Skip to main content
  • Systematic Review
  • Open access
  • Published:

Phytochemicals regulate cancer metabolism through modulation of the AMPK/PGC-1α signaling pathway

Abstract

Background

Due to the complex pathophysiological mechanisms involved in cancer progression and metastasis, current therapeutic approaches lack efficacy and have significant adverse effects. Therefore, it is essential to establish novel strategies for combating cancer. Phytochemicals, which possess multiple biological activities, such as antioxidant, anti-inflammatory, antimutagenic, immunomodulatory, antiproliferative, anti-angiogenesis, and antimetastatic properties, can regulate cancer progression and interfere in various stages of cancer development by suppressing various signaling pathways.

Methods

The current systematic and comprehensive review was conducted based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria, using electronic databases, including PubMed, Scopus, and Science Direct, until the end of December 2023. After excluding unrelated articles, 111 related articles were included in this systematic review.

Results

In this current review, the major signaling pathways of cancer metabolism are highlighted with the promising anticancer role of phytochemicals. This was through their ability to regulate the AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) signaling pathway. The AMPK/PGC-1α signaling pathway plays a crucial role in cancer cell metabolism via targeting energy homeostasis and mitochondria biogenesis, glucose oxidation, and fatty acid oxidation, thereby generating ATP for cell growth. As a result, targeting this signaling pathway may represent a novel approach to cancer treatment. Accordingly, alkaloids, phenolic compounds, terpene/terpenoids, and miscellaneous phytochemicals have been introduced as promising anticancer agents by regulating the AMPK/PGC-1α signaling pathway. Novel delivery systems of phytochemicals targeting the AMPK/PGC-1α pathway in combating cancer are also highlighted in this review.

Peer Review reports

Introduction

Cancer is one of the major causes of death worldwide. There are several manifestations of this deadly disease, such as lumps, unusual bleeding, protracted coughing, weight loss, and decreased appetite [1,2,3]. Factors effective in causing cancer include smoking, obesity, an unhealthy diet, excess alcohol consumption, lack of exercise, sickness, exposure to ionizing radiation and environmental pollutants, or infection with viruses, bacteria, and certain parasites. Cancer occurs due to neoplastic growth in an irregular manner, which often forms a mass of cancer cells [4,5,6]. Under physiologic conditions, complex signaling pathways are involved in the development of cancer. Oxidative stress, apoptosis, autophagy, and inflammation are the most important pathways that affect cancer, engaging pro-inflammatory cytokines and apoptotic mediators. Therefore, the control of regulatory mechanisms, such as the release of cytokines or chemokines, oxidative stress and the process of apoptosis, has a significant function in the management of cancer [7]. Studies have demonstrated that several signaling pathways have considerable function in cancer pathogenesis. These include the Ras/extracellular signal-regulated protein kinase (ERK), phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) pathways [8]. Several other dysregulated signaling pathways are responsible for cancer metabolism such as carbohydrate, lipid, and protein metabolism. An altered metabolism may advance the proliferation and survival of cancer cells [9].

Cancer treatments include a variety of approaches tailored to improve patient outcomes and effectively manage the disease, depending on the specific type and stage of cancer. These approaches include surgical intervention, chemotherapy, targeted therapy, radiation therapy, hormone therapy, and immunotherapy, as well as complementary and integrated therapies, such as herbal treatments. Healthcare professionals select treatment modalities based on the patient’s clinical status, medical history, and potential side effects. The appropriate and judicious combination of these modalities may lead to improved treatment outcomes and reduced adverse effects, ultimately contributing to an enhanced quality of life for the patient. Over the years, more than 300 chemotherapeutic drugs have been used in the treatment of cancer, such as taxol, vincrictine, cis-platin, 5-fluorouracil, bevacizumab, erlotinib, nivolumab, ipilimib, santin, and olaparib. However, these drugs are very toxic and may create resistance, which leads to tumor recurrence and metastasis [10]. Therefore, alternative treatment methods, such as the use of bioactive plant secondary metabolites (phytochemicals) should be considered [1, 11].

Phytochemicals, also known as plant secondary metabolites, have been the focus of extensive research in recent years due to their potential health benefits, including their anticancer properties. Phytochemicals may be obtained from various sources, including whole grains, fruits, vegetables, nuts, and spices [12]. Phytochemicals have remarkable chemical diversity and possesses various bioactivities, including, anti-inflammation, antioxidative, antimicrobial, antiviral and antiaging properties, and have been widely investigated for their anticancer potential [13]. Preclinical studies have indicated that phytochemicals are able to modulate various oncogenic and oncosuppressive cell signaling pathways, such as the PI3K/Akt/mTOR/P70S6K [14], peroxisome proliferator-activated receptor (PPARs) [15], nuclear factor erythroid 2-related factor 2 (Nrf2), Janus kinase (JAK)–signal transducer and activator of transcription (STAT) [16], hypoxia-inducible factor-1 (HIF-1), transforming growth factor-beta (TGF-β) [17], toll-like receptors (TLR)/nuclear factor-κB (NF-κB)/Nod-like receptor protein (NLRP), MAPK, ERK, and p38 signaling pathways [18,19,20]. Phytochemicals also target multiple dysregulated pathways of cancer metabolism, such as carbohydrate, lipid, and protein metabolism. This ability of phytochemicals to target multiple pathways involved in cancer development and progression is one of the key reasons why they may have potential in cancer treatment. Traditional cancer therapies often target specific molecular pathways, but phytochemicals simultaneously modulate multiple signaling pathways, potentially making them more effective in combating the complex nature of cancer. Phytochemicals not only have direct anticancer effects, but also influence the metabolic pathways within cancer cells. For instance, researchers have discovered that several phytochemicals hinder cancer cell growth by disrupting crucial metabolic processes, such as carbohydrate metabolism, lipid metabolism, and protein synthesis [21]. By disrupting these essential pathways, phytochemicals can potentially starve cancer cells of the nutrients they need to survive and proliferate.

The use of phytochemicals has unique advantages for medical applications, providing a novel approach to cancer prevention and improving patient outcomes and treatment efficacy, resulting in more effective treatment strategies. Additionally, studies have demonstrated that phytochemicals are a cost-effective option that mitigates the common adverse side effects of traditional treatments and decreases drug resistance, providing a safer and more patient-friendly alternative. These benefits are crucial in the field of medicine, where personalized and targeted therapies are essential for optimizing patient care and improving overall health outcomes. In this line novel analytical methodologies (e.g., Liquid chromatography-tandem mass spectrometry) is necessary to identify active phytochemicals of traditional medicine [22]. In addition, novel drug delivery systems will pave the road in combating different diseases [23, 24].

Numerous phytochemicals are being tested in vitro and in vivo experiments. Moreover, various clinical trials also utilize phytochemicals in combination with other approved drugs. These clinical trials involve the rigorous testing and evaluation of phytochemicals in real-world settings, providing valuable insights into the effectiveness and safety of plant ingredients. By integrating plant-based therapeutic appriaches with established treatments, it is possible to enhance the overall efficacy and outcomes of clinical trials, ultimately contributing to the advancement of medical science and patient care.

Cancer metabolism refers to the unique metabolic characteristics and alterations that occur in cancer cells when compared to normal cells. Cancer cells exhibit specific metabolic requirements and mechanisms to maintain their rapid growth and reproduction. Metabolic reprogramming has been established as the hallmark of cancer. Key aspects of cancer metabolism are heightened glucose absorption and glycolysis, regardless of oxygen availability (the Warburg effect), modified lipid processing, elevated glutamine utilization, and alterations in mitochondrial activity. These pathways provide cancer cells with a flexible metabolic characteristic and offer chances for survival for cancer cells under stress. Comprehending cancer metabolism is crucial to developing specific treatments that capitalize on the metabolic defects of cancer cells without damaging normal cells. Researchers are investigating the disrupted metabolic pathways in cancer cells to pinpoint possible drug targets and create novel therapeutic approaches. There are several signaling pathways that play a crucial role in regulating cancer cell metabolism. Some of the key signaling pathways involved in cancer cell metabolism include, PI3K/Akt/mTOR pathway, AMPK, HIF, p53, Wnt/β-catenin, and Nrf2 pathway. These signaling pathways interact with each other and with other cellular processes to coordinate the metabolic reprogramming that occurs in cancer cells. Targeting these pathways with specific inhibitors or modulators holds promise for developing novel therapeutic strategies to selectively target cancer metabolism.

As a critical signaling pathway, 5´-adenosine monophosphate-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), has recently been highlighted as a promising target in combating cancer [25]. The dysregulation of the AMPK/PGC-1α axis is associated with many types of malignancies and may affect tumor development, metastasis, and treatment outcomes. In cancer cells, alterations in AMPK and PGC-1α activity can lead to metabolic reprogramming that supports the high energy demands of rapidly proliferating cells. For example, AMPK activation regulates mTOR signaling and inhibits cell growth, whereas PGC-1α overexpression promotes mitochondrial function and accelerates oxidative metabolism. The modulation of the AMPK and PGC-1α signaling pathways by phytochemicals presents a novel approach to targeting cancer cells and disrupting their metabolic processes. By elucidating the complex processes and mechanisms by which phytochemicals interact with these keys signaling pathways, researchers aim to develop innovative strategies for combating cancer and improving patient outcomes.

The influence of phytochemicals on cancer cell metabolism via altering the AMPK/PGC-1α signaling pathway has not been thoroughly investigated in existing literature, and no previous reviews have been published in this area. The aim of this review is to explore the impact of phytochemicals on the regulation of cancer metabolism through modulation of the AMPK/PGC-1α signaling pathway. By examining the current literature and summarizing the findings from in vitro and in vivo studies, we aim to elucidate the molecular mechanisms underlying the anticancer effects of phytochemicals targeting this key metabolic pathway. Ultimately, this review aims to provide a systematic review of the therapeutic potential of phytochemicals in cancer treatment and highlight their role in modulating cellular metabolism for improved patient outcomes. It’s the first systematic review on the promising anticancer role of phytochemicals through the regulation of AMPK/PGC-1α signaling pathway. We have also summarized various novel drug delivery systems for phytochemicals targeting AMPK/PGC-1α pathway and combat cancer.

The core role of AMPK/PGC-1α signaling pathway in cancer metabolism

The AMPK, a type of enzyme called a serine/threonine kinase, has remained similar in various species through evolution. It has a significant function in controlling cellular energy and redox homeostasis and affects all aspects of energy metabolism and mitochondrial biogenesis [26]. The AMPK enzyme has a heterotrimeric structure consisting of three subunits: α, β, and γ. The α subunit plays a catalytic role, the β subunit has a scaffolding function, and the γ subunit is a regulatory component. There are at least 12 known isoforms of AMPK identified in various cells and tissues [27, 28]. The α subunit is present in both the cell membrane and nucleus [29]. The N-terminus of the β subunit binds to carbohydrates and inhibits AMPK signaling, affecting glycogen synthesis in the liver and muscle [28, 30]. The γ subunit connects to the β subunit through its N-terminus and helps activate the enzyme [31]. AMPK gets activated when adenosine triphosphate (ATP) production decreases due to factors like hypoxia, or low oxygen, or lack of nutrients [32]. This phosphorylation mediates by liver kinase B1 (LKB1), calcium/calmodulin-dependent protein kinase kinase-β (CaMKKβ), and protein phosphatase 2 C and 2 A (PP2C and PP2A) [33, 34]. However, evidence indicates that LKB1 has an important function in AMPK phosphorylation and activation in energetic stress conditions [33].

The activation of AMPK can cause inhibition of the anabolic pathway and stimulation of the catabolic pathway [34, 35]. Adenosine monophosphate (AMP) has a higher affinity for attachment to the γ subunit of AMPK than adenosine diphosphate (ADP) [33]. Furthermore, AMP, in contrast to ADP, can promote LKB1-induced AMPK phosphorylation [33]. In times of abundant energy, enzymes like TGF-β-activated kinase 1 (TAK1), PP2A, PP2C, and dependent protein phosphatase 1E (PPM1E) inactive AMPK through dephosphorylation [36].

Reactive oxygen species (ROS) can active AMPK signaling, through direct and indirect mechanisms. Direct activation takes place via S-glutathionylation of cysteines on the AMPK α and β subunits. While indirect activation is mediated by changes in the cellular ATP/AMP and ATP/ADP ratios [37]. Numerous studies indicate that the activation of AMPK could be a potential treatment for various cancer types by inhibiting cancer promoting metabolic processes, arresting the cell cycle, and acting as a cyclooxygenase-2 (COX-2) inhibitor, decreasing cancer stemness [38]. AMPK increases autophagy and mitophagy by activating UNC-51-like kinase 1 (ULK1) and death-associated protein 1 (DAP1), respectively.

AMPK starts the apoptotic program through the activation of p53, p21, p27 and retinoblastoma protein (pRb) arrests cell cycle through the inhibition of HUR and the concomitant activation of cyclin A, cyclin B1, and cyclin D1 [39,40,41]. AMPK inhibits metabolic pathways in tumor cells, such as the Warburg effect, depriving cancer cells of energy and fuels [42]. The activation of AMPK signaling results in the reduction of mammalian target of rapamycin complex 1 (mTORC1) via the phosphorylation of tuberous sclerosis complex 2, also known as tuberin (TSC2) and regulatory-associated protein of mTOR (RAPTOR) [43]. This process suppresses oncogenic signaling, thereby restricting the proliferation and migration of cancer cells [44]. Furthermore, AMPK activation induces autophagy, inhibits glycolysis, and promotes mitochondrial oxidative metabolism through mTORC inhibition [45]. AMPK phosphorylates and deactivates oncogenic yes-associated protein (YAP) through Hippo tumor suppressive signal during glucose deprivation [46]. Studies suggest that AMPK phosphorylates and counteracts the oncogenic activity of glioma-associated oncogene 1 (Gli-1), the central transcription factor that regulates cell proliferation and differentiation in the Hedgehog pathway, particularly in medulloblastoma [47]. The LKB1, also called serine/threonine kinase 11 (STK11), is a known activator of AMPK, is tumor suppressor that act via the inhibition of mTORC1, and its deactivation is indicated in various cancers, include lung adenocarcinoma [43].

In epigenetic reprogramming, AMPK affects DNA methylation by phosphorylating and activating the tumor suppressor TET methylcytosine dioxygenase 2 (TET2) at S99 [48]. Metformin, a biguanide antidiabetic drug, activates AMPK, leading to the phosphorylation of checkpoint blocker programmed cell death ligand 1 (PD-L1) at S195. This process causes PD-L1 degradation through endoplasmic reticulum-associated protein degradation (ERAD) and enhances antitumor immunity [49]. Metformin enhances the expression of NF-κB via the AMPK/SIRT1 pathway, ultimately resulting in cellular apoptosis [50]. AMPK-mediated phosphorylation of enhancer of zeste homolog 2 (EZH2) inhibits polycomb repressive complex 2 (PRC2) cancer-promoting function and associated with improved survival outcome in breast and ovarian cancer [51].

Numerous experimental evidence in rats supports the tumor-suppressor action of AMPK. First, the loss of AMPKβ1 in the prostate accelerates the onset of prostate adenocarcinoma [52]. Second, elevated ubiquitin conjugating enzyme E2 (UBE2O) expression in various cancers induced the degradation of AMPKα2, therefore increasing the development and spread of tumors [53]. Third, the loss of AMPKα1 in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) accelerates the formation of leukemia and lymphoma [54]. Fourth, the inactivation of AMPKα1 promotes MYC (a hallmark of tumorigenesis)-driven lymphomagenesis by inducing HIF-1α-dependent aerobic glycolysis [43]. Fifth, depleting AMPKβ1 accelerates the onset of T-cell lymphoma after the inactivation of the p53 gene [43]. Finally, MAGE-A3/6-TRIM28, a cancer-specific ubiquitin ligase, induces the ubiquitination and degradation of AMPK. This process promotes mTOR signaling and contributes to malignant transformation in lung, breast, and colon tissues [55].

There are conflicting perspectives on whether AMPK actions may function in cancer development. AMPK plays a role in metabolic adaptation by participating in the signaling pathways associated with metabolic stress in tumor microenvironment (TME), which include nutrient starvation, matrix detachment, oxidative stress, and hypoxia [34]. As a result, activating AMPK may enhance the resistance of tumor cells to metabolic stress and maintain ATP levels by reprogramming energy metabolism [25, 34]. One study indicates that in low glucose conditions, the AMPK-p38-PGC-1α pathway induces metabolic homeostasis in cancer cells [56]. AMPK provides the metabolic requirements for cancer cells that are growing during autophagy, thus promoting cell growth and survival. On the other hand, autophagy causes chemoresistance in cancer cells [25]. AMPK enhances mitochondrial respiration through MYC, resulting in efficient glutamine metabolism for energy production [34]. It has been reported that the effects of AMPK on tumor development may depend on the nutrient levels in the TME. In the absence of nutrients, AMPK may promote tumor growth and support survival, while in the presence of nutrients, AMPK may suppress tumor development [57]. AMPK phosphorylates and deactivates acetyl-CoA carboxylases 1/2 (ACC1/2) to maintain nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) homeostasis, thereby promoting cancer cell survival and breast cancer progression [58].

AMPK is involved in EGF/Akt signaling, a process disrupted in malignant transformation and cancer metastasis through the activation of Akt, leading to oncogenic procedure [59]. AMPK promotes T-ALL cells survive by inhibiting aerobic glycolysis, increasing mitochondrial oxidative metabolism, and decreasing metabolic stress and apoptosis [45]. During metabolic stress, AMPK activation causes resistance to oxidative stress and DNA damage in tumor cells in the bone marrow [60]. Studies indicated that the activation of AMPK-SKP2-Akt axis and AMPK-PDHc cascade is linked to a dismal prognosis in breast cancer [61]. Sirtuin 3 (SIRT3), a mitochondrial protein that removes acetyl groups, increases AMPK expression and leads to increased lymph node metastasis in cervical cancer cells. In colorectal cancer cells, non-muscle myosin IIA (NMIIA) triggers AMPK signaling, promoting the expression of mTOR, which increases growth and invasion [50]. Experimental evidence in rats supports the continuous function of AMPK in cancer advancement. In an acute myeloid leukemia (AML) model, AMPK inactivation decreased leukemia-initiating cells (LIC) and decelerated leukemogenesis [60]. In a lung cancer model, AMPK loss diminished tumor development, underscoring the essential role of AMPK activation in tumorigenesis [62].

One of the key regulators of cell metabolism is a member of the PGC-1 transcriptional co-activator family, known as PGC-1α. The PGC-1 family has three members: PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC) [63]. PGC-1α primarily regulates mitochondrial biogenesis (mitobiogenesis) and function, including oxidative phosphorylation (OXPHOS), fatty acid/lipid metabolism, and the regulation of ROS levels [64]. The activation of PGC-1α in brown adipose tissue and in skeletal muscle contributes to an increase in metabolic activity and heat production as a response to cold exposure, exercise, and fasting [63, 65]. The PGC-1α predominantly localizes within metabolically active tissues, including the liver, heart, muscles, kidneys, adipose tissue, and the brain [66]. PGC1α activity and mitochondrial function decline with age, potentially contributing to age-related cancer. However, exercise and a calorie-limit diet have been shown to enhance PGC1α activity, promoting healthy aging and potentially acting as protective factors against age-related cancer [64].

Numerous studies indicate the expression of PGC-1α is closely associated with cancer progression. PGC-1α has a function in maintaining metabolic homeostasis in microenvironments with high energy demands and restricted nutrition supplies in cancer cells. Overexpression of PGC-1α is identified in various type of cancers [67]. Similar to normal cells, PGC-1α mainly influences mitochondrial respiration, detoxification ROS, fatty acid oxidation (FAO), and glucose- or glutamine-derived lipogenesis in cancer cells [68, 69]. However, literature contains conflicting reports, as both increased and decreased levels of PGC1α expression were related to cancer and a worse prognosis. Even within a specific cancer, include breast cancer, there exist discrepancies in the reported PGC1α levels [64]. Melanoma cells that overexpress PGC-1α demonstrate elevated mitochondrial oxidative metabolism, effective detoxification of ROS, dependence on OXPHOS, and resistance to apoptosis and chemotherapy [63, 70, 71]. In contrast, melanoma cells expressing low levels of PGC-1α, depend on glycolysis to survive, making them more vulnerable to apoptosis induced by ROS [70]. Nevertheless, despite the enhanced proliferation and survival associated with PGC-1α overexpression, it concurrently suppresses the invasive properties of these cells [71]. Furthermore, in melanoma, BRAF and inhibition of mTORC1/2 inhibits the melanocyte lineage factor (MITF), which, in turn, downregulates PGC-1α and increases glycolytic metabolism [72].

Under metabolic stress, the switch from glucose to fatty acid usage helps cells survive and occurs in various cancer types. PGC-1α induces the transactivation of FAO genes via PPARα and sirtuin 1 (SIRT1) [67, 73, 74]. When cells are deprived of glucose, PGC1α breaks down, leading to the aggregation of ROS and apoptosis [75]. In nutrient deprivation conditions, p53 has both cytoprotective and cytotoxic functions, while PGC-1α regulates p53 stress-dependent transcription, enhancing its activation of genes for cell cycle arrest and metabolism [75, 76]. Receptor-interacting protein 1 (RIP1) regulates p53 via PGC-1α signaling. RIP1 inactivation reduces PGC-1α expression and OXPHOS, promoting glycolysis. Excessive glycolysis lowers nicotinamide adenine dinucleotide (NAD) levels, impairing DNA repair and activating p53-mediated cell growth inhibition [77]. MYC regulates glucose and glutamine metabolism, along with mitobiogenesis in cancer cells. C-MYC attaches to the promoter of PGC-1α and inhibits its transcription. The ratios of PGC-1α to MYC are associated with metabolic phenotypes in tumors, including pancreatic ductal adenocarcinoma, ranging from OXPHOS-based to glycolytic [78, 79].

Pancreatic cancer stem cells express high levels of PGC-1α due to the absence of c-MYC. The strong expression of PGC-1α is implicated in mitochondrial respiration, as well as sensitivity to metformin treatment [78]. This ratio can be controlled by the transcription factor Forkhead box O3a (FOXO3a). Similar to PGC-1α, FoxO3a expression levels are correlated with cancer and adverse outcomes at both high and low levels [64, 80]. Estrogen-related receptor (ERR) and PGC-1α are downstream proteins of kinase suppressor of Ras 1 (KSR1), a molecule that promotes Ras-induced transformation in breast cancer [81]. PGC-1α can mimic the actions of the natural ligands that ERR typically binds to, even though ERRα itself doesn’t bind to estrogens. Similar to PGC-1α, ERRα functions in the quick response to metabolic stress [64, 82]. The PGC-1α/ERRα axis can influence glucose, glutamate, and fatty acid metabolism, as well as the TCA cycle, thereby stimulating the proliferation of breast cancer cells, even in conditions of low nutrients or hypoxia. Furthermore, in breast tumors, the PGC-1α/ERRα axis is involved in angiogenesis, metastasis, and resistance to chemotherapy [64, 83]. High level of PGC-1α in mammary tumor cells induces dependence on the folate cycle for nucleotide synthesis and tumor proliferation [84]. Studies indicate that increased levels of PGC-1α, and its associated glutaminolysis genes, forecast a poor prognosis in breast cancer and demonstrate a negative association between the expression of PGC-1α and patient survival [83].

Evidence indicates a mutual influence and activation between androgen receptor (AR) and PGC-1α in prostate cancer. The androgen/AR/AMPK/PGC-1α signaling axis promotes mitochondria biogenesis, glucose oxidation, and FAO, which provides structural units and ATP for cancer cell growth [67, 85]. PGC-1α-mediated tumor suppression primarily occurs through the induction of cell death. In addition, PGC-1α plays a role in tumor suppression by enhancing antioxidant defense via the upregulation of enzymes, including manganese-dependent superoxide dismutase and Nrf2, which can help prevent oxidative DNA damage and the development of cancer [64, 86]. The function of PGC-1α in advanced tumor stages is hypothesized to be shaped by both the microenvironment and the metabolic conditions of the tumor [87]. Evidence indicates that PGC-1α has a function in promoting and inhibiting tumor progression, depending on the microenvironment and the metabolic conditions of the tumor [64]. For instance, one study demonstrated that in melanoma, treatments that inhibit BRAF upregulate PGC-1α and ID-2, its downstream target, leading to the suppression of metastasis-related genes [71]. In contrast, evidence suggests that the downstream effectors of PGC-1α, specifically β-oxidation and fatty acid/lipid metabolism, play an essential role in promoting metastasis [88]. Furthermore, chemoresistant metastatic cells exhibit heightened metabolic patterns involving both glycolysis and increased OXPHOS [64]. Expression of PGC is upregulated by AMPK, ERRα, and p53. In contrast, PGC is downregulated through hypermethylation.

To fully understand the mechanisms underlying PPARGC1A methylation and its possible therapeutic implications, more research is necessary, as the precise significance of this alteration in cancer is currently unclear [64, 67, 89, 90]. Glycogen synthase kinase-3β (GSK-3β) phosphorylates the PGC-1α protein, which is then degraded via the ubiquitin-proteasome pathway. The breakdown of the PGC process is inhibited by the nuclear protein necdin, a tumor suppressor, thus preserving OXPHOS integrity [64, 91]. Inflammatory cytokines, including TGF-β, tumor necrosis factor-α (TNF-α), interleukin- 6 (IL-6), and TNF-related weak inducer of apoptosis (TWEAK), suppress the expression of PGC1α [64, 92, 93]. The E3 ligase Parkin orchestrates the elimination of dysfunctional mitochondria via mitophagy. Deactivating mutations of Parkin result in the accumulation of ZNF746, a transcriptional repressor of PGC-1α. Numerous studies suggest the presence of Parkin deletions in various types of cancers [64, 94].

MicroRNAs (miRNAs or miR)-485, miR-485-3p, and miR-5p, along with miR-23a and miR-217, downregulate PGC-1α. However, these miRNAs are inhibited in certain tumors [95,96,97]. Finally, in various types of carcinomas, the expression of MYC is upregulated, subsequently inhibiting PGC1 through acetylation by general control non-depressible 5 (GCN5) enzyme. This finding reinforces the inverse relationship between MYC and PGC-1α [64]. Autophagy exhibits tumor-suppressive functions during early tumorigenesis, but it may support the survival of cancer cells in established tumors [64]. Mitophagy has an important function in normal tissue function but is implicated in tumor cell resistance to stress, hypoxia, and DNA damage [98, 99]. Recent studies indicated that melanoma has two types of cells, one with high levels of PGC-1α and the other with very low levels of PGC-1α. In breast cancer, PGC-1α activates PPARα, ERRα, Nrf1, and Nrf2, which leads to development of mitochondrial biogenesis and OXPHOS and can generate large amounts of ATP for tumor growth [100]. The function of PGC-1α in prostate cancer is similar to that of melanoma. It has been demonstrated that c-MYC directly regulates PGC-1α in pancreatic adenocarcinoma. C-MYC binds to the PGC-1α promoter and inhibits its transcription. Further, the ratio of c-MYC/PGC-1α controls the metabolic behavior of pancreatic cancer cells [63].

Methodology of literature search

Based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria, the current systematic and comprehensive review was conducted. Thus, a systematic literature search was performed through electronic databases, including PubMed, Scopus, and Science Direct for articles written in the English language. The last search was performed at the end of December 2023. The systematic search in databases was carried out by applying the following keywords: (tumor OR cancer OR malignant* OR neoplasm OR melanoma OR leukemia) found in title or abstract AND (herb* OR plant* OR natural product OR polyphenol* OR phenolic compound* OR terpene* OR alkaloid* OR flavonoid* OR glucosinolate* OR coumarin*) AND (AMPK OR PGC-1alpha OR PGC-1α) were found in title/abstract. The search process was executed by two independent researchers (S.F. and S.Z.M.). Accordingly, the review process was finalized through discussion with a senior author (A.B.) to resolve disagreements. Initially, 265 articles of the 717 articles, which were obtained from the primary electronic search, were excluded due to duplication. Furthermore, 101 review articles were excluded. To evaluate the results based on the title and full text of the articles, 177 and 61 unrelated articles were exempted, respectively. Finally, after excluding unrelated articles, 111 related articles were included in this systematic review (Fig. 1).

Fig. 1
figure 1

PRISMA flowchart on the process of literature search and selection of relevant studies

Anticancer phytochemicals targeting AMPK/PGC-1α signaling pathway

Several recent studies have highlighted the modulatory roles of various phytochemicals against AMPK/PGC-1α. Accordingly, alkaloids, phenolic compounds, terpenes/terpenoids, and several miscellaneous compounds have shown potential in the modulation of AMPK/PGC-1α pathway to combat cancer.

Phenolic compounds

The most widespread type of secondary metabolite in plants are phenolic compounds. Phenolic compounds are identified by an aromatic ring with a characteristic hydroxylated structure. There are two main types of phenolic compounds: non-flavonoids and flavonoids. Non-flavonoids include stilbenes, phenolic acids, tannins, coumarins, and lignans [101,102,103,104]. Flavonoids include flavanols, flavonols, flavones, flavanones, anthocyanidins, and isoflavonoids. Phenolic compounds display a variety of advantageous traits, such as antibacterial, anticancer, anti-inflammatory, cardioprotective, antiviral, and antimutagenic effects [101,102,103,104]. The most notable phenolic substances with anticancer properties are the subject of research presented in the following paragraphs, emphasizing the pathways linked to cancer metabolism, especially AMPK/PGC-1α, that underlie their anticancer action.

Resveratrol

Resveratrol (Fig. 2), an extensively researched polyphenol, possesses a broad range of pharmacological properties, including anticarcinogenic, antibacterial, antioxidant, and anti-inflammatory effects [105,106,107,108]. Resveratrol induced apoptosis and inhibited the migration, proliferation, and invasion of in vitro and in vivo models of ovarian [109], breast [110], leukemia [111], colon [112, 113], prostate [114], and glioblastoma [115] cell lines via inhibiting glycolysis and fatty acid synthase. Resveratrol also reversed the Warburg effect and downregulating acetyl-CoA carboxylase-α via targeting pyruvate dehydrogenase, AMPK/mTOR, STIM1, NF-κB, AMPK-YAP, and c-Jun NH2-terminal kinase (JNK)-mediated p62/SQSTM1 signaling pathways.

Fig. 2
figure 2

Chemical structures of common phenolic compounds in the regulation of cancer metabolism AMPK/PGC-1α

Quercetin

Quercetin (Fig. 2) is a naturally occurring flavonoid that is prevalent in fruits and vegetables and mechanistically displays antioxidant, anti-inflammatory, and anticancer properties in a variety of cellular and animal models. It has been shown to have biological actions that include anticancer, anti-inflammatory, immunoprotective, and antiviral activities [116,117,118,119]. Quercetin induced apoptosis and autophagy and suppressed viability, migration, and proliferation of various human cancer cell lines, such as lung [120], cervical [121], colon [122, 123], and breast [124] cancer. This was due to quercetin’s ability to interfere with AMPK, epidermal growth factor receptor (EGFR), Akt/AMPK/mTOR, and SIRT1/AMPK signaling pathways.

Isoquercitrin

It has been reported that isoquercitrin (Fig. 2), a naturally occurring flavonoid found in various plant species, including Mangifera indica and Rheum nobile, significantly inhibited the proliferation of human liver cancer cells Huh7 and HepG2 [7, 125]. This was achieved through the inhibition of viability and colony growth, activation of the apoptotic pathway, and dysregulation of autophagy via the activation of the AMPK/mTOR/p70S6K pathway [126].

Curcumin

Curcumin (Fig. 2) is a phenol compound that can be extracted from Curcuma longa L. (Zingiberaceae family). Curcumin possesses significant biological activities, including antioxidant, anti-inflammatory, antimicrobial, neuroprotective, and anticancer activities [127,128,129,130]. It has been reported that curcumin inhibits growth, angiogenesis, and metastasis of 4T1, B16, CT26, A204, RD, SJCRH30, and SMMC-7721 cell lines by inducing apoptosis, fatal energetic impairment, and cell cycle arrest, mainly through the inhibition of NF-κB, suppression of ATP-synthase activity, and the activation of AMPK [131,132,133].

Epigallocatechin gallate

Epigallocatechin gallate (EGCG) (Fig. 2) is a phenolic compound derived from green tea. It is well-known for its antioxidant, anti-inflammatory, neuroprotective, and antineoplastic properties [134,135,136,137,138]. EGCG exerted significant anticarcinogenic activity in H1299 lung cancer cells in vitro via targeting AMPK, mTOR, and Akt signaling [139]. Moreover, treatment with EGCG leads to apoptosis and suppression of the proliferation of HT-29 cells via interfering with COX-2, AMPK, vascular endothelial growth factor (VEGF), and Glut1-related pathways. In addition, treatment decreased the formation and synthesis of fatty acid, lipid droplets formation, energy metabolism, mitochondrial oxidation/glycolysis, lipolysis, and fatty acid β-oxidation was reported as the main anticancer activities of EGCG in both HCT-116 and HT-29 cancer cells [140, 141].

Apigenin

Apigenin (Fig. 2), a 4′,5,7-trihydroxyflavone, is predominantly found in plants and belongs to the Apium genus, including parsley and Chinese celery. This bioactive molecule exhibits a wide range of biological activities, including antiviral, anti-inflammatory, and antioxidant, and anticancer properties [128,129,130, 142]. Apigenin promoted autophagy and apoptosis in the H1975 cell line via targeting HIF-1α, c-MYC, glucose metabolism, Glut1, Glut4, monocarboxylate transporter 1 (MCT1), and AMPK [143]. Moreover, apigenin exert significant in vitro and in vivo anti-carcinoma activities in gastric cancer models by interfering with AMPK, ULK1, mTOR, p62, protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), and HIF-1α [144].

Isoliquiritigenin

Isoliquiritigenin (Fig. 2), a chalcone compound from the natural source locorice, has exhibited noteworthy biological activities, such as antitumor, antiviral, anti-inflammatory, antispasmodic, antidiabetic, and antioxidant effects [145, 146]. Treatment with isoliquiritigenin led to the inhibition of growth of colorectal cancer cells via regulating AMPK/mTOR-mediated glycolysis and HIF-1α signaling. Similarly, isoliquiritigenin promoted the suppression of Glut4-mediated glucose uptake by targeting PDHK1/PGC-1α in gastric cancer cells [147, 148].

Hispidulin

Hispidulin (Fig. 2) is a naturally occurring flavone isolated from the Artemisa and Salvia plants, which are widely accepted as traditional medicinal plants. Hispidulin possesses various biological activities, including antioxidant, antifungal, neuroprotective, and antiproliferative properties [105, 106, 149]. Hispidulin suppressed the growth, proliferation, and metastasis of hepatocellular carcinoma cells by targeting ERK, AMPK, and AMPK/mTOR signaling pathways [150].

Rottlerin

Rottlerin (Fig. 2), a phytoconstituent with vital biological properties, such as anticancer, antibacterial, antifilarial, and anti-inflammatory activities, is found in the pericarp of Mallotus philippensis. Its multifaceted pharmacological effects against cancer are promising. Rottlerin promoted apoptosis and autophagy in prostate cancer cells by interfering with the AMPK and PI3K/Akt/mTOR signaling pathways [151]. Moreover, suppression of the Wnt/β-catenin and mTORC1 signaling pathways were noted to be the main antitumor effects of rottlerin in several in vitro models of breast and prostate cancer cells [152].

Baicalin and baicalein

Baicalin (Fig. 2) is a major bioactive glycosyloxyflavone that can be isolated from root of the Scutellaria baicalensis plant [153]. Baicalin has anticancer, hepatoprotective, anti-inflammatory, neuroprotective, cardioprotective, antioxidant, renal protective, and antibacterial advantages [154,155,156]. Baicalin exerts significant anticancer activity against several in vitro models of cancer, including non-small cell lung cancer (NSCLC) via regulating AMPK/Nrf2 and activating SIRT1/AMPK signaling [157, 158]. Baicalin can hydrolyzed to its metabolite and aglycone form, baicalein (Fig. 2), which has garnered significant attention from cosmetic, food, and pharmaceutical industries for its exceptional antioxidant, neuroprotective, anti-inflammatory, cardioprotective, anticancer, hepatoprotective, and antiviral properties [154,155,156]. Baicalein suppresses the growth of PC-3 and DU145 cells in in vitro models of prostate cancer by activating AMPK/ULK1 and inhibiting mTORC1 signaling [159]. In a similar study, baicalein promoted apoptosis and autophagy in glioblastoma cells (U251) through activation of AMPK [160]. Moreover, baicalein showed meaningful anticarcinoma activities in H1299, A549, PC9, and H1650 cell lines as in vitro models of NSCLC via facilitation of apoptosis and increasing p-ERK1/2, FOXO3a, and RUNX3 [161].

Other phenolic compounds

Kaempfero, (Fig. 2, a natural flavonol found in plants and plant-derived foods) [162], luteolin (Fig. 2, a flavonoid that is known for its antioxidant and anti-inflammatory properties) [163], morusin (Fig. 2, prenylated flavonoid produced from Morus alba Linn) [164], glycycoumarin (Fig. 2, a major coumarin in licorice) [165], and cyanidin 3-O-glucoside (Fig. 2, an anthocyanidin glycoside found in legumes) [166], are some of the known polyphenolic compounds that inhibit growth and glycolysis in various in vitro and in vivo models of hepatic carcinoma. This was achieved by inducing apoptosis, senescence, and cell cycle arrest through targeting AMPK, NF-κB, Akt, and CDK1/cyclin B. In addition, interfering with AMPK, HIF-1α, AMPK-mTOR, Sirt3/HIF-1α, PI3K/Akt/mTOR, and CaMKKβ-AMPK-mTOR signaling pathways was identified as the primary anticarcinogenic activity of ellagic acid (Fig. 2, a bioactive polyphenolic agent found Punica granatum L.) [167], honokiol (Fig. 2, a lignan polyphenol found in several Magnolia species) [168], gallotannin (Fig. 2, a specific type of hydrolyzable tannin present in vegetables) [169], and pomiferin (Fig. 2, a bioactive prenylated flavonoid isolated from Derris montana, Citrus aurantium) [170] in different in vitro and in vivo lung cancer models. Furthermore, multiple preclinical models of colon carcinoma illustrated the capability of other phenolic compounds to promote apoptosis, inhibit epithelial-mesenchymal transition (EMT), invasion, and growth, and induce cell cycle arrest by targeting several signaling pathways, including AMPK/mTOR, AMPK/MAPK/XAF1, TGF-β1, NF-κB, CaMKKβ-AMPK, and JAK2/STAT3 signaling. Of those phenolics, such as ampelopsin (Fig. 2, a flavanonol flavonoid known as dihydromyricetin) [171], isoangustone A (Fig. 2, a flavonoid obtained from Glycyrrhiza glabra) [172], brosimone I (Fig. 2, a flavonoid isolated from jackfruit) [173], and salidroside (Fig. 2, a tyrosol glucoside isolated from Rhodiola rosea) [174], have exerted promising antineoplastic potential by targeting AMPK signaling. Moreover, in PANC-1 and MIA-PaCa2 pancreatic cancer cells, fisetin (Fig. 2, a flavonoid found in several fruits and vegetables) [175], eupatilin (Fig. 2, a flavonoid derived from Artemisia asiatica) [176], and isoorientin (Fig. 2, a flavone C-glycoside constituent) [177] interfered with AMPK/mTOR, VEGF, and AMPK. Also, isorhamnetin (Fig. 2, a monomethoxyflavonol extracted from leaf of Ginkgo biloba) [178, 179], hyperoside (Fig. 2, a flavonol glycoside present in genera Hypericum and Crataegus) [180], silibinin (Fig. 2, a bioactive compound derived from Silybum marianum L.) [181], and typhaneoside (Fig. 2, a phenolic component isolated from Typha angustifolia L.) [182] are some of the known polyphenolic compounds with variant biological effects that showed significant anticarcinoma potential against in vitro and in vivo models of breast, skin, renal, melanoma, and leukemia cancers by interfering with several metabolism-related pathways, including AMPK/mTOR/p70S6K, PI3K, and Akt signaling pathways (Table 1).

Table 1 Phenolic compounds targeting AMPK/PGC-1α in cancer

Alkaloids

Alkaloids are naturally occurring organic nitrogen compounds found in various organisms, particularly plants, and are hypothesized to have substantial pharmacological and biological activities. These activities include neuroprotective, antibacterial, antifungal, and antitumor properties [129, 153, 183, 184]. The following section presents information on how various alkaloids target the AMPK/PGC-1α signaling pathway, which ultimately regulates cancer metabolism.

Berberine

Berberine (Fig. 3) is a phytocompound extracted from Berberis vulgaris and Berberis aristata plants. It is classified as an isoquinoline alkaloid and has a variety of pharmacological properties, including anti-inflammatory, immunomodulatory, antidepressant, and antineoplastic effects [185,186,187]. Berberine led to the suppression of growth, migration, and invasion of a colorectal cancer cell line via suppressing fatty acid synthesis and downregulating the activities of AMPK, NF-κB, and integrin β1 signaling [188,189,190]. Similarly, berberine attenuated the growth and proliferation of U87MG [191], HepG2 [192], PANC-1 [193], B16F10 [194], and AGS [195] cells in in vitro models of glioblastoma, hepatocellular, pancreatic, melanoma, and gastric cancer, respectively. This was done by inhibiting DNA synthesis, AMPK/mTOR/ULK1, mTORC1, COX-2, ERK, and the AMPK/HNF4α/WNT5A pathway.

Fig. 3
figure 3

Chemical structures of common alkaloids in the regulation of cancer metabolism through AMPK/PGC-1α

Chaetocochin J and neferine

Chaetocochin J (Fig. 3) and neferine (Fig. 3) are natural alkaloids with significant antimetastatic, antiproliferative, and neuroprotective properties. These alkaloids play a crucial role against HCT-116 and SW480 in vitro colorectal cancer cell models via interfering with Ulk-1-PERK, AMPK, AMPK-mTOR, and PI3K/Akt/mTOR pathways [196].

Stachydrine

Targeting the LIF/AMPK and PI3K/Akt/mTOR axis was reported to be the main anticarcinoma activity of stachydrine (Fig. 3, an active constituent obtained from of Castanea sativa Mill.) and coptisine (Fig. 3, an isoquinoline alkaloid present in Chinese goldthread) in several in vitro models of hepatocellular carcinoma [197, 198].

Other alkaloids

Fangchinoline (Fig. 3), a miscellaneous alkaloid extracted from Stephania tetrandra, exerted anticancer effects on lung and colorectal cancer via promoting apoptosis, autophagy, suppression of metastasis, and EMT through regulation of the AMPK/mTOR/ULK1 and Akt-mTOR pathways [199, 200]. Aloperine (Fig. 3, a quinolizidine alkaloid isolated from Sophora alopecuroides L.) [201], hydroxycamptothecin (Fig. 3, an active ingredient found in Nothapodytes nimmoniana) [202], hernandezine (Fig. 3, a bisbenzylisoquinoline alkaloid derived from Thalictrum glandulosissimum) [203], ethoxysanguinarine (Fig. 3, a benzophenanthridine alkaloid obtained from Macleaya cordata) [204], cryptolepine (Fig. 3, an alkaloid that can be found in Cryptolepis sanguinolenta) [205], angustoline (Fig. 3, an active ingredient of Camptotheca acuminata) [206], and 11-methoxytabersonine (Fig. 3, an active ingredient isolated from M. cochinchinensis) [207], are some of the valuable alkaloids found in several vegetables and fruits. They have been shown to possess crucial antimetastatic potential against various in vitro and in vivo cancer models, including thyroid, bladder, pancreatic, breast, melanoma, esophageal, and lung cancers. They do so by targeting AMPK, Akt, AMPK-mTOR-ULK1, ERK, ROS/AMPK, AMPKα1/2-LKB1, JNK, LKB1/AMPK/ELAVL1/LPACT2, and AMPK/mTOR signaling pathways (Table 2).

Table 2 Alkaloids targeting AMPK/PGC-1α in cancer

Terpenes/terpenoids

Terpenes, a hydrocarbon, and terpenoids, which are terpenoids that contain oxygen, are potent organic compounds obtained from plants that provide a wide variety of therapeutic benefits. Terpenes and terpenoids consist of a structure made of isoprene units, which are five-carbon unit components. The bonds between various isoprene units form the main structure of these compounds. Terpenes and terpenoids possess significant anticancer activities against variant types of carcinomas via interaction with apoptosis, autophagy, and metabolism-related signaling pathways [208,209,210]. In the following section, we provide an overview of research carried out on the most notable terpenes and terpenoid substances that possess substantial anticancer properties via modulation of the AMPK/PGC-1α signaling pathway.

Furanodiene and β-elemene

Furanodiene (Fig. 4) and β-elemene (Fig. 4) are two bioactive sesquiterpenes that exert significant anticancer activity against MCF-7, breast cancer [211], and A549, lung cancer [212], cells by alteration of mitochondrial function, suppression of DNA methyltransferase 1 expression, and activation of the AMPKα and ERK1/2 signaling pathways.

Fig. 4
figure 4

Chemical structures of common terpenes/terpenoids in the regulation of cancer metabolism through AMPK/PGC-1α

Ursolic acid

Ursolic acid (Fig. 4) is a naturally occurring pentacyclic triterpenoid carboxylic acid that is present in many fruits and vegetables, such as oregano, apples, peppermint, cranberries, lavender, bilberries, and elder flower. Ursolic acid possesses a wide spectrum of pharmacological activities such as antibacterial, cardioprotective, hepatoprotective, anti-inflammatory, and antiproliferative effects [213]. It can inhibit tumor growth and is increasingly being recognized as a promising molecule for both preventing and treating cancer. Ursolic acid suppresses the growth of hepatocellular carcinoma cells through the alteration of the glycolytic pathway, AMPKα, and pERK1/2 signaling pathways [214]. Moreover, treatment with ursolic acid leads to the induction of apoptosis and the suppression of the proliferation in MCF-7 and MDA-MB-231 cells by interfering with Akt, ERK, ROS, and AMPK pathways [215].

Triptolide

Triptolide (Fig. 4) is a primary active ingredient isolated from Tripterygium wilfordii Hook F. with notable anticancer potential. This phytocompounds can promote apoptosis and autophagy in lung and prostate cancer cells via activation of the CaMKKβ-AMPK and regulation of AMPK/mTORC2 signaling pathways [216, 217].

Yuanhuacine

Yuanhuacine (Fig. 4) is another diterpene terpenoid that showed antitumor activity in the A549 cell line via regulating AMPK/mTOR axis [218].

Other terpenoids

Nummularic acid (Fig. 4, a triterpenoid derived from the Fraxinus xanthoxyloides) [219], celastrol (Fig. 4, an active ingredients isolated from Tripterygium wilfordii) [220, 221], gitogenin (Fig. 4, a natural component extracted from Allium rotundum and Yucca gloriosa) [222], oleanolic acid (Fig. 4, a pentacyclic triterpenoid derived from Phytolacca americana) [223], poricoic acid A (Fig. 4, a tricyclic triterpenoid isolated from Poria cocos) [224], and plectranthoic acid (Fig. 4, a pentacyclic triterpenoid in Ficus microcarpa) [225] are some of the other triterpenoid components that possess considerable anticarcinoma activities in several in vitro and in vivo models of prostate, colorectal, lung, leukemia, and breast cancer via inducing cycle arrest and apoptosis and interfering with numerous signaling pathways, including mTOR/S6K, AMPK/mTOR, and AMPK signaling (Table 3).

Table 3 Terpenes/terpenoids targeting AMPK/PGC-1α in cancer

Miscellaneous phytochemicals

Research on the most important miscellaneous phytocompounds with notable anticancer activities are summarized in the following section, with an emphasis on the cancer metabolic pathways that underlie their anticancer action. Osthole (Fig. 5) is a natural coumarin agent with several pharmacological activities that can be isolated from Cnidium spp. Osthole suppressed the growth of various malignant phenotypes by promoting ferroptosis and apoptosis and suppressing glycolysis, AMPK/Akt, and the GSK-3β/AMPK/mTOR signaling pathway [226,227,228]. Targeting of AMPK/miR-299-5p/ATF2, AMPK, E-cadherin/AMPK/mTOR, and ROS/AMPK/mTOR axes was described as the major anticancer mechanism of numerous miscellaneous phytocompounds, such as 6′-O-galloylpaeoniflorin (Fig. 5, an active ingredients extracted from paeoniflorin) [229], gambogic acid (Fig. 5, a xanthonoid compound isolated from brownish) [230], gracillin (Fig. 5, a steroidal saponin that can be extracted from Dracaena draco) [231], aspiletrein A (Fig. 5, a steroidal saponin found in Aspidistra letreae) [232], and schizandrin A (Fig. 5, a dibenzocyclooctadiene lignan) [233], against several in vitro and in vivo models of lung carcinoma. In addition, ginkgolic acid (Fig. 5, an active ingredient in Ginkgo biloba L.) and periplocin (Fig. 5, a plant-derived glycoside) diminished the growth and development of PANC-1 and BxPC-3 cell lines in vitro models of pancreatic cancer by suppressing lipogenesis, promoting apoptosis, and activating AMPK-mTOR signaling [234, 235]. In similar studies, panduratin A (Fig. 5, a major active ingredient isolated from Boesenbergia rotunda) [236], bixin (Fig. 5, an apocarotenoid extracted from Bixa orellana) [237], β-sitosterol (Fig. 5, an active phytosterol) [238], physciosporin (Fig. 5, a natural constituent obtained from Pseudocyphellaria faveolata) [239], hydroxycitric acid (Fig. 5, a derivative of citric acid in Garcinia cambogia) [240], and isogambogenic acid (Fig. 5, an active component from Garcinia hanburyi) [241], showed remarkable antineoplastic effects against numerous in vitro models of cancer, including melanoma, glioma, colorectal, breast, leukemia, and gastric adenocarcinoma, by diminishing metastasis, mitochondrial respiration, aerobic glycolysis, EMT, cancer metabolism, and cell proliferation by disrupting AMPK/PGC-1α, mTOR, and AMPK and interconnected signaling pathways. Table 4 presents miscellaneous phytochemicals targeting AMPK/PGC-1α in cancer.

Fig. 5
figure 5

Chemical structures of miscellaneous phytochemicals in the regulation of cancer metabolism through AMPK/PGC-1α

Table 4 Miscellaneous phytochemicals targeting AMPK/PGC-1α in cancer

Novel delivery systems of phytochemicals in cancer

Novel delivery systems and formulations of phytochemicals play a crucial role in enhancing the bioavailability, stability, and efficacy of these natural compounds. Phytochemicals obtained from plants have favorable health effects such as anti-inflammatory, antioxidant, and anticancer characteristics. However, their poor solubility, low stability, and limited absorption in the body can hinder their therapeutic potential. By developing innovative delivery systems, such as nanoparticles, liposomes, micelles, and nanoemulsions, researchers can overcome these challenges and improve the targeted delivery of phytochemicals to specific tissues or cells [242, 243]. This progress improves the therapeutic impact of phytochemicals and introduces new opportunities for personalized medicine and disease therapy.

The effectiveness of many phytochemicals is limited due to rapid metabolism, low bioavailability, poor water solubility, and systemic elimination. To address these challenges, scientists have investigated novel drug delivery systems, including lipid-based nanoparticles, polymeric nanoparticles, micelles, exosomes, nanogels, and mesoporous silica nanoparticles [130, 244,245,246]. In the following section, novel delivery systems of phytochemicals, with their anticancer potentials, are reviewed.

Curcumin

Liposomal curcumin has demonstrated potential in cancer treatment through in vitro, in vivo, and clinical studies. Other novel delivery systems for curcumin against cancer include graphene oxide, poly(glycerol subcategory) nanoparticles, and nanodelivery systems, such as polymer nanoparticles, solid lipid nanoparticles, nanostructured lipid carriers, liposomes, niosomes, and nanoemulsions. These delivery systems have enhanced the therapeutic effectiveness of curcumin against various types of cancer, primarily because of its antiproliferative and proapoptotic effects on tumor cells. However, potentially serious side effects, including interactions with other drugs and the toxic aspects of nanoparticles, may occur. More high-quality studies are needed to determine clinical efficacy [247,248,249,250]. The potential of curcumin-loaded nanoparticles to reduce oxidative stress and apoptosis in various conditions, including cancer, through the AMPK pathway, has been analyzed. A study on curcumin nanoparticles demonstrated their ability to reduce palmitate-induced oxidative stress in the heart and protect it from apoptosis by activating the AMPK pathway [251]. Curcumin phytosomes combine curcumin with phospholipids to enhance this phytochemical’s bioavailability. Curcumin has been extensively studied for its potential health benefits, including its effects in fighting cancer. Research has shown that phytosome curcumin can inhibit thrombin-induced cell growth and migration through AMPK in breast cancer [252]. Curcumin’s phytosome formulation, such as Meriva® Phytosome® technology, enhanced the bioavailability of curcuminoids and ensured optimal absorption in the body [253]. Phytosome curcumin has demonstrated potential effects on cancer, especially in relation to breast cancer. Research has shown that phytosome curcumin can inhibit thrombin-induced cell growth and migration through AMPK in breast cancer [252].

EGCG

Several nanodelivery systems have been developed to enhance the effectiveness of EGCG in cancer treatment. It has been shown that liposomal co-delivery systems, including EGCG and paclitaxel, induce apoptosis in cancer cells more effectively than either of these compounds alone [254]. Gold and lipid-based nanoparticles have also been investigated as delivery systems for EGCG [255, 256].

A recent study demonstrated that delivering EGCG in nanoparticles increased cytotoxicity in breast cancer cell lines. Furthermore, a co-delivery nanosystem of EGCG and rutin has been suggested for their potential anticancer and antibacterial effects [257]. These studies suggest that nanodelivery systems may enhance the potential of EGCG in cancer treatment. The anticancer effects of EGCG nanoemulsion on lung cancer have been analyzed, and the results indicate that nano EGCG may inhibit lung cancer cell proliferation, colony formation, migration, and invasion by activating protein signaling pathways and inhibiting AMPK [139].

Resveratrol

In one study, resveratrol was bioconjugated with gold nanoparticles using polyvinylpyrrolidone as a cross-linker. This led to improved delivery performance and enhanced the antitumor effects of resveratrol [258]. Another study investigated the targeted delivery of resveratrol to mitochondria by conjugating it to triphenylphosphonium (TPP), resulting in the potentiation and induction of mitochondrial-mediated apoptosis. Similarly, nanocarrier-based delivery systems have also been investigated to enhance the bioavailability and therapeutic potential of resveratrol [259].

Ginsenosides

Ginsenosides are important bioactive compounds that can be found in ginseng roots. These compounds have been studied for their potential health benefits, such as anticarcinoma, antioxidant, neuroprotective, and anti-inflammatory properties. Ginsenosides have demonstrated therapeutic potential in triggering apoptosis in tumor cells, decreasing proliferation, invasion, and metastasis, as well as reversing multidrug resistance [260]. These ginsenosides have been encapsulated or modified in various nanodelivery systems, such as polymeric nanoparticles, liposomes, micelles, and biomimetic nanoparticles, to enhance drug bioavailability and targeting ability [261]. In addition, ginsenosides serve as chemotherapeutic adjuvants and membrane stabilizers in a novel multifunctional liposome system that demonstrates antitumor efficacy and active targeting abilities [262]. Ginsenosides have been reported to activate the AMPK pathway, which can regulate metabolic reprogramming and reverse the Warburg effect in breast cancer [263]. New ginsenoside delivery systems have been developed to effectively target the AMPK pathway for cancer therapy, according to recent findings [264,265,266].

Conclusion, challenges/pitfalls, and future perspectives

As a major area of extensive attention, cancer metabolism has been an emerging hallmark of cancer. Targeting the major dysregulated metabolic pathways by multi-targeting phytochemicals could be a promising strategy to combat cancer cells. Despite the benefits behind targeted therapies, many cancer cells cannot be treated using just one type of treatment. Furthermore, considering the complexity of cancer metabolism, it is necessary to find new treatments that can target multiple dysregulated pathways. Accordingly, phytocompounds have indicated potential in combating cancer dysregulated pathways, but with fewer side effects than other commonly used treatments. Such potentials have introduced phytochemicals as promising compounds for prevention and treatment of cancer through regulating different signaling pathways.

Amongst those dysregulated pathways, AMPK/PGC-1α and interconnected pathways plays critical roles in cancer metabolism. In this line, alkaloids, phenolic compounds, terpenes/terpenoids, and several miscellaneous phytochemicals, represent major candidates in modulating cancer metabolism (Fig. 6). Phytochemicals have also demonstrated potential in the regulation of downstream signaling pathways of AMPK/PGC-1α, including angiogenesis, apoptosis, inflammation, and oxidative stress (Fig. 7).

Fig. 6
figure 6

Targeting AMPK/PGC-1α and interconnected signaling pathways by phytochemicals in cancer

Fig. 7
figure 7

Targeting the downstream signaling pathways of AMPK/PGC-1α by phytochemicals including angiogenesis, apoptosis, inflammation, and oxidative stress

The instability, low solubility/selectivity, poor bioavailability, rapid metabolism, and chemical degradation of phytochemicals limit their therapeutic applications in cancer. Employing novel delivery systems kindly overwhelmed such pharmacokinetic limitations by increasing bioavailability. Accordingly, lipid-based nanoparticles, polymeric nanoparticles, micelles, nanogels, cyclodextrin, gold, and mesoporous silica nanoparticles, have been critically employed to drawback the pharmacokinetic limitations of phytochemicals in targeting the AMPK/PGC-1α signaling pathway. Several phytochemicals have demonstrated notable antitumor effects by regulating cancer metabolism via influencing different signaling pathways, such as AMPK/PGC-1α, NF-κB, PI3K/Akt/mTOR, HIF-1α, ERK1/2, and the AMPK/mTORC2 axis. As a result, they suppress cancer metabolism, cell growth, invasion, and EMT. Phytocompounds are recognized as excellent and promising substances for the treatment of cancer.

Phytochemicals targeting AMPK/PGC-1α in cancer treatment face several challenges and limitations. One major challenge is the limited bioavailability of phytochemicals due to poor absorption and quick metabolism in the body. This can lead to decreased levels and lower concentrations of the phytochemicals when reaching their targeted tissues, diminishing their efficacy. It can also be difficult to determine how a certain phytochemical will interact with AMPK/PGC-1α signaling pathways in various types of cancer due to cancer biology being very complicated and cancer cells differing greatly from one another. Furthermore, the potential for off-target effects and interactions with other medications or treatments can pose as a risk. Another significant challenge and limitation in the use of phytochemicals targeting AMPK/PGC-1α in cancer treatment is the lack of extensive clinical studies to confirm and validate their effectiveness and safety. Promising outcomes in cell cultures and animal models require accurate clinical trials before being used in clinical practice. Insufficient clinical evidence makes it difficult to safely endorse the use of phytochemicals as a conventional therapy for cancer patients. Therefore, more well-designed clinical trials are needed to bridge the gap between preclinical research and the clinical application of phytochemicals targeting AMPK/PGC-1α in cancer therapy. While phytochemicals show promise as potential therapeutic agents in cancer treatment, further research is needed to address these challenges and optimize their use in clinical settings.

In summary, the current systematic review underlines the significance of targeting the AMPK/PGC-1α signaling pathway in cancer metabolism by multi-targeted phytochemicals. Future studies should focus on evaluating the effects of phytochemicals during well-controlled clinical trials in combating cancer.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ACC1/2:

Acetyl-CoA carboxylases 1/2

Akt:

Protein kinase B

AML:

Acute myeloid leukemia

AMP:

Adenosine monophosphate

AMPK:

Adenosine monophosphate-activated protein kinase

AR:

Androgen receptor

ATP:

Adenosine triphosphate

CaMKKβ:

Calcium/calmodulin-dependent protein kinase kinase-β

COX-2:

Cyclooxygenase-2

DAP1:

Death-associated protein 1

EGCG:

Epigallocatechin gallate

EGFR:

Epidermal growth factor receptor

EMT:

Epithelial-mesenchymal transition

ERAD:

Endoplasmic reticulum-associated protein degradation

ERK:

Extracellular signal-regulated protein kinase

ERR:

Estrogen-related receptor

EZH2:

Enhancer of zeste homolog 2

FAO:

Fatty acid oxidation

FAS:

Fatty acid synthase

FOXO3a:

Forkhead box O3a

GCN5:

General control non-depressible 5

Gli-1:

Glioma-associated oncogene 1

GSK-3β:

Glycogen synthase kinase-3β

HIF-1:

Hypoxia-inducible factor-1

IL-6:

Interleukin- 6

JAK:

Janus kinase

JNK:

c-Jun NH2-terminal kinase

KSR1:

Kinase suppressor of Ras 1

LIC:

Leukemia-initiating cells

LKB1:

Liver kinase B1

MAPK:

Mitogen-activated protein kinase

MCT1:

Monocarboxylate transporter 1

miRNAs or miR:

MicroRNAs

mTOR:

Mammalian target of rapamycin

mTORC1:

Mammalian target of rapamycin complex 1

NAD:

Nicotinamide adenine dinucleotide

NADPH:

Nicotinamide adenine dinucleotide phosphate hydrogen

NF-κB:

Nuclear factor-κB

NLRP:

Nod-like receptor protein

NMIIA:

Non-muscle myosin IIA

Nrf2:

Nuclear factor erythroid 2-related factor 2

NSCLC:

Non-small cell lung cancer

OXPHOS:

Oxidative phosphorylation

PD-L1:

Programmed cell death ligand 1

PERK:

Protein kinase R (PKR)-like endoplasmic reticulum kinase

PGC-1α:

Peroxisome proliferator-activated receptor-gamma coactivator-1α

PI3K:

Phosphoinositide 3-kinase

PP2A:

Protein phosphatase 2 A

PP2C:

Protein phosphatase 2 C

PPM1E:

Protein phosphatase 1E

pRb:

Retinoblastoma protein

PRC:

PGC-1-related coactivator

PRC2:

Polycomb repressive complex 2

RAPTOR:

Regulatory-associated protein of mTOR

RIP1:

Receptor-interacting protein 1

ROS:

Reactive oxygen species

SIRT3:

Sirtuin 3

STAT:

Signal transducer and activator of transcription

STK11:

Serine/threonine kinase 11

TAK1:

TGF-β-activated kinase 1

T-ALL:

T-cell acute lymphoblastic leukemia/lymphoma

TET2:

TET methylcytosine dioxygenase 2

TGF-β:

Transforming growth factor-beta

TLR:

Toll-like receptors

TME:

Tumor microenvironment

TNF-α:

Tumor necrosis factor-α

TPP:

Triphenylphosphonium

TSC2:

Tuberous sclerosis complex 2, also known as Tuberin

TWEAK:

TNF-related weak inducer of apoptosis

UBE2O:

Ubiquitin conjugating enzyme E2

ULK1:

UNC-51- like kinase 1

VEGF:

Vascular endothelial growth factor

YAP:

Yes-associated protein

References

  1. HemaIswarya S, Doble M. Potential synergism of natural products in the treatment of cancer. Phytotherapy Research: Int J Devoted Pharmacol Toxicol Evaluation Nat Prod Derivatives. 2006;20(4):239–49.

    Article  CAS  Google Scholar 

  2. Fakhri S, Piri S, Khan H. Cachexia and phytonutrients. The role of phytonutrients in metabolic disorders. edn.: Elsevier; 2022. pp. 397–417.

  3. Stark L, Tofthagen C, Visovsky C, McMillan SC. The Symptom experience of patients with Cancer. J Hospice Palliat Nursing: JHPN : Official J Hospice Palliat Nurses Association. 2012;14(1):61–70.

    Article  Google Scholar 

  4. Menendez JA, Alarcón T. Metabostemness: a new cancer hallmark. Front Oncol. 2014;4:262.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Gutschner T, Diederichs S. The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol. 2012;9(6):703–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46.

    Article  PubMed  CAS  Google Scholar 

  7. Gravandi MM, Abdian S, Tahvilian M, Iranpanah A, Moradi SZ, Fakhri S, Echeverría J. Therapeutic targeting of Ras/Raf/MAPK pathway by natural products: a systematic and mechanistic approach for neurodegeneration. Phytomedicine. 2023;115:154821.

    Article  PubMed  CAS  Google Scholar 

  8. Yao CA, Ortiz-Vega S, Sun YY, Chien CT, Chuang JH, Lin Y. Association of mSin1 with mTORC2 Ras and Akt reveals a crucial domain on mSin1 involved in akt phosphorylation. Oncotarget. 2017;8(38):63392–404.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Fakhri S, Moradi SZ, Farzaei MH, Bishayee A. Modulation of dysregulated cancer metabolism by plant secondary metabolites: a mechanistic review. Seminars in cancer biology: 2020. Elsevier; 2020.

  10. Bordoloi D, Roy K, Monisha N, Padmavathi J, Kunnumakkara GB A. Multi-targeted agents in cancer cell chemosensitization: what we learnt from curcumin thus far. Recent Pat Anti-cancer Drug Discov. 2016;11(1):67–97.

    Article  CAS  Google Scholar 

  11. Bishayee A, Sethi G. Bioactive natural products in cancer prevention and therapy: progress and promise. Sem Cancer Biol. 2016;40–41:1–3.

    Article  Google Scholar 

  12. Kumar A, P N, Kumar M, Jose A, Tomer V, Oz E, Proestos C, Zeng M, Elobeid T, K S et al. (2023) Major phytochemicals: recent advances in Health benefits and extraction method. Molecules, 28(2).

  13. Huang M, Lu J-J, Ding J. Natural products in cancer therapy: past, present and future. Nat Prod Bioprospecting. 2021;11:5–13.

    Article  Google Scholar 

  14. Tewari D, Patni P, Bishayee A, Sah AN, Bishayee A. Natural products targeting the PI3K-Akt-mTOR signaling pathway in cancer: a novel therapeutic strategy. Sem Cancer Biol. 2022;80:1–17.

    Article  Google Scholar 

  15. Kaur C, Sahu SK, Bansal K, DeLiberto LK, Zhang J, Tewari D, Bishayee A. Targeting peroxisome proliferator-activated Receptor-β/δ, Reactive Oxygen Species and Redox Signaling with Phytocompounds for Cancer Therapy. Antioxid Redox Signal; 2024.

  16. Bose S, Banerjee S, Mondal A, Chakraborty U, Pumarol J, Croley CR, Bishayee A. (2020) Targeting the JAK/STAT signaling pathway using Phytocompounds for Cancer Prevention and Therapy. Cells, 9(6).

  17. Tewari D, Priya A, Bishayee A, Bishayee A. Targeting transforming growth factor-β signalling for cancer prevention and intervention: recent advances in developing small molecules of natural origin. Clin Translational Med. 2022;12(4):e795.

    Article  CAS  Google Scholar 

  18. Choudhari AS, Mandave PC, Deshpande M, Ranjekar P, Prakash O. Phytochemicals in Cancer Treatment: from preclinical studies to clinical practice. Front Pharmacol. 2019;10:1614.

    Article  PubMed  CAS  Google Scholar 

  19. Kim MO, Lee M-H, Oi N, Kim S-H, Bae KB, Huang Z, Kim DJ, Reddy K, Lee S-Y, Park SJ. [6]-Shogaol inhibits growth and induces apoptosis of non-small cell lung cancer cells by directly regulating Akt1/2. Carcinogenesis. 2014;35(3):683–91.

    Article  PubMed  CAS  Google Scholar 

  20. Wang T, Jiang Y, Chu L, Wu T, You J. Alpinumisoflavone suppresses tumour growth and metastasis of clear-cell renal cell carcinoma. Am J cancer Res. 2017;7(4):999.

    PubMed  PubMed Central  CAS  Google Scholar 

  21. Kooshki L, Mahdavi P, Fakhri S, Akkol EK, Khan H. Targeting lactate metabolism and glycolytic pathways in the tumor microenvironment by natural products: a promising strategy in combating cancer. BioFactors (Oxford, England; 2021.

  22. İpek P, Atalar MN, Baran A, Baran MF, Ommati MM, Karadag M, Zor M, Eftekhari A, Alma MH, Benis KZ, et al. Determination of chemical components of the endemic species Allium Turcicum L. plant extract by LC-MS/MS and evaluation of medicinal potentials. Heliyon. 2024;10(6):e27386.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Khalilov R, Bakishzade A, Nasibova A. Future prospects of biomaterials in nanomedicine. Adv Biology Earth Sci. 2024;9:5–10.

    Article  Google Scholar 

  24. Rosic G, Selakovic D, Omarova S. (2024) CANCER SIGNALING, CELL/GENE THERAPY, DIAGNOSIS AND ROLE OF NANOBIOMATERIALS. Adv Biology Earth Sci, 9.

  25. Keerthana CK, Rayginia TP, Shifana SC, Anto NP, Kalimuthu K, Isakov N, Anto RJ. The role of AMPK in cancer metabolism and its impact on the immunomodulation of the tumor microenvironment. Front Immunol. 2023;14:1114582.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Kemp BE, Oakhill JS, Scott JW. AMPK structure and regulation from three angles. Structure. 2007;15(10):1161–3.

    Article  PubMed  CAS  Google Scholar 

  27. Hardie DG, Schaffer BE, Brunet A. AMPK: an energy-sensing pathway with multiple inputs and outputs. Trends Cell Biol. 2016;26(3):190–201.

    Article  PubMed  CAS  Google Scholar 

  28. Hardie DG. AMPK—sensing energy while talking to other signaling pathways. Cell Metabol. 2014;20(6):939–52.

    Article  CAS  Google Scholar 

  29. Towler MC, Hardie DG. AMP-activated protein kinase in metabolic control and insulin signaling. Circul Res. 2007;100(3):328–41.

    Article  CAS  Google Scholar 

  30. McBride A, Hardie D. AMP-activated protein kinase–a sensor of glycogen as well as AMP and ATP? Acta Physiol. 2009;196(1):99–113.

    Article  CAS  Google Scholar 

  31. CHEUNG PC, SALT IP, DAVIES SP, HARDIE DG, CARLING D. Characterization of AMP-activated protein kinase γ-subunit isoforms and their role in AMP binding. Biochem J. 2000;346(3):659–69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 2012;13(4):251–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Gowans GJ, Hawley SA, Ross FA, Hardie DG. AMP is a true physiological regulator of AMP-activated protein kinase by both allosteric activation and enhancing net phosphorylation. Cell Metabol. 2013;18(4):556–66.

    Article  CAS  Google Scholar 

  34. Jeon S-M, Hay N. The double-edged sword of AMPK signaling in cancer and its therapeutic implications. Arch Pharm Res. 2015;38:346–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Yan Y, Zhou XE, Xu HE, Melcher K. Structure and physiological regulation of AMPK. Int J Mol Sci. 2018;19(11):3534.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Vara-Ciruelos D, Russell FM, Hardie DG. The strange case of AMPK and cancer: Dr Jekyll or Mr Hyde? Open Biology. 2019;9(7):190099.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Hinchy EC, Gruszczyk AV, Willows R, Navaratnam N, Hall AR, Bates G, Bright TP, Krieg T, Carling D, Murphy MP. Mitochondria-derived ROS activate AMP-activated protein kinase (AMPK) indirectly. J Biol Chem. 2018;293(44):17208–17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Park CE, Yun H, Lee E-B, Min B-I, Bae H, Choe W, Kang I, Kim S-S, Ha J. The antioxidant effects of genistein are associated with AMP-activated protein kinase activation and PTEN induction in prostate cancer cells. J Med Food. 2010;13(4):815–20.

    Article  PubMed  CAS  Google Scholar 

  39. Naik PP, Mukhopadhyay S, Praharaj PP, Bhol CS, Panigrahi DP, Mahapatra KK, Patra S, Saha S, Panda AK, Panda K. Secretory clusterin promotes oral cancer cell survival via inhibiting apoptosis by activation of autophagy in AMPK/mTOR/ULK1 dependent pathway. Life Sci. 2021;264:118722.

    Article  PubMed  CAS  Google Scholar 

  40. Sinha RA, Singh BK, Zhou J, Wu Y, Farah BL, Ohba K, Lesmana R, Gooding J, Bay B-H, Yen PM. Thyroid hormone induction of mitochondrial activity is coupled to mitophagy via ROS-AMPK-ULK1 signaling. Autophagy. 2015;11(8):1341–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Cai X, Hu X, Cai B, Wang Q, Li Y, Tan X, Hu H, Chen X, Huang J, Cheng J. Metformin suppresses hepatocellular carcinoma cell growth through induction of cell cycle G1/G0 phase arrest and p21CIP and p27KIP expression and downregulation of cyclin D1 in vitro and in vivo. Oncol Rep. 2013;30(5):2449–57.

    Article  PubMed  CAS  Google Scholar 

  42. Rao E, Zhang Y, Zhu G, Hao J, Persson X-MT, Egilmez NK, Suttles J, Li B. Deficiency of AMPK in CD8 + T cells suppresses their anti-tumor function by inducing protein phosphatase-mediated cell death. Oncotarget. 2015;6(10):7944.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Hsu C-C, Peng D, Cai Z, Lin H-K. AMPK signaling and its targeting in cancer progression and treatment. Seminars in cancer biology: 2022. Elsevier; 2022. pp. 52–68.

  44. Sun W, Qian K, Guo K, Chen L, Xiang J, Li D, Wu Y, Ji Q, Sun T, Wang Z. LHPP inhibits cell growth and migration and triggers autophagy in papillary thyroid cancer by regulating the AKT/AMPK/mTOR signaling pathway. Acta Biochim Biophys Sin. 2020;52(4):382–9.

    Article  PubMed  CAS  Google Scholar 

  45. Kishton RJ, Barnes CE, Nichols AG, Cohen S, Gerriets VA, Siska PJ, Macintyre AN, Goraksha-Hicks P, De Cubas AA, Liu T. AMPK is essential to balance glycolysis and mitochondrial metabolism to control T-ALL cell stress and survival. Cell Metabol. 2016;23(4):649–62.

    Article  CAS  Google Scholar 

  46. Wang W, Xiao Z-D, Li X, Aziz KE, Gan B, Johnson RL, Chen J. AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat Cell Biol. 2015;17(4):490–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Li Y-H, Luo J, Mosley Y-YC, Hedrick VE, Paul LN, Chang J, Zhang G, Wang Y-K, Banko MR, Brunet A. AMP-activated protein kinase directly phosphorylates and destabilizes hedgehog pathway transcription factor GLI1 in medulloblastoma. Cell Rep. 2015;12(4):599–609.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Wu D, Hu D, Chen H, Shi G, Fetahu IS, Wu F, Rabidou K, Fang R, Tan L, Xu S. Glucose-regulated phosphorylation of TET2 by AMPK reveals a pathway linking diabetes to cancer. Nature. 2018;559(7715):637–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Cha J-H, Yang W-H, Xia W, Wei Y, Chan L-C, Lim S-O, Li C-W, Kim T, Chang S-S, Lee H-H. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol Cell. 2018;71(4):606–20. e607.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Ashrafizadeh M, Mirzaei S, Hushmandi K, Rahmanian V, Zabolian A, Raei M, Farahani MV, Goharrizi MASB, Khan H, Zarrabi A. Therapeutic potential of AMPK signaling targeting in lung cancer: advances, challenges and future prospects. Life Sci. 2021;278:119649.

    Article  PubMed  CAS  Google Scholar 

  51. Wan L, Xu K, Wei Y, Zhang J, Han T, Fry C, Zhang Z, Wang YV, Huang L, Yuan M. Phosphorylation of EZH2 by AMPK suppresses PRC2 methyltransferase activity and oncogenic function. Mol Cell. 2018;69(2):279–91. e275.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Penfold L, Woods A, Muckett P, Nikitin AY, Kent TR, Zhang S, Graham R, Pollard A, Carling D. CAMKK2 promotes prostate cancer independently of AMPK via increased lipogenesis. Cancer Res. 2018;78(24):6747–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Pineda CT, Ramanathan S, Tacer KF, Weon JL, Potts MB, Ou Y-H, White MA, Potts PR. Degradation of AMPK by a cancer-specific ubiquitin ligase. Cell. 2015;160(4):715–28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Vila IK, Yao Y, Kim G, Xia W, Kim H, Kim S-J, Park M-K, Hwang JP, González-Billalabeitia E, Hung M-C. A UBE2O-AMPKα2 axis that promotes tumor initiation and progression offers opportunities for therapy. Cancer Cell. 2017;31(2):208–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Houde VP, Donzelli S, Sacconi A, Galic S, Hammill JA, Bramson JL, Foster RA, Tsakiridis T, Kemp BE, Grasso G. AMPK β1 reduces tumor progression and improves survival in p53 null mice. Mol Oncol. 2017;11(9):1143–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Chaube B, Malvi P, Singh SV, Mohammad N, Viollet B, Bhat MK. AMPK maintains energy homeostasis and survival in cancer cells via regulating p38/PGC-1α-mediated mitochondrial biogenesis. Cell Death Discovery. 2015;1(1):1–11.

    Article  Google Scholar 

  57. Sadria M, Seo D, Layton AT. The mixed blessing of AMPK signaling in Cancer treatments. BMC Cancer. 2022;22(1):1–16.

    Article  Google Scholar 

  58. Jeon S-M, Chandel NS, Hay N. AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature. 2012;485(7400):661–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Han F, Li C-F, Cai Z, Zhang X, Jin G, Zhang W-N, Xu C, Wang C-Y, Morrow J, Zhang S. The critical role of AMPK in driving akt activation under stress, tumorigenesis and drug resistance. Nat Commun. 2018;9(1):4728.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Saito Y, Chapple RH, Lin A, Kitano A, Nakada D. AMPK protects leukemia-initiating cells in myeloid leukemias from metabolic stress in the bone marrow. Cell Stem Cell. 2015;17(5):585–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Cai Z, Li C-F, Han F, Liu C, Zhang A, Hsu C-C, Peng D, Zhang X, Jin G, Rezaeian A-H. Phosphorylation of PDHA by AMPK drives TCA cycle to promote cancer metastasis. Mol Cell. 2020;80(2):263–78. e267.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Eichner LJ, Brun SN, Herzig S, Young NP, Curtis SD, Shackelford DB, Shokhirev MN, Leblanc M, Vera LI, Hutchins A. Genetic analysis reveals AMPK is required to support tumor growth in murine Kras-dependent lung cancer models. Cell Metabol. 2019;29(2):285–302. e287.

    Article  CAS  Google Scholar 

  63. Bost F, Kaminski L. The metabolic modulator PGC-1α in cancer. Am J Cancer Res. 2019;9(2):198–211.

    PubMed  PubMed Central  CAS  Google Scholar 

  64. Mastropasqua F, Girolimetti G, Shoshan M. (2018) PGC1α: friend or foe in Cancer? Genes (Basel), 9(1).

  65. Handschin C, Spiegelman BM. The role of exercise and PGC1alpha in inflammation and chronic disease. Nature. 2008;454(7203):463–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Villena JA. New insights into PGC-1 coactivators: redefining their role in the regulation of mitochondrial function and beyond. FEBS J. 2015;282(4):647–72.

    Article  PubMed  CAS  Google Scholar 

  67. Tan Z, Luo X, Xiao L, Tang M, Bode AM, Dong Z, Cao Y. The role of PGC1α in Cancer Metabolism and its therapeutic implications. Mol Cancer Ther. 2016;15(5):774–82.

    Article  PubMed  CAS  Google Scholar 

  68. Bhalla K, Hwang BJ, Dewi RE, Ou L, Twaddel W, Fang HB, Vafai SB, Vazquez F, Puigserver P, Boros L, et al. PGC1α promotes tumor growth by inducing gene expression programs supporting lipogenesis. Cancer Res. 2011;71(21):6888–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. McGuirk S, Gravel SP, Deblois G, Papadopoli DJ, Faubert B, Wegner A, Hiller K, Avizonis D, Akavia UD, Jones RG, et al. PGC-1α supports glutamine metabolism in breast cancer. Cancer Metabolism. 2013;1(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Vazquez F, Lim JH, Chim H, Bhalla K, Girnun G, Pierce K, Clish CB, Granter SR, Widlund HR, Spiegelman BM, et al. PGC1α expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell. 2013;23(3):287–301.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Luo C, Lim JH, Lee Y, Granter SR, Thomas A, Vazquez F, Widlund HR, Puigserver P. A PGC1α-mediated transcriptional axis suppresses melanoma metastasis. Nature. 2016;537(7620):422–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Haq R, Shoag J, Andreu-Perez P, Yokoyama S, Edelman H, Rowe GC, Frederick DT, Hurley AD, Nellore A, Kung AL, et al. Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. Cancer Cell. 2013;23(3):302–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Peters JM, Shah YM, Gonzalez FJ. The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention. Nat Rev Cancer. 2012;12(3):181–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Gerhart-Hines Z, Rodgers JT, Bare O, Lerin C, Kim SH, Mostoslavsky R, Alt FW, Wu Z, Puigserver P. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. EMBO J. 2007;26(7):1913–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Sen N, Satija YK, Das S. PGC-1α, a key modulator of p53, promotes cell survival upon metabolic stress. Mol Cell. 2011;44(4):621–34.

    Article  PubMed  CAS  Google Scholar 

  76. Cioce M, Blandino G. PGC1α confers specificity-metabolic stress and p53-dependent transcription. Mol Cell. 2011;44(4):515–6.

    Article  PubMed  CAS  Google Scholar 

  77. Chen W, Wang Q, Bai L, Chen W, Wang X, Tellez CS, Leng S, Padilla MT, Nyunoya T, Belinsky SA, et al. RIP1 maintains DNA integrity and cell proliferation by regulating PGC-1α-mediated mitochondrial oxidative phosphorylation and glycolysis. Cell Death Differ. 2014;21(7):1061–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Sancho P, Burgos-Ramos E, Tavera A, Bou Kheir T, Jagust P, Schoenhals M, Barneda D, Sellers K, Campos-Olivas R, Graña O, et al. MYC/PGC-1α balance determines the metabolic phenotype and plasticity of pancreatic Cancer stem cells. Cell Metab. 2015;22(4):590–605.

    Article  PubMed  CAS  Google Scholar 

  79. Dang CV. (2013) MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harbor Perspect Med, 3(8).

  80. Olmos Y, Valle I, Borniquel S, Tierrez A, Soria E, Lamas S, Monsalve M. Mutual dependence of Foxo3a and PGC-1alpha in the induction of oxidative stress genes. J Biol Chem. 2009;284(21):14476–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Fisher KW, Das B, Kortum RL, Chaika OV, Lewis RE. Kinase suppressor of ras 1 (KSR1) regulates PGC1α and estrogen-related receptor α to promote oncogenic ras-dependent anchorage-independent growth. Mol Cell Biol. 2011;31(12):2453–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Deblois G, St-Pierre J, Giguère V. The PGC-1/ERR signaling axis in cancer. Oncogene. 2013;32(30):3483–90.

    Article  PubMed  CAS  Google Scholar 

  83. Klimcakova E, Chénard V, McGuirk S, Germain D, Avizonis D, Muller WJ, St-Pierre J. PGC-1α promotes the growth of ErbB2/Neu-induced mammary tumors by regulating nutrient supply. Cancer Res. 2012;72(6):1538–46.

    Article  PubMed  CAS  Google Scholar 

  84. Audet-Walsh É, Papadopoli DJ, Gravel SP, Yee T, Bridon G, Caron M, Bourque G, Giguère V, St-Pierre J. The PGC-1α/ERRα Axis represses one-Carbon Metabolism and promotes sensitivity to anti-folate therapy in breast Cancer. Cell Rep. 2016;14(4):920–31.

    Article  PubMed  CAS  Google Scholar 

  85. Tennakoon JB, Shi Y, Han JJ, Tsouko E, White MA, Burns AR, Zhang A, Xia X, Ilkayeva OR, Xin L, et al. Androgens regulate prostate cancer cell growth via an AMPK-PGC-1α-mediated metabolic switch. Oncogene. 2014;33(45):5251–61.

    Article  PubMed  CAS  Google Scholar 

  86. Neill T, Torres A, Buraschi S, Owens RT, Hoek JB, Baffa R, Iozzo RV. Decorin induces mitophagy in breast carcinoma cells via peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) and mitostatin. J Biol Chem. 2014;289(8):4952–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Morandi A, Giannoni E, Chiarugi P. Nutrient Exploitation within the Tumor-Stroma metabolic crosstalk. Trends cancer. 2016;2(12):736–46.

    Article  PubMed  Google Scholar 

  88. Porporato PE, Payen VL, Baselet B, Sonveaux P. Metabolic changes associated with tumor metastasis, part 2: Mitochondria, lipid and amino acid metabolism. Cell Mol Life Sci. 2016;73(7):1349–63.

    Article  PubMed  CAS  Google Scholar 

  89. Jäger S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc Natl Acad Sci USA. 2007;104(29):12017–22.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Barrès R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, Krook A, Zierath JR. Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density. Cell Metab. 2009;10(3):189–98.

    Article  PubMed  Google Scholar 

  91. Anderson RM, Barger JL, Edwards MG, Braun KH, O’Connor CE, Prolla TA, Weindruch R. Dynamic regulation of PGC-1alpha localization and turnover implicates mitochondrial adaptation in calorie restriction and the stress response. Aging Cell. 2008;7(1):101–11.

    Article  PubMed  CAS  Google Scholar 

  92. Li J, Ke W, Zhou Q, Wu Y, Luo H, Zhou H, Yang B, Guo Y, Zheng Q, Zhang Y. Tumour necrosis factor-α promotes liver ischaemia-reperfusion injury through the PGC-1α/Mfn2 pathway. J Cell Mol Med. 2014;18(9):1863–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Barroso WA, Victorino VJ, Jeremias IC, Petroni RC, Ariga SKK, Salles TA, Barbeiro DF, de Lima TM, de Souza HP. High-fat diet inhibits PGC-1α suppressive effect on NFκB signaling in hepatocytes. Eur J Nutr. 2018;57(5):1891–900.

    Article  PubMed  CAS  Google Scholar 

  94. Bernardini JP, Lazarou M, Dewson G. Parkin and mitophagy in cancer. Oncogene. 2017;36(10):1315–27.

    Article  PubMed  CAS  Google Scholar 

  95. Lou C, Xiao M, Cheng S, Lu X, Jia S, Ren Y, Li Z. MiR-485-3p and mir-485-5p suppress breast cancer cell metastasis by inhibiting PGC-1α expression. Cell Death Dis. 2016;7(3):e2159.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Wang B, Hsu SH, Frankel W, Ghoshal K, Jacob ST. Stat3-mediated activation of microRNA-23a suppresses gluconeogenesis in hepatocellular carcinoma by down-regulating glucose-6-phosphatase and peroxisome proliferator-activated receptor gamma, coactivator 1 alpha. Hepatology (Baltimore MD). 2012;56(1):186–97.

    Article  PubMed  CAS  Google Scholar 

  97. Zhang S, Liu X, Liu J, Guo H, Xu H, Zhang G. PGC-1 alpha interacts with microRNA-217 to functionally regulate breast cancer cell proliferation. Biomed Pharmacotherapy = Biomedecine Pharmacotherapie. 2017;85:541–8.

    Article  CAS  Google Scholar 

  98. Kulikov AV, Luchkina EA, Gogvadze V, Zhivotovsky B. Mitophagy: link to cancer development and therapy. Biochem Biophys Res Commun. 2017;482(3):432–9.

    Article