and various matrix metalloproteinases (MMPs), as well as anti-apoptotic proteins like Bcl-2 and Bcl-xL. Additionally, it reduced the levels of pro-inflammatory cytokines (TNF-α, IL-1β) and iNOS, while enhancing the expression of anti-cancer genes, including tissue inhibitors of metalloproteinases (TIMPs), IκB-α, p53, p21, and pro-apoptotic proteins. The synergy between resveratrol and cisplatin is particularly notable in reducing metastasis. This combination not only inhibits cell proliferation and invasion but also reduces telomerase activity and enhances ß-galactosidase activity. It induces cell cycle arrest at the G0/G1 phase, promotes apoptosis, and supports cell senescence through the P38/P53 and P16/P21 pathways [125]. Additionally, resveratrol and cisplatin together induce endoplasmic reticulum stress-mediated apoptosis and cause cell cycle arrest at the G2/M phase in gastric cancer cells [126]. Resveratrol has demonstrated significant potential as an adjunct therapy in osteosarcoma treatment, enhancing the effects of conventional chemotherapy regimens [127].
Overall, resveratrol has demonstrated substantial potential as an adjunct therapy in cancer treatment, particularly when combined with conventional chemotherapy regimens. Its ability to enhance the efficacy of various chemotherapeutic agents while mitigating their adverse effects highlights its promise as a valuable therapeutic agent in the fight against cancer.

4.2. Radiotherapy

Resveratrol shows promise as an adjunct treatment in cancer therapy when combined with radiotherapy. Radiotherapy is a standard cancer treatment that uses ionizing radiation to kill cancer cells [128]. However, it can also harm surrounding normal tissues, leading to adverse side effects. Resveratrol enhances the efficacy of radiotherapy while protecting normal cells from radiation-induced damage, making it an attractive candidate for combination therapy [129].
A key benefit of combining resveratrol with radiotherapy is its ability to reduce oxidative stress and protect normal cells from apoptosis [130]. This protective effect is mediated through the activation of the SIRT1/FOXO3a and PI3K/Akt pathways, which help in reducing radiation-induced intestinal damage [131]. Additionally, it has been found to increase the tumor cell sensitivity to radiation and increase apoptosis. Its radio-sensitizing effects are attributed to its ability to disrupt DNA repair mechanisms early on, inhibit cell proliferation, induce autophagy and promote apoptosis in both laboratory and animal models [132].
In addition to its radioprotective properties, numerous studies have demonstrated resveratrol’s synergistic effects when combined with ionizing radiation [133,134,135]. Although the radiosensitizing effects of resveratrol are well documented in vitro, only a few in vivo studies have confirmed its ability to enhance the effectiveness of radiotherapy. One study identified that its radiosensitizing mechanism involves increased autophagy and apoptosis [132]. Another study by Mikami et al. [136] demonstrated that it enhances irradiation efficacy via the expression pathway of the regenerating gene (REG) III. Furthermore, a study investigating the effects of radiotherapy combined with resveratrol on radioresistant glioma stem cells (GSCs) found that it inhibited GSC proliferation and enhanced radiosensitivity. This combination treatment decreased the expression of the neural stem cell marker CD133, induced cell differentiation, and significantly increased levels of autophagy and apoptosis both in vitro and in vivo [132]. Additional research suggests that resveratrol, when used in combination with ionizing radiation, delays the repair of radiation-induced DNA double-strand breaks (DSBs) and prolongs G2/M cell cycle arrest, leading to increased apoptosis [137].

4.3. Target Therapies and Immunotherapy

Resveratrol’s ability to modulate signaling pathways and immune responses suggests potential benefits when used alongside targeted therapies and immunotherapies.
Targeted therapies are designed to interfere with specific molecules involved in cancer growth and progression [138]. These therapies aim to selectively target cancer cells while sparing normal cells, offering a more precise approach compared to traditional chemotherapy [139]. Resveratrol has been studied for its ability to enhance the effects of targeted therapies through some mechanisms. One significant mechanism is the inhibition of key signaling pathways, such as the PI3K/Akt/mTOR pathway, a critical signaling cascade that is frequently overactive in various cancers, contributing to tumor growth and survival [140]. By downregulating this pathway, resveratrol can promote apoptosis and inhibit cancer cell proliferation, as observed in studies involving glioma cells, where it not only reduces Akt activation but also decreases the phosphorylation of downstream targets like S6 kinase [43]. Studies have shown that it can potentiate the effects of tyrosine kinase inhibitors, leading to increased cancer cell apoptosis and reduced proliferation [141]. Moreover, it has been shown to modulate the expression of key proteins involved in cell survival and apoptosis, such as Bcl-xL and Bax. For example, resveratrol enhances the effectiveness of trastuzumab through synergistic interactions that notably reduce Bcl-xL gene expression and increase apoptosis in breast cancer cells [142]. Another significant challenge in cancer treatment is overcoming drug resistance. Resveratrol modulates molecular pathways that help to counteract multidrug resistance (MDR), including the downregulation of proteins such as P-glycoprotein (P-gp) [143] and multidrug-resistance-associated proteins (MRPs) [144], often overexpressed in resistant cancer cells. By modulating these pathways, it enhances the sensitivity of cancer cells to targeted therapies, making it a valuable adjunct in treatment regimens [145].
Immunotherapy has revolutionized cancer treatment by harnessing the immune system to identify and eradicate cancer cells [146]. However, its efficacy may be compromised by the immunosuppressive tumor microenvironment and the ability of cancer cells to evade immune detection [147]. Resveratrol’s diverse biological activities may enhance immunotherapy’s effectiveness in several ways. Firstly, it modulates the immune response by enhancing the activity of various immune cells, including T cells, natural killer (NK) cells, and dendritic cells [148]. By boosting the function of these immune cells, it may augment the effectiveness of immunotherapies, such as checkpoint inhibitors, which depend on an active immune response to target cancer cells [149]. Secondly, chronic inflammation within the tumor microenvironment can suppress the immune response against cancer’s [150] anti-inflammatory properties, mediated through the inhibition of NF-κB signaling and the reduction in pro-inflammatory cytokines, creating a more favorable environment for immunotherapy to operate [13,40]. By mitigating inflammation, it enhances the infiltration and activity of immune cells in tumors. Additionally, checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 pathways have shown substantial success in treating specific cancers, but not all patients respond favorably [149,151,152,153]. Resveratrol has been shown to downregulate the expression of PD-L1 in cancer cells, which can potentiate the efficacy of PD-1/PD-L1 inhibitors [154,155]. This synergistic effect could improve the response rates and outcomes for patients undergoing checkpoint inhibitor therapy. Moreover, inducing immunogenic cell death is an objective of cancer therapy, where dying cancer cells release signals that attract and activate immune cells [156]. Resveratrol has been shown to induce such immunogenic cell death, enhancing the immune system’s ability to recognize and attack cancer cells [157]. This property positions resveratrol as a promising candidate for combination with immunotherapy. Lastly, some tumors develop resistance to immunotherapy by creating an immunosuppressive microenvironment or upregulating immune checkpoints [158]. Resveratrol’s ability to modulate multiple pathways involved in immune evasion and suppression may help to overcome these resistance mechanisms, enhancing the efficacy of immunotherapy [159,160,161].

5. Adverse Effects and Safety Considerations

Although resveratrol is widely recognized for its therapeutic potential and general safety, its use at higher therapeutic doses may be associated with adverse effects and safety concerns [162]. Commonly reported side effects include gastrointestinal disturbances such as nausea, diarrhea, and abdominal pain, which are often dose-dependent and may become more pronounced at elevated doses [163]. Resveratrol has also been linked to hepatotoxicity, with some studies suggesting that high doses may adversely affect liver function [162]. While such effects are relatively rare, monitoring liver health is advisable, especially for individuals with pre-existing liver conditions or those on other medications that affect liver function.
Drug interactions are another area of concern; resveratrol may influence the metabolism of medications processed by the liver’s cytochrome P450 enzymes, potentially altering the efficacy and safety of concomitant therapies [164]. This is especially pertinent for patients on anticoagulants or antiplatelet medications, as resveratrol may enhance the risk of bleeding [165,166].
Particular caution is advised for patients with hormone-responsive cancers, such as breast cancer, due to resveratrol’s influence on hormone signaling pathways. Resveratrol may exhibit estrogenic effects, which could potentially affect hormone-driven tumor growth [167,168].
To ensure safe and effective use, comprehensive safety evaluations are essential, necessitating thorough clinical trials to establish appropriate dosing regimens and identify potential adverse effects [169]. Optimal dosing should balance therapeutic benefits with minimal side effects, and regular monitoring of liver function and gastrointestinal health should be included in the management strategy. Patients should consult their healthcare providers before initiating resveratrol supplementation, especially if they are on other medications or have underlying health conditions.

6. Limitations of Current Studies

Although preclinical data highlight resveratrol’s potential as a cancer therapeutic, several limitations must be addressed before its clinical application can be fully realized. One of the major challenges is its poor bioavailability [170]. Resveratrol is rapidly metabolized and eliminated from the body, resulting in low and fluctuating plasma levels that are often insufficient to achieve the therapeutic concentrations needed for effective cancer treatment [171]. This challenge complicates the ability to maintain adequate drug levels in target tissues.
Additionally, variability in the study results adds further complexity. While some preclinical studies show significant anticancer effects and enhanced apoptosis in various cancer cell lines, others report inconsistent or modest results. Differences in study design, dosage, formulation, and methodologies contribute to this variability, making it difficult to generalize findings across different models and cancer types.
The clinical trial landscape also reveals limitations regarding the use of resveratrol as a cancer therapy. Currently, most clinical trials are small-scale and often focus on resveratrol’s bioavailability and pharmacokinetics rather than its direct anti-cancer effects. This disparity between preclinical success and clinical application highlights the urgent need for larger, more comprehensive trials to fully elucidate the therapeutic potential of resveratrol in cancer treatment.
In addition, formulation and delivery issues present a significant challenge. Resveratrol’s low bioavailability requires the development of advanced delivery systems to enhance its stability and effectiveness. Nanoparticle-based and liposomal formulations are being explored, but their success and safety require thorough validation.
Furthermore, a detailed understanding of the molecular mechanisms through which resveratrol acts is still evolving. This knowledge is crucial for optimizing dosing strategies and predicting patient responses. Without clear insights into its mechanisms of action, determining the most effective treatment regimens and identifying appropriate patient populations remains challenging.
Lastly, while resveratrol is generally considered safe, its potential side effects and interactions with other medications must be carefully evaluated. Managing these factors is essential to ensuring that resveratrol can be used safely and effectively in cancer treatment.

7. Future Perspectives

The exploration of resveratrol’s role in cancer therapy offers numerous promising research avenues. As studies continue to assess its potential to enhance the efficacy of traditional chemotherapeutic agents and mitigate their adverse effects, it becomes increasingly important to deepen our understanding of its mechanisms and optimize its application. Future research should not only focus on elucidating the precise pathways through which resveratrol improves treatment outcomes and reduces side effects, but also emphasize the need to first understand these mechanisms individually before exploring their combined effects with other therapies. This includes a detailed examination of its effects on various signaling pathways, gene expression, and cellular processes involved in cancer progression and drug resistance.
Optimizing the dosage and administration of resveratrol is crucial to harness its full therapeutic potential. Investigating the most effective dosages and delivery methods will help to maximize the benefits of resveratrol while minimizing potential toxicity. Additionally, further research into combining resveratrol with other emerging therapies should be carefully designed to reveal potential synergistic effects while also considering the complexities of how these therapies may interact on a molecular level. Exploring its interactions with targeted therapies, immunotherapies, and novel small molecules could provide valuable insights into developing more effective combination treatments. However, it is essential to first elucidate resveratrol’s isolated mechanisms of action before fully understanding its role in combination therapies.
Expanding the scope of research to include a wider range of cancer types will be essential. While current studies have focused on specific cancers, there is significant potential to investigate resveratrol’s effects across other malignancies, including those resistant to conventional treatments. This broader exploration could uncover new therapeutic applications and improve outcomes for a wider range of cancer patients.
To transition from preclinical findings to clinical practice, well-designed clinical trials are necessary. These trials should be meticulously planned to evaluate the safety, efficacy, and optimal dosing of resveratrol in combination with standard chemotherapies. Randomized, controlled trials will provide robust data on treatment outcomes and help identify which patient populations are most likely to benefit. Identifying biomarkers that predict patient response to resveratrol-based therapies will also be important for personalizing treatment and improving patient outcomes.
Long-term follow-up in clinical trials will be crucial to assess the durability of therapeutic responses, monitor long-term side effects, and evaluate the overall impact on patients’ quality of life. Navigating regulatory pathways to facilitate the approval of resveratrol-based therapies will require addressing safety concerns, standardizing formulations, and ensuring compliance with clinical trial regulations.
In conclusion, while the current research on resveratrol is promising, advancing this work involves exploring new research directions, developing innovative combined treatments, and conducting rigorous clinical trials. By addressing these areas, the potential for resveratrol to significantly impact cancer therapy can be realized, ultimately improving treatment outcomes and enhancing patient quality of life.

8. Conclusions

Resveratrol, a naturally occurring polyphenolic compound, has demonstrated significant potential in cancer management through its multifaceted mechanisms of action. Its antioxidant, anti-inflammatory, antiproliferative and epigenetic modulation properties provide a strong foundation for its application in cancer therapy. In vitro and in vivo studies have consistently shown its efficacy in inhibiting cancer cell growth, inducing apoptosis and enhancing the effects of conventional treatments such as chemotherapy and radiotherapy. However, despite these promising findings, the clinical translation of resveratrol is challenged by issues related to its bioavailability and potential toxicity. Future research should focus on optimizing delivery methods, understanding the long-term effects, and conducting comprehensive clinical trials to fully realize its therapeutic potential in cancer treatment.

Author Contributions

Conceptualization, N.V.; methodology E.R.; formal analysis, E.R. and N.V.; investigation, E.R.; writing—original draft preparation, E.R.; writing—review and editing, N.V.; supervision, N.V.; project administration, N.V.; funding acquisition, N.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financed by Fundo Europeu de Desenvolvimento Regional (FEDER) funds through the COMPETE 2020 Operational Program for Competitiveness and Internationalization (POCI), Portugal 2020, and by Portuguese funds through Fundação para a Ciência e a Tecnologia (FCT) in the framework of the projects IF/00092/2014/CP1255/CT0004 and CHAIR in Onco-Innovation from the Faculty of Medicine, University of Porto (FMUP).

Acknowledgments

E.R. acknowledges CHAIR in Onco-Innovation/FMUP for funding her Ph.D. grant.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brown, J.S.; Amend, S.R.; Austin, R.H.; Gatenby, R.A.; Hammarlund, E.U.; Pienta, K.J. Updating the Definition of Cancer. Mol. Cancer Res. 2023, 21, 1142–1147. [Google Scholar] [CrossRef] [PubMed]
  2. Ma, X.; Yu, H. Global burden of cancer. Yale J. Biol. Med. 2006, 79, 85–94. [Google Scholar] [PubMed]
  3. Debela, D.T.; Muzazu, S.G.; Heraro, K.D.; Ndalama, M.T.; Mesele, B.W.; Haile, D.C.; Kitui, S.K.; Manyazewal, T. New approaches and procedures for cancer treatment: Current perspectives. SAGE Open Med. 2021, 9, 20503121211034366. [Google Scholar] [CrossRef]
  4. Mitra, A.K.; Agrahari, V.; Mandal, A.; Cholkar, K.; Natarajan, C.; Shah, S.; Joseph, M.; Trinh, H.M.; Vaishya, R.; Yang, X.; et al. Novel delivery approaches for cancer therapeutics. J. Control. Release 2015, 219, 248–268. [Google Scholar] [CrossRef]
  5. Chakraborty, S.; Rahman, T. The difficulties in cancer treatment. Ecancermedicalscience 2012, 6, ed16. [Google Scholar] [CrossRef]
  6. Zhu, L.; Jiang, M.; Wang, H.; Sun, H.; Zhu, J.; Zhao, W.; Fang, Q.; Yu, J.; Chen, P.; Wu, S.; et al. A narrative review of tumor heterogeneity and challenges to tumor drug therapy. Ann. Transl. Med. 2021, 9, 1351. [Google Scholar] [CrossRef]
  7. Lim, Z.-F.; Ma, P.C. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J. Hematol. Oncol. 2019, 12, 134. [Google Scholar] [CrossRef]
  8. Koushki, M.; Amiri-Dashatan, N.; Ahmadi, N.; Abbaszadeh, H.A.; Rezaei-Tavirani, M. Resveratrol: A miraculous natural compound for diseases treatment. Food Sci. Nutr. 2018, 6, 2473–2490. [Google Scholar] [CrossRef]
  9. Catalgol, B.; Batirel, S.; Taga, Y.; Ozer, N.K. Resveratrol: French paradox revisited. Front. Pharmacol. 2012, 3, 141. [Google Scholar] [CrossRef]
  10. Vidavalur, R.; Otani, H.; Singal, P.K.; Maulik, N. Significance of wine and resveratrol in cardiovascular disease: French paradox revisited. Exp. Clin. Cardiol. 2006, 11, 217–225. [Google Scholar]
  11. Neves, A.R.; Lucio, M.; Lima, J.L.; Reis, S. Resveratrol in medicinal chemistry: A critical review of its pharmacokinetics, drug-delivery, and membrane interactions. Curr. Med. Chem. 2012, 19, 1663–1681. [Google Scholar] [CrossRef] [PubMed]
  12. Constantinescu, T.; Mihis, A.G. Resveratrol as a privileged molecule with antioxidant activity. Food Chem. Adv. 2023, 3, 100539. [Google Scholar] [CrossRef]
  13. Meng, T.; Xiao, D.; Muhammed, A.; Deng, J.; Chen, L.; He, J. Anti-Inflammatory Action and Mechanisms of Resveratrol. Molecules 2021, 26, 229. [Google Scholar] [CrossRef] [PubMed]
  14. Kursvietiene, L.; Kopustinskiene, D.M.; Staneviciene, I.; Mongirdiene, A.; Kubová, K.; Masteikova, R.; Bernatoniene, J. Anti-Cancer Properties of Resveratrol: A Focus on Its Impact on Mitochondrial Functions. Antioxidants 2023, 12, 2056. [Google Scholar] [CrossRef] [PubMed]
  15. Gal, R.; Deres, L.; Toth, K.; Halmosi, R.; Habon, T. The Effect of Resveratrol on the Cardiovascular System from Molecular Mechanisms to Clinical Results. Int. J. Mol. Sci. 2021, 22, 10152. [Google Scholar] [CrossRef]
  16. Ren, B.; Kwah, M.X.-Y.; Liu, C.; Ma, Z.; Shanmugam, M.K.; Ding, L.; Xiang, X.; Ho, P.C.-L.; Wang, L.; Ong, P.S.; et al. Resveratrol for cancer therapy: Challenges and future perspectives. Cancer Lett. 2021, 515, 63–72. [Google Scholar] [CrossRef]
  17. Rege, S.D.; Geetha, T.; Griffin, G.D.; Broderick, T.L.; Babu, J.R. Neuroprotective effects of resveratrol in Alzheimer disease pathology. Front. Aging Neurosci. 2014, 6, 218. [Google Scholar] [CrossRef]
  18. Sun, A.Y.; Wang, Q.; Simonyi, A.; Sun, G.Y. Resveratrol as a therapeutic agent for neurodegenerative diseases. Mol. Neurobiol. 2010, 41, 375–383. [Google Scholar] [CrossRef]
  19. Agbadua, O.G.; Kúsz, N.; Berkecz, R.; Gáti, T.; Tóth, G.; Hunyadi, A. Oxidized Resveratrol Metabolites as Potent Antioxidants and Xanthine Oxidase Inhibitors. Antioxidants 2022, 11, 1832. [Google Scholar] [CrossRef]
  20. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  21. Jelic, M.D.; Mandic, A.D.; Maricic, S.M.; Srdjenovic, B.U. Oxidative stress and its role in cancer. J. Cancer Res. Ther. 2021, 17, 22–28. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, J.Q.; Kosten, T.R.; Zhang, X.Y. Free radicals, antioxidant defense systems, and schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2013, 46, 200–206. [Google Scholar] [CrossRef] [PubMed]
  23. Aaseth, J.; Skalny, A.V.; Roos, P.M.; Alexander, J.; Aschner, M.; Tinkov, A.A. Copper, Iron, Selenium and Lipo-Glycemic Dysmetabolism in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 9461. [Google Scholar] [CrossRef] [PubMed]
  24. Farhan, M. Cytotoxic Activity of the Red Grape Polyphenol Resveratrol against Human Prostate Cancer Cells: A Molecular Mechanism Mediated by Mobilization of Nuclear Copper and Generation of Reactive Oxygen Species. Life 2024, 14, 611. [Google Scholar] [CrossRef]
  25. Chaudhary, P.; Janmeda, P.; Docea, A.O.; Yeskaliyeva, B.; Abdull Razis, A.F.; Modu, B.; Calina, D.; Sharifi-Rad, J. Oxidative stress, free radicals and antioxidants: Potential crosstalk in the pathophysiology of human diseases. Front. Chem. 2023, 11, 1158198. [Google Scholar] [CrossRef]
  26. Iqbal, M.J.; Kabeer, A.; Abbas, Z.; Siddiqui, H.A.; Calina, D.; Sharifi-Rad, J.; Cho, W.C. Interplay of oxidative stress, cellular communication and signaling pathways in cancer. Cell Commun. Signal. 2024, 22, 7. [Google Scholar] [CrossRef]
  27. Gligorijević, N.; Stanić-Vučinić, D.; Radomirović, M.; Stojadinović, M.; Khulal, U.; Nedić, O.; Ćirković Veličković, T. Role of Resveratrol in Prevention and Control of Cardiovascular Disorders and Cardiovascular Complications Related to COVID-19 Disease: Mode of Action and Approaches Explored to Increase Its Bioavailability. Molecules 2021, 26, 2834. [Google Scholar] [CrossRef]
  28. Bartra, C.; Yuan, Y.; Vuraić, K.; Valdés-Quiroz, H.; Garcia-Baucells, P.; Slevin, M.; Pastorello, Y.; Suñol, C.; Sanfeliu, C. Resveratrol Activates Antioxidant Protective Mechanisms in Cellular Models of Alzheimer’s Disease Inflammation. Antioxidants 2024, 13, 177. [Google Scholar] [CrossRef]
  29. Su, T.; Zhang, Z.; Han, X.; Yang, F.; Wang, Z.; Cheng, Y.; Liu, H. Systematic Insight of Resveratrol Activated SIRT1 Interactome through Proximity Labeling Strategy. Antioxidants 2022, 11, 2330. [Google Scholar] [CrossRef]
  30. Guarente, L.; Picard, F. Calorie restriction—The SIR2 connection. Cell 2005, 120, 473–482. [Google Scholar] [CrossRef]
  31. Nogueiras, R.; Habegger, K.M.; Chaudhary, N.; Finan, B.; Banks, A.S.; Dietrich, M.O.; Horvath, T.L.; Sinclair, D.A.; Pfluger, P.T.; Tschöp, M.H. Sirtuin 1 and sirtuin 3: Physiological modulators of metabolism. Physiol. Rev. 2012, 92, 1479–1514. [Google Scholar] [CrossRef] [PubMed]
  32. Daenthanasanmak, A.; Iamsawat, S.; Chakraborty, P.; Nguyen, H.D.; Bastian, D.; Liu, C.; Mehrotra, S.; Yu, X.Z. Targeting Sirt-1 controls GVHD by inhibiting T-cell allo-response and promoting Treg stability in mice. Blood 2019, 133, 266–279. [Google Scholar] [CrossRef] [PubMed]
  33. Iside, C.; Scafuro, M.; Nebbioso, A.; Altucci, L. SIRT1 Activation by Natural Phytochemicals: An Overview. Front. Pharmacol. 2020, 11, 1225. [Google Scholar] [CrossRef]
  34. Wenz, T. Mitochondria and PGC-1α in Aging and Age-Associated Diseases. J. Aging Res. 2011, 2011, 810619. [Google Scholar] [CrossRef]
  35. Li, Q.; Jiang, X.; Zhou, Y.; Gu, Y.; Ding, Y.; Luo, J.; Pang, N.; Sun, Y.; Pei, L.; Pan, J.; et al. Improving Mitochondrial Function in Skeletal Muscle Contributes to the Amelioration of Insulin Resistance by Nicotinamide Riboside. Int. J. Mol. Sci. 2023, 24, 10015. [Google Scholar] [CrossRef]
  36. Solomon, J.M.; Pasupuleti, R.; Xu, L.; McDonagh, T.; Curtis, R.; DiStefano, P.S.; Huber, L.J. Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage. Mol. Cell. Biol. 2006, 26, 28–38. [Google Scholar] [CrossRef]
  37. Yi, J.; Luo, J. SIRT1 and p53, effect on cancer, senescence and beyond. Biochim. Biophys. Acta 2010, 1804, 1684–1689. [Google Scholar] [CrossRef]
  38. Engeland, K. Cell cycle regulation: p53-p21-RB signaling. Cell Death Differ. 2022, 29, 946–960. [Google Scholar] [CrossRef]
  39. Shen, Y.; White, E. p53-dependent apoptosis pathways. Adv. Cancer Res. 2001, 82, 55–84. [Google Scholar] [CrossRef]
  40. Xu, L.; Botchway, B.O.A.; Zhang, S.; Zhou, J.; Liu, X. Inhibition of NF-κB Signaling Pathway by Resveratrol Improves Spinal Cord Injury. Front. Neurosci. 2018, 12, 690. [Google Scholar] [CrossRef]
  41. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed]
  42. Ghafouri-Fard, S.; Bahroudi, Z.; Shoorei, H.; Hussen, B.M.; Talebi, S.F.; Baig, S.G.; Taheri, M.; Ayatollahi, S.A. Disease-associated regulation of gene expression by resveratrol: Special focus on the PI3K/AKT signaling pathway. Cancer Cell Int. 2022, 22, 298. [Google Scholar] [CrossRef] [PubMed]
  43. Jiang, H.; Shang, X.; Wu, H.; Gautam, S.C.; Al-Holou, S.; Li, C.; Kuo, J.; Zhang, L.; Chopp, M. Resveratrol downregulates PI3K/Akt/mTOR signaling pathways in human U251 glioma cells. J. Exp. Ther. Oncol. 2009, 8, 25–33. [Google Scholar]
  44. Wu, H.; Chen, L.; Zhu, F.; Han, X.; Sun, L.; Chen, K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins 2019, 11, 731. [Google Scholar] [CrossRef]
  45. Singh, S.K.; Banerjee, S.; Acosta, E.P.; Lillard, J.W.; Singh, R. Resveratrol induces cell cycle arrest and apoptosis with docetaxel in prostate cancer cells via a p53/ p21WAF1/CIP1 and p27KIP1 pathway. Oncotarget 2017, 8, 17216–17228. [Google Scholar] [CrossRef]
  46. Hu, X.; Wang, J.; Xia, Y.; Simayi, M.; Ikramullah, S.; He, Y.; Cui, S.; Li, S.; Wushouer, Q. Resveratrol induces cell cycle arrest and apoptosis in human eosinophils from asthmatic individuals. Mol. Med. Rep. 2016, 14, 5231–5236. [Google Scholar] [CrossRef]
  47. Ding, L.; Cao, J.; Lin, W.; Chen, H.; Xiong, X.; Ao, H.; Yu, M.; Lin, J.; Cui, Q. The Roles of Cyclin-Dependent Kinases in Cell-Cycle Progression and Therapeutic Strategies in Human Breast Cancer. Int. J. Mol. Sci. 2020, 21, 1960. [Google Scholar] [CrossRef]
  48. Ye, M.; Tian, H.; Lin, S.; Mo, J.; Li, Z.; Chen, X.; Liu, J. Resveratrol inhibits proliferation and promotes apoptosis via the androgen receptor splicing variant 7 and PI3K/AKT signaling pathway in LNCaP prostate cancer cells. Oncol. Lett. 2020, 20, 169. [Google Scholar] [CrossRef]
  49. Takashina, M.; Inoue, S.; Tomihara, K.; Tomita, K.; Hattori, K.; Zhao, Q.-L.; Suzuki, T.; Noguchi, M.; Ohashi, W.; Hattori, Y. Different effect of resveratrol to induction of apoptosis depending on the type of human cancer cells. Int. J. Oncol. 2017, 50, 787–797. [Google Scholar] [CrossRef]
  50. Jang, J.Y.; Im, E.; Kim, N.D. Mechanism of Resveratrol-Induced Programmed Cell Death and New Drug Discovery against Cancer: A Review. Int. J. Mol. Sci. 2022, 23, 13689. [Google Scholar] [CrossRef]
  51. Gogvadze, V.; Orrenius, S.; Zhivotovsky, B. Multiple pathways of cytochrome c release from mitochondria in apoptosis. Biochim. Et Biophys. Acta (BBA) Bioenerg. 2006, 1757, 639–647. [Google Scholar] [CrossRef] [PubMed]
  52. Laurindo, L.F.; Santos, A.; Carvalho, A.C.A.; Bechara, M.D.; Guiguer, E.L.; Goulart, R.A.; Vargas Sinatora, R.; Araújo, A.C.; Barbalho, S.M. Phytochemicals and Regulation of NF-kB in Inflammatory Bowel Diseases: An Overview of In Vitro and In Vivo Effects. Metabolites 2023, 13, 96. [Google Scholar] [CrossRef] [PubMed]
  53. Zykova, T.A.; Zhu, F.; Zhai, X.; Ma, W.Y.; Ermakova, S.P.; Lee, K.W.; Bode, A.M.; Dong, Z. Resveratrol directly targets COX-2 to inhibit carcinogenesis. Mol. Carcinog. 2008, 47, 797–805. [Google Scholar] [CrossRef]
  54. Dupont, C.; Armant, D.R.; Brenner, C.A. Epigenetics: Definition, mechanisms and clinical perspective. Semin. Reprod. Med. 2009, 27, 351–357. [Google Scholar] [CrossRef]
  55. Wang, Z.A.; Hsu, W.; Liu, W.R. Role of SIRT1 in Epigenetics. In Handbook of Nutrition, Diet, and Epigenetics; Patel, V.B., Preedy, V.R., Eds.; Springer International Publishing: Cham, Switzerland, 2019; pp. 311–329. [Google Scholar]
  56. Jin, B.; Li, Y.; Robertson, K.D. DNA methylation: Superior or subordinate in the epigenetic hierarchy? Genes. Cancer 2011, 2, 607–617. [Google Scholar] [CrossRef]
  57. Fernandes, G.F.S.; Silva, G.D.B.; Pavan, A.R.; Chiba, D.E.; Chin, C.M.; Dos Santos, J.L. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017, 9, 1201. [Google Scholar] [CrossRef]
  58. Bae, S.; Lee, E.M.; Cha, H.J.; Kim, K.; Yoon, Y.; Lee, H.; Kim, J.; Kim, Y.J.; Lee, H.G.; Jeung, H.K.; et al. Resveratrol alters microRNA expression profiles in A549 human non-small cell lung cancer cells. Mol. Cells 2011, 32, 243–249. [Google Scholar] [CrossRef]
  59. Otsuka, K.; Yamamoto, Y.; Ochiya, T. Regulatory role of resveratrol, a microRNA-controlling compound, in HNRNPA1 expression, which is associated with poor prognosis in breast cancer. Oncotarget 2018, 9, 24718–24730. [Google Scholar] [CrossRef]
  60. Di Cerbo, V.; Schneider, R. Cancers with wrong HATs: The impact of acetylation. Brief. Funct. Genom. 2013, 12, 231–243. [Google Scholar] [CrossRef]
  61. Ohshiro, K.; Rayala, S.K.; Kondo, S.; Gaur, A.; Vadlamudi, R.K.; El-Naggar, A.K.; Kumar, R. Identifying the estrogen receptor coactivator PELP1 in autophagosomes. Cancer Res. 2007, 67, 8164–8171. [Google Scholar] [CrossRef]
  62. Whyte, L.; Huang, Y.Y.; Torres, K.; Mehta, R.G. Molecular mechanisms of resveratrol action in lung cancer cells using dual protein and microarray analyses. Cancer Res. 2007, 67, 12007–12017. [Google Scholar] [CrossRef] [PubMed]
  63. Ebi, H.; Tomida, S.; Takeuchi, T.; Arima, C.; Sato, T.; Mitsudomi, T.; Yatabe, Y.; Osada, H.; Takahashi, T. Relationship of Deregulated Signaling Converging onto mTOR with Prognosis and Classification of Lung Adenocarcinoma Shown by Two Independent In silico Analyses. Cancer Res. 2009, 69, 4027–4035. [Google Scholar] [CrossRef] [PubMed]
  64. Saxton, R.A.; Sabatini, D.M. mTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef]
  65. Wright, C.; Iyer, A.K.V.; Yakisich, J.S.; Azad, N. Anti-Tumorigenic Effects of Resveratrol in Lung Cancer Cells Through Modulation of c-FLIP. Curr. Cancer Drug Targets 2017, 17, 669–680. [Google Scholar] [CrossRef]
  66. Li, W.; Li, C.; Ma, L.; Jin, F. Resveratrol inhibits viability and induces apoptosis in the small-cell lung cancer H446 cell line via the PI3K/Akt/c-Myc pathway. Oncol. Rep. 2020, 44, 1821–1830. [Google Scholar] [CrossRef]
  67. Hsieh, T.C.; Wu, J.M. Grape-derived chemopreventive agent resveratrol decreases prostate-specific antigen (PSA) expression in LNCaP cells by an androgen receptor (AR)-independent mechanism. Anticancer. Res. 2000, 20, 225–228. [Google Scholar]
  68. Benitez, D.A.; Pozo-Guisado, E.; Clementi, M.; Castellón, E.; Fernandez-Salguero, P.M. Non-genomic action of resveratrol on androgen and oestrogen receptors in prostate cancer: Modulation of the phosphoinositide 3-kinase pathway. Br. J. Cancer 2007, 96, 1595–1604. [Google Scholar] [CrossRef]
  69. Wang, T.T.; Hudson, T.S.; Wang, T.C.; Remsberg, C.M.; Davies, N.M.; Takahashi, Y.; Kim, Y.S.; Seifried, H.; Vinyard, B.T.; Perkins, S.N.; et al. Differential effects of resveratrol on androgen-responsive LNCaP human prostate cancer cells in vitro and in vivo. Carcinogenesis 2008, 29, 2001–2010. [Google Scholar] [CrossRef]
  70. Jones, S.B.; DePrimo, S.E.; Whitfield, M.L.; Brooks, J.D. Resveratrol-induced gene expression profiles in human prostate cancer cells. Cancer Epidemiol. Biomark. Prev. 2005, 14, 596–604. [Google Scholar] [CrossRef]
  71. Seeni, A.; Takahashi, S.; Takeshita, K.; Tang, M.; Sugiura, S.; Sato, S.Y.; Shirai, T. Suppression of prostate cancer growth by resveratrol in the transgenic rat for adenocarcinoma of prostate (TRAP) model. Asian Pac. J. Cancer Prev. 2008, 9, 7–14. [Google Scholar]
  72. ALkharashi, N.A. Efficacy of resveratrol against breast cancer and hepatocellular carcinoma cell lines. Saudi Med. J. 2023, 44, 246–252. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, Y.N.; Choe, S.R.; Cho, K.H.; Cho, D.Y.; Kang, J.; Park, C.G.; Lee, H.Y. Resveratrol suppresses breast cancer cell invasion by inactivating a RhoA/YAP signaling axis. Exp. Mol. Med. 2017, 49, e296. [Google Scholar] [CrossRef] [PubMed]
  74. Buhrmann, C.; Shayan, P.; Goel, A.; Shakibaei, M. Resveratrol Regulates Colorectal Cancer Cell Invasion by Modulation of Focal Adhesion Molecules. Nutrients 2017, 9, 1073. [Google Scholar] [CrossRef] [PubMed]
  75. Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef]
  76. Chen, H.J.; Hsu, L.S.; Shia, Y.T.; Lin, M.W.; Lin, C.M. The β-catenin/TCF complex as a novel target of resveratrol in the Wnt/β-catenin signaling pathway. Biochem. Pharmacol. 2012, 84, 1143–1153. [Google Scholar] [CrossRef]
  77. Fontecave, M.; Lepoivre, M.; Elleingand, E.; Gerez, C.; Guittet, O. Resveratrol, a remarkable inhibitor of ribonucleotide reductase. FEBS Lett. 1998, 421, 277–279. [Google Scholar] [CrossRef]
  78. Dai, H.; Li, M.; Yang, W.; Sun, X.; Wang, P.; Wang, X.; Su, J.; Wang, X.; Hu, X.; Zhao, M. Resveratrol inhibits the malignant progression of hepatocellular carcinoma via MARCH1-induced regulation of PTEN/AKT signaling. Aging 2020, 12, 11717–11731. [Google Scholar] [CrossRef]
  79. Wang, Y.; Su, L.; Hu, Z.; Peng, S.; Li, N.; Fu, H.; Wang, B.; Wu, H. Resveratrol suppresses liver cancer progression by downregulating AKR1C3: Targeting HCC with HSA nanomaterial as a carrier to enhance therapeutic efficacy. Apoptosis 2024, 29, 1429–1453. [Google Scholar] [CrossRef]
  80. Gao, F.; Deng, G.; Liu, W.; Zhou, K.; Li, M. Resveratrol suppresses human hepatocellular carcinoma via targeting HGF-c-Met signaling pathway. Oncol. Rep. 2017, 37, 1203–1211. [Google Scholar] [CrossRef]
  81. Ratajczak, K.; Glatzel-Plucińska, N.; Ratajczak-Wielgomas, K.; Nowińska, K.; Borska, S. Effect of Resveratrol Treatment on Human Pancreatic Cancer Cells through Alterations of Bcl-2 Family Members. Molecules 2021, 26, 6560. [Google Scholar] [CrossRef]
  82. Yang, L.; Yang, L.; Tian, W.; Li, J.; Liu, J.; Zhu, M.; Zhang, Y.; Yang, Y.; Liu, F.; Zhang, Q.; et al. Resveratrol plays dual roles in pancreatic cancer cells. J. Cancer Res. Clin. Oncol. 2014, 140, 749–755. [Google Scholar] [CrossRef] [PubMed]
  83. Qin, T.; Cheng, L.; Xiao, Y.; Qian, W.; Li, J.; Wu, Z.; Wang, Z.; Xu, Q.; Duan, W.; Wong, L.; et al. NAF-1 Inhibition by Resveratrol Suppresses Cancer Stem Cell-Like Properties and the Invasion of Pancreatic Cancer. Front. Oncol. 2020, 10, 1038. [Google Scholar] [CrossRef] [PubMed]
  84. Mo, W.; Xu, X.; Xu, L.; Wang, F.; Ke, A.; Wang, X.; Guo, C. Resveratrol inhibits proliferation and induces apoptosis through the hedgehog signaling pathway in pancreatic cancer cell. Pancreatology 2011, 11, 601–609. [Google Scholar] [CrossRef]
  85. Ding, X.Z.; Adrian, T.E. Resveratrol inhibits proliferation and induces apoptosis in human pancreatic cancer cells. Pancreas 2002, 25, e71–e76. [Google Scholar] [CrossRef]
  86. Li, W.; Ma, J.; Ma, Q.; Li, B.; Han, L.; Liu, J.; Xu, Q.; Duan, W.; Yu, S.; Wang, F.; et al. Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-κB pathway. Curr. Med. Chem. 2013, 20, 4185–4194. [Google Scholar] [CrossRef]
  87. Tessitore, L.; Davit, A.; Sarotto, I.; Caderni, G. Resveratrol depresses the growth of colorectal aberrant crypt foci by affecting bax and p21(CIP) expression. Carcinogenesis 2000, 21, 1619–1622. [Google Scholar] [CrossRef]
  88. Majumdar, A.P.; Banerjee, S.; Nautiyal, J.; Patel, B.B.; Patel, V.; Du, J.; Yu, Y.; Elliott, A.A.; Levi, E.; Sarkar, F.H. Curcumin synergizes with resveratrol to inhibit colon cancer. Nutr. Cancer 2009, 61, 544–553. [Google Scholar] [CrossRef]
  89. Sengottuvelan, M.; Nalini, N. Dietary supplementation of resveratrol suppresses colonic tumour incidence in 1,2-dimethylhydrazine-treated rats by modulating biotransforming enzymes and aberrant crypt foci development. Br. J. Nutr. 2006, 96, 145–153. [Google Scholar] [CrossRef]
  90. Alfaras, I.; Juan, M.E.; Planas, J.M. trans-Resveratrol reduces precancerous colonic lesions in dimethylhydrazine-treated rats. J. Agric. Food Chem. 2010, 58, 8104–8110. [Google Scholar] [CrossRef]
  91. Bove, K.; Lincoln, D.W.; Tsan, M.F. Effect of resveratrol on growth of 4T1 breast cancer cells in vitro and in vivo. Biochem. Biophys. Res. Commun. 2002, 291, 1001–1005. [Google Scholar] [CrossRef]
  92. Bhat, K.P.; Lantvit, D.; Christov, K.; Mehta, R.G.; Moon, R.C.; Pezzuto, J.M. Estrogenic and antiestrogenic properties of resveratrol in mammary tumor models. Cancer Res. 2001, 61, 7456–7463. [Google Scholar] [PubMed]
  93. Provinciali, M.; Re, F.; Donnini, A.; Orlando, F.; Bartozzi, B.; Di Stasio, G.; Smorlesi, A. Effect of resveratrol on the development of spontaneous mammary tumors in HER-2/neu transgenic mice. Int. J. Cancer 2005, 115, 36–45. [Google Scholar] [CrossRef] [PubMed]
  94. Castillo-Pichardo, L.; Cubano, L.A.; Dharmawardhane, S. Dietary grape polyphenol resveratrol increases mammary tumor growth and metastasis in immunocompromised mice. BMC Complement. Altern. Med. 2013, 13, 6. [Google Scholar] [CrossRef] [PubMed]
  95. Rajasekaran, D.; Elavarasan, J.; Sivalingam, M.; Ganapathy, E.; Kumar, A.; Kalpana, K.; Sakthisekaran, D. Resveratrol interferes with N-nitrosodiethylamine-induced hepatocellular carcinoma at early and advanced stages in male Wistar rats. Mol. Med. Rep. 2011, 4, 1211–1217. [Google Scholar] [CrossRef]
  96. Lin, H.C.; Chen, Y.F.; Hsu, W.H.; Yang, C.W.; Kao, C.H.; Tsai, T.F. Resveratrol helps recovery from fatty liver and protects against hepatocellular carcinoma induced by hepatitis B virus X protein in a mouse model. Cancer Prev. Res. (Phila.) 2012, 5, 952–962. [Google Scholar] [CrossRef]
  97. Salado, C.; Olaso, E.; Gallot, N.; Valcarcel, M.; Egilegor, E.; Mendoza, L.; Vidal-Vanaclocha, F. Resveratrol prevents inflammation-dependent hepatic melanoma metastasis by inhibiting the secretion and effects of interleukin-18. J. Transl. Med. 2011, 9, 59. [Google Scholar] [CrossRef]
  98. Luther, D.J.; Ohanyan, V.; Shamhart, P.E.; Hodnichak, C.M.; Sisakian, H.; Booth, T.D.; Meszaros, J.G.; Bishayee, A. Chemopreventive doses of resveratrol do not produce cardiotoxicity in a rodent model of hepatocellular carcinoma. Investig. New Drugs 2011, 29, 380–391. [Google Scholar] [CrossRef]
  99. Yin, H.T.; Tian, Q.Z.; Guan, L.; Zhou, Y.; Huang, X.E.; Zhang, H. In vitro and in vivo evaluation of the antitumor efficiency of resveratrol against lung cancer. Asian Pac. J. Cancer Prev. 2013, 14, 1703–1706. [Google Scholar] [CrossRef]
  100. Savio, M.; Ferraro, D.; Maccario, C.; Vaccarone, R.; Jensen, L.D.; Corana, F.; Mannucci, B.; Bianchi, L.; Cao, Y.; Stivala, L.A. Resveratrol analogue 4,4′-dihydroxy-trans-stilbene potently inhibits cancer invasion and metastasis. Sci. Rep. 2016, 6, 19973. [Google Scholar] [CrossRef]
  101. Zhao, W.; Bao, P.; Qi, H.; You, H. Resveratrol down-regulates survivin and induces apoptosis in human multidrug-resistant SPC-A-1/CDDP cells. Oncol. Rep. 2010, 23, 279–286. [Google Scholar] [CrossRef]
  102. Wu, J.Y.; Tsai, K.W.; Shee, J.J.; Li, Y.Z.; Chen, C.H.; Chuang, J.J.; Liu, Y.W. 4′-Chloro-3,5-dihydroxystilbene, a resveratrol derivative, induces lung cancer cell death. Acta Pharmacol. Sin. 2010, 31, 81–92. [Google Scholar] [CrossRef] [PubMed]
  103. Sun, L.; Chen, B.; Jiang, R.; Li, J.; Wang, B. Resveratrol inhibits lung cancer growth by suppressing M2-like polarization of tumor associated macrophages. Cell. Immunol. 2017, 311, 86–93. [Google Scholar] [CrossRef] [PubMed]
  104. Dias, S.J.; Li, K.; Rimando, A.M.; Dhar, S.; Mizuno, C.S.; Penman, A.D.; Levenson, A.S. Trimethoxy-resveratrol and piceatannol administered orally suppress and inhibit tumor formation and growth in prostate cancer xenografts. Prostate 2013, 73, 1135–1146. [Google Scholar] [CrossRef] [PubMed]
  105. Harper, C.E.; Patel, B.B.; Wang, J.; Arabshahi, A.; Eltoum, I.A.; Lamartiniere, C.A. Resveratrol suppresses prostate cancer progression in transgenic mice. Carcinogenesis 2007, 28, 1946–1953. [Google Scholar] [CrossRef]
  106. Roy, S.K.; Chen, Q.; Fu, J.; Shankar, S.; Srivastava, R.K. Resveratrol inhibits growth of orthotopic pancreatic tumors through activation of FOXO transcription factors. PLoS ONE 2011, 6, e25166. [Google Scholar] [CrossRef]
  107. Shankar, S.; Nall, D.; Tang, S.N.; Meeker, D.; Passarini, J.; Sharma, J.; Srivastava, R.K. Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial-mesenchymal transition. PLoS ONE 2011, 6, e16530. [Google Scholar] [CrossRef]
  108. Oi, N.; Jeong, C.H.; Nadas, J.; Cho, Y.Y.; Pugliese, A.; Bode, A.M.; Dong, Z. Resveratrol, a red wine polyphenol, suppresses pancreatic cancer by inhibiting leukotriene A4hydrolase. Cancer Res. 2010, 70, 9755–9764. [Google Scholar] [CrossRef]
  109. Harikumar, K.B.; Kunnumakkara, A.B.; Sethi, G.; Diagaradjane, P.; Anand, P.; Pandey, M.K.; Gelovani, J.; Krishnan, S.; Guha, S.; Aggarwal, B.B. Resveratrol, a multitargeted agent, can enhance antitumor activity of gemcitabine in vitro and in orthotopic mouse model of human pancreatic cancer. Int. J. Cancer 2010, 127, 257–268. [Google Scholar] [CrossRef]
  110. Sudha, T.; El-Far, A.H.; Mousa, D.S.; Mousa, S.A. Resveratrol and Its Nanoformulation Attenuate Growth and the Angiogenesis of Xenograft and Orthotopic Colon Cancer Models. Molecules 2020, 25, 1412. [Google Scholar] [CrossRef]
  111. Abdul-Rahman, T.; Dunham, A.; Huang, H.; Bukhari, S.M.A.; Mehta, A.; Awuah, W.A.; Ede-Imafidon, D.; Cantu-Herrera, E.; Talukder, S.; Joshi, A.; et al. Chemotherapy Induced Cardiotoxicity: A State of the Art Review on General Mechanisms, Prevention, Treatment and Recent Advances in Novel Therapeutics. Curr. Probl. Cardiol. 2023, 48, 101591. [Google Scholar] [CrossRef]
  112. Volkova, M.; Russell, R., III. Anthracycline cardiotoxicity: Prevalence, pathogenesis and treatment. Curr. Cardiol. Rev. 2011, 7, 214–220. [Google Scholar] [CrossRef] [PubMed]
  113. Rawat, P.S.; Jaiswal, A.; Khurana, A.; Bhatti, J.S.; Navik, U. Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed. Pharmacother. 2021, 139, 111708. [Google Scholar] [CrossRef] [PubMed]
  114. Cappetta, D.; De Angelis, A.; Sapio, L.; Prezioso, L.; Illiano, M.; Quaini, F.; Rossi, F.; Berrino, L.; Naviglio, S.; Urbanek, K. Oxidative Stress and Cellular Response to Doxorubicin: A Common Factor in the Complex Milieu of Anthracycline Cardiotoxicity. Oxidative Med. Cell. Longev. 2017, 2017, 1521020. [Google Scholar] [CrossRef] [PubMed]
  115. Sheu, M.T.; Jhan, H.J.; Hsieh, C.M.; Wang, C.J.; Ho, H.O. Efficacy of antioxidants as a Complementary and Alternative Medicine (CAM) in combination with the chemotherapeutic agent doxorubicin. Integr. Cancer Ther. 2015, 14, 184–195. [Google Scholar] [CrossRef]
  116. Ito, Y.; Mitani, T.; Harada, N.; Isayama, A.; Tanimori, S.; Takenaka, S.; Nakano, Y.; Inui, H.; Yamaji, R. Identification of carbonyl reductase 1 as a resveratrol-binding protein by affinity chromatography using 4′-amino-3,5-dihydroxy-trans-stilbene. J. Nutr. Sci. Vitaminol. (Tokyo) 2013, 59, 358–364. [Google Scholar] [CrossRef]
  117. Wang, L.; Kou, M.C.; Weng, C.Y.; Hu, L.W.; Wang, Y.J.; Wu, M.J. Arsenic modulates heme oxygenase-1, interleukin-6, and vascular endothelial growth factor expression in endothelial cells: Roles of ROS, NF-κB, and MAPK pathways. Arch. Toxicol. 2012, 86, 879–896. [Google Scholar] [CrossRef]
  118. Emadi, A.; Gore, S.D. Arsenic trioxide—An old drug rediscovered. Blood Rev. 2010, 24, 191–199. [Google Scholar] [CrossRef]
  119. Zhao, X.Y.; Li, G.Y.; Liu, Y.; Chai, L.M.; Chen, J.X.; Zhang, Y.; Du, Z.M.; Lu, Y.J.; Yang, B.F. Resveratrol protects against arsenic trioxide-induced cardiotoxicity in vitro and in vivo. Br. J. Pharmacol. 2008, 154, 105–113. [Google Scholar] [CrossRef]
  120. Akbarali, H.I.; Muchhala, K.H.; Jessup, D.K.; Cheatham, S. Chemotherapy induced gastrointestinal toxicities. Adv. Cancer Res. 2022, 155, 131–166. [Google Scholar] [CrossRef]
  121. Ko, J.L.; Tsai, C.H.; Liu, T.C.; Lin, M.Y.; Lin, H.L.; Ou, C.C. Differential effects of grape juice on gastric emptying and renal function from cisplatin-induced acute adverse toxicity. Hum. Exp. Toxicol. 2016, 35, 808–817. [Google Scholar] [CrossRef]
  122. Hu, W.H.; Chan, G.K.; Duan, R.; Wang, H.Y.; Kong, X.P.; Dong, T.T.; Tsim, K.W. Synergy of Ginkgetin and Resveratrol in Suppressing VEGF-Induced Angiogenesis: A Therapy in Treating Colorectal Cancer. Cancers 2019, 11, 1828. [Google Scholar] [CrossRef] [PubMed]
  123. Tomikoshi, Y.; Nomura, M.; Okudaira, N.; Sakagami, H.; Wakabayashi, H. Enhancement of cytotoxicity of three apoptosis-inducing agents against human oral squamous cell carcinoma cell line by benzoxazinotropone. Vivo 2016, 30, 645–650. [Google Scholar]
  124. Kong, F.; Zhang, R.; Zhao, X.; Zheng, G.; Wang, Z.; Wang, P. Resveratrol raises in vitro anticancer effects of paclitaxel in NSCLC cell line A549 through COX-2 expression. Korean J. Physiol. Pharmacol. 2017, 21, 465–474. [Google Scholar] [CrossRef]
  125. Rahimifard, M.; Baeeri, M.; Mousavi, T.; Azarnezhad, A.; Haghi-Aminjan, H.; Abdollahi, M. Combination therapy of cisplatin and resveratrol to induce cellular aging in gastric cancer cells: Focusing on oxidative stress, and cell cycle arrest. Front. Pharmacol. 2023, 13, 1068863. [Google Scholar] [CrossRef] [PubMed]
  126. Ren, M.; Zhou, X.; Gu, M.; Jiao, W.; Yu, M.; Wang, Y.; Liu, S.; Yang, J.; Ji, F. Resveratrol synergizes with cisplatin in antineoplastic effects against AGS gastric cancer cells by inducing endoplasmic reticulum stress-mediated apoptosis and G2/M phase arrest. Oncol. Rep. 2020, 44, 1605–1615. [Google Scholar] [CrossRef]
  127. De Luca, A.; Bellavia, D.; Raimondi, L.; Carina, V.; Costa, V.; Fini, M.; Giavaresi, G. Multiple Effects of Resveratrol on Osteosarcoma Cell Lines. Pharmaceuticals 2022, 15, 342. [Google Scholar] [CrossRef]
  128. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.W. Cancer and radiation therapy: Current advances and future directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef]
  129. Agbele, A.T.; Fasoro, O.J.; Fabamise, O.M.; Oluyide, O.O.; Idolor, O.R.; Bamise, E.A. Protection Against Ionizing Radiation-Induced Normal Tissue Damage by Resveratrol: A Systematic Review. Eurasian J. Med. 2020, 52, 298–303. [Google Scholar] [CrossRef]
  130. Komorowska, D.; Radzik, T.; Kalenik, S.; Rodacka, A. Natural Radiosensitizers in Radiotherapy: Cancer Treatment by Combining Ionizing Radiation with Resveratrol. Int. J. Mol. Sci. 2022, 23, 10627. [Google Scholar] [CrossRef]
  131. Wang, Z.; Dabrosin, C.; Yin, X.; Fuster, M.M.; Arreola, A.; Rathmell, W.K.; Generali, D.; Nagaraju, G.P.; El-Rayes, B.; Ribatti, D. Broad targeting of angiogenesis for cancer prevention and therapy. In Proceedings of the Seminars in Cancer Biology, New Paltz, NY, USA, 15–17 June 2015; pp. S224–S243. [Google Scholar]
  132. Wang, L.; Long, L.; Wang, W.; Liang, Z. Resveratrol, a potential radiation sensitizer for glioma stem cells both in vitro and in vivo. J. Pharmacol. Sci. 2015, 129, 216–225. [Google Scholar] [CrossRef]
  133. Kma, L. Synergistic effect of resveratrol and radiotherapy in control of cancers. Asian Pac. J. Cancer Prev. 2013, 14, 6197–6208. [Google Scholar] [CrossRef] [PubMed]
  134. Scarlatti, F.; Sala, G.; Ricci, C.; Maioli, C.; Milani, F.; Minella, M.; Botturi, M.; Ghidoni, R. Resveratrol sensitization of DU145 prostate cancer cells to ionizing radiation is associated to ceramide increase. Cancer Lett. 2007, 253, 124–130. [Google Scholar] [CrossRef] [PubMed]
  135. Rashid, A.; Liu, C.; Sanli, T.; Tsiani, E.; Singh, G.; Bristow, R.G.; Dayes, I.; Lukka, H.; Wright, J.; Tsakiridis, T. Resveratrol enhances prostate cancer cell response to ionizing radiation. Modulation of the AMPK, Akt and mTOR pathways. Radiat. Oncol. 2011, 6, 144. [Google Scholar] [CrossRef] [PubMed]
  136. Mikami, S.; Ota, I.; Masui, T.; Uchiyama, T.; Okamoto, H.; Kimura, T.; Takasawa, S.; Kitahara, T. Resveratrol-induced REG III expression enhances chemo-and radiosensitivity in head and neck cancer in xenograft mice. Oncol. Rep. 2019, 42, 436–442. [Google Scholar] [CrossRef]
  137. Chen, Y.-A.; Lien, H.-M.; Kao, M.-C.; Lo, U.-G.; Lin, L.-C.; Lin, C.-J.; Chang, S.-J.; Chen, C.-C.; Hsieh, J.-T.; Lin, H. Sensitization of radioresistant prostate cancer cells by resveratrol isolated from arachis hypogaea stems. PLoS ONE 2017, 12, e0169204. [Google Scholar] [CrossRef]
  138. Min, H.Y.; Lee, H.Y. Molecular targeted therapy for anticancer treatment. Exp. Mol. Med. 2022, 54, 1670–1694. [Google Scholar] [CrossRef]
  139. Yadav, P. Targeted cancer therapy; researchers reflection. Int. Peer-Rev. J. 2017, 1, 11–17. [Google Scholar]
  140. 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. [Google Scholar] [CrossRef]
  141. Jin, Z.; Feng, W.; Ji, Y.; Jin, L. Resveratrol mediates cell cycle arrest and cell death in human esophageal squamous cell carcinoma by directly targeting the EGFR signaling pathway. Oncol. Lett. 2017, 13, 347–355. [Google Scholar] [CrossRef]
  142. Abdel-Latif, G.A.; Al-Abd, A.M.; Tadros, M.G.; Al-Abbasi, F.A.; Khalifa, A.E.; Abdel-Naim, A.B. The chemomodulatory effects of resveratrol and didox on herceptin cytotoxicity in breast cancer cell lines. Sci. Rep. 2015, 5, 12054. [Google Scholar] [CrossRef]
  143. Zhang, Y.; Zhang, Z.; Mousavi, M.; Moliani, A.; Bahman, Y.; Bagheri, H. Resveratrol inhibits glioblastoma cells and chemoresistance progression through blockade P-glycoprotein and targeting AKT/PTEN signaling pathway. Chem. Biol. Interact. 2023, 376, 110409. [Google Scholar] [CrossRef]
  144. Hu, Y.; Li, C.; Li, H.; Li, M.; Shu, X. Resveratrol-mediated reversal of tumor multi-drug resistance. Curr. Drug Metab. 2014, 15, 703–710. [Google Scholar] [CrossRef]
  145. Zhu, Y.; He, W.; Gao, X.; Li, B.; Mei, C.; Xu, R.; Chen, H. Resveratrol overcomes gefitinib resistance by increasing the intracellular gefitinib concentration and triggering apoptosis, autophagy and senescence in PC9/G NSCLC cells. Sci. Rep. 2015, 5, 17730. [Google Scholar] [CrossRef]
  146. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef] [PubMed]
  147. Tormoen, G.W.; Crittenden, M.R.; Gough, M.J. Role of the immunosuppressive microenvironment in immunotherapy. Adv. Radiat. Oncol. 2018, 3, 520–526. [Google Scholar] [CrossRef] [PubMed]
  148. Alesci, A.; Nicosia, N.; Fumia, A.; Giorgianni, F.; Santini, A.; Cicero, N. Resveratrol and Immune Cells: A Link to Improve Human Health. Molecules 2022, 27, 424. [Google Scholar] [CrossRef]
  149. Basudan, A.M. The Role of Immune Checkpoint Inhibitors in Cancer Therapy. Clin. Pract. 2022, 13, 22–40. [Google Scholar] [CrossRef]
  150. Nigam, M.; Mishra, A.P.; Deb, V.K.; Dimri, D.B.; Tiwari, V.; Bungau, S.G.; Bungau, A.F.; Radu, A.-F. Evaluation of the association of chronic inflammation and cancer: Insights and implications. Biomed. Pharmacother. 2023, 164, 115015. [Google Scholar] [CrossRef]
  151. Chen, Y.; Pei, Y.; Luo, J.; Huang, Z.; Yu, J.; Meng, X. Looking for the Optimal PD-1/PD-L1 Inhibitor in Cancer Treatment: A Comparison in Basic Structure, Function, and Clinical Practice. Front. Immunol. 2020, 11, 1088. [Google Scholar] [CrossRef]
  152. Wang, B.; Bai, W.; Ma, H.; Li, F. Regulatory Effect of PD1/PD-Ligand 1 (PD-L1) on Treg Cells in Patients with Idiopathic Pulmonary Fibrosis. Med. Sci. Monit. 2021, 27, e927577. [Google Scholar] [CrossRef]
  153. Curran, M.A.; Montalvo, W.; Yagita, H.; Allison, J.P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 4275–4280. [Google Scholar] [CrossRef] [PubMed]
  154. Delmas, D.; Hermetet, F.; Aires, V. PD-1/PD-L1 Checkpoints and Resveratrol: A Controversial New Way for a Therapeutic Strategy. Cancers 2021, 13, 4509. [Google Scholar] [CrossRef] [PubMed]
  155. Pradhan, R.; Paul, S.; Acharya, S.S.; Sinha, S.; Dash, S.R.; Kundu, C.N. Nano formulated Resveratrol inhibits PD-L1 in oral cancer cells by deregulating the association between tumor associated macrophages and cancer associated fibroblasts through IL-6/JAK2/STAT3 signaling axis. J. Nutr. Biochem. 2024, 125, 109568. [Google Scholar] [CrossRef] [PubMed]
  156. Calvillo-Rodríguez, K.M.; Lorenzo-Anota, H.Y.; Rodríguez-Padilla, C.; Martínez-Torres, A.C.; Scott-Algara, D. Immunotherapies inducing immunogenic cell death in cancer: Insight of the innate immune system. Front. Immunol. 2023, 14, 1294434. [Google Scholar] [CrossRef]
  157. Zhang, Y.; Yang, S.; Yang, Y.; Liu, T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect. Agents Cancer 2019, 14, 27. [Google Scholar] [CrossRef]
  158. Yi, W.; Yan, D.; Wang, D.; Li, Y. Smart drug delivery systems to overcome drug resistance in cancer immunotherapy. Cancer Biol. Med. 2023, 20, 248–267. [Google Scholar] [CrossRef]
  159. Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef]
  160. Khalil, A.; Al-Massarani, G.; Aljapawe, A.; Ekhtiar, A.; Bakir, M.A. Resveratrol Modulates the Inflammatory Profile of Immune Responses and Circulating Endothelial Cells’ (CECs’) Population during Acute Whole Body Gamma Irradiation. Front. Pharmacol. 2020, 11, 528400. [Google Scholar] [CrossRef]
  161. Lafta, H.A.; AbdulHussein, A.H.; Al-Shalah, S.A.J.; Alnassar, Y.S.; Mohammed, N.M.; Akram, S.M.; Qasim, M.T.; Najafi, M. Tumor-associated Macrophages (TAMs) in Cancer Resistance; Modulation by Natural Products. Curr. Top. Med. Chem. 2023, 23, 1104–1122. [Google Scholar] [CrossRef]
  162. Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef]
  163. Kemper, C.; Benham, D.; Brothers, S.; Wahlestedt, C.; Volmar, C.H.; Bennett, D.; Hayward, M. Safety and pharmacokinetics of a highly bioavailable resveratrol preparation (JOTROL (TM)). AAPS Open 2022, 8, 11. [Google Scholar] [CrossRef] [PubMed]
  164. Detampel, P.; Beck, M.; Krähenbühl, S.; Huwyler, J. Drug interaction potential of resveratrol. Drug Metab. Rev. 2012, 44, 253–265. [Google Scholar] [CrossRef] [PubMed]
  165. Bertelli, A.A.; Giovannini, L.; Giannessi, D.; Migliori, M.; Bernini, W.; Fregoni, M.; Bertelli, A. Antiplatelet activity of synthetic and natural resveratrol in red wine. Int. J. Tissue React. 1995, 17, 1–3. [Google Scholar]
  166. Shen, M.Y.; Hsiao, G.; Liu, C.L.; Fong, T.H.; Lin, K.H.; Chou, D.S.; Sheu, J.R. Inhibitory mechanisms of resveratrol in platelet activation: Pivotal roles of p38 MAPK and NO/cyclic GMP. Br. J. Haematol. 2007, 139, 475–485. [Google Scholar] [CrossRef]
  167. Gehm, B.D.; McAndrews, J.M.; Chien, P.Y.; Jameson, J.L. Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc. Natl. Acad. Sci. USA 1997, 94, 14138–14143. [Google Scholar] [CrossRef]
  168. Andreani, C.; Bartolacci, C.; Wijnant, K.; Crinelli, R.; Bianchi, M.; Magnani, M.; Hysi, A.; Iezzi, M.; Amici, A.; Marchini, C. Resveratrol fuels HER2 and ERα-positive breast cancer behaving as proteasome inhibitor. Aging 2017, 9, 508–523. [Google Scholar] [CrossRef]
  169. Kandi, V.; Vadakedath, S. Clinical Trials and Clinical Research: A Comprehensive Review. Cureus 2023, 15, e35077. [Google Scholar] [CrossRef]
  170. Smoliga, J.M.; Blanchard, O. Enhancing the delivery of resveratrol in humans: If low bioavailability is the problem, what is the solution? Molecules 2014, 19, 17154–17172. [Google Scholar] [CrossRef]
  171. Boocock, D.J.; Faust, G.E.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J.; et al. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1246–1252. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of resveratrol (3,5,4′-trihydroxylstilbene).
Figure 1. Chemical structure of resveratrol (3,5,4′-trihydroxylstilbene).
Targets 02 00018 g001
Table 1. Summary of the anticancer mechanisms of resveratrol.
Table 1. Summary of the anticancer mechanisms of resveratrol.
MechanismDescriptionImplications for Cancer
Antioxidant ActivityScavenges ROS and RNS
Chelates iron and copper
Reduces oxidative stress
Prevents DNA, protein, and lipid damage
Modulation of Cellular SignalingRegulates cell proliferation, apoptosis, inflammation
Affects oxidative stress regulation
Inhibits cancer growth and metastasis
SIRT1 ActivationActivates SIRT1 deacetylase
Deacetylates PGC-1α and p53
Enhances mitochondrial function
Regulates apoptosis and reduces inflammation
PI3K/Akt InhibitionInhibits PI3K/Akt pathway
Reduces phosphorylation of Akt
Decreases cell proliferation
Promotes apoptosis
Antiproliferative EffectsInduces cell cycle arrest (G1, G2/M phases)
Modulates cyclins and CDKs
Halts cancer cell growth
Promotes programmed cell death
Anti-inflammatory EffectsInhibits NF-κB pathway
Reduces pro-inflammatory cytokines (TNF-α, IL-6)
Mitigates chronic inflammation
Reduces cancer-promoting inflammation
Epigenetic Modulation- Affects histone acetylation
- Alters DNA methylation and microRNA expression
- Regulates oncogene and tumor suppressor gene expression
- Enhances anticancer potential
Table 2. Effects of resveratrol against various types of cancer: in vivo studies.
Table 2. Effects of resveratrol against various types of cancer: in vivo studies.
Type of CancerAnimal ModelDoseOutcomeReferences
ColorectalF344 rats200 μg/kg/daySuppresses growth[87]
SCID 1 mice150 mg/kg/daySuppresses growth[88]
Wistar rats8 mg/kg/dayInhibits tumorigenesis[89]
Sprague–Dawley rats60 mg/kg/dayDecreases aberrant crypt foci and mucin depleted foci[90]
BreastBALB/c mice 21, 3, or 5 mg/kh/dayNo effect[91]
Sprague–Dawley rats10 or 100 mg/kg/5×/weekInhibition of carcinogen-induced preneoplastic lesions and mammary tumors[92]
HER2/neu mice4 μg/dayReduces the metastasizing[93]
SCID mice0.5, 5 or 50 mg/kg/5×/weekIncreases tumor growth[94]
Nude mice0.5, 5 or 50 mg/kg/5×/weekIncreases tumor growth[94]
LiverWistar rats20 mg/kg/dayInduces apoptosis[95]
HBx mice30 mg/kg/dayDelayed hepatocarcinogenesis[96]
C57BL/6J mice1 mg/kg/dayPrevention of inflammation-dependent melanoma metastasis[97]
Sprague–Dawley rats0, 100 or 300 mg/kg/dayPrevents chemically induced liver tumorigenesis[98]
LungNude mice15, 30, or 60 mg/kg/dayDecreases lung cancer growth in a dose-dependent manner[99]
C57B6 mice25 mg/kg/dayDecreases tumor volume, cell proliferation, tumor angiogenesis and liver metastatic lesions[100]
BALB/c nude mice1 g/kg/day or 3 g/kg/daySuppresses growth[101]
Nude mice50 mg/kg/daySuppresses growth[102]
C57B6 mice 100 mg/kg/dayDecreases F4/80+ macrophage[103]
ProstateNude mice50 mg/kg/daySuppresses growth[104]
TRAP rats0.005, 0.01, or 0.02%/daySuppresses growth[71]
TRAMP mice0.0625%/daySuppresses growth[105]
PancreaticNude mice20, 40, or 60 mg/kg/daySuppresses growth[106]
KrasG12D mice40 mg/kg/5×/weekInhibits pancreatic cancer stem cell[107]
Nude mice10 or 50 mg/kg/5×/weekSuppresses growth[108]
Nude mice40 mg/kg/dayPotentiate the effects of gemcitabine[109]
1 severe combined immunodeficiency; 2 Bagg Albino/c.
Table 3. Clinical trials involving resveratrol against types of cancer.
Table 3. Clinical trials involving resveratrol against types of cancer.
Type of CancerClinical TrialStatusStudy ID (https://clinicaltrials.gov) Accessed on 31 July 2024
ColorectalPhase 1Completed (2009)NCT00920803
Phase 1Completed (2009)NCT00433576
Phase 1Completed (2007)NCT00578396
Neuroendocrine TumorN.A *Completed (2018)NCT01476592
LiverPhase 1/2Completed (2016)NCT02261844
ColonPhase 1Completed (2009)NCT00256334
Multiple MyelomaPhase 2Completed (2010)NCT00920556
* Not applicable.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ribeiro, E.; Vale, N. The Role of Resveratrol in Cancer Management: From Monotherapy to Combination Regimens. Targets 2024, 2, 307-326. https://doi.org/10.3390/targets2040018

AMA Style

Ribeiro E, Vale N. The Role of Resveratrol in Cancer Management: From Monotherapy to Combination Regimens. Targets. 2024; 2(4):307-326. https://doi.org/10.3390/targets2040018

Chicago/Turabian Style

Ribeiro, Eduarda, and Nuno Vale. 2024. "The Role of Resveratrol in Cancer Management: From Monotherapy to Combination Regimens" Targets 2, no. 4: 307-326. https://doi.org/10.3390/targets2040018

APA Style

Ribeiro, E., & Vale, N. (2024). The Role of Resveratrol in Cancer Management: From Monotherapy to Combination Regimens. Targets, 2(4), 307-326. https://doi.org/10.3390/targets2040018

Article Metrics

Back to TopTop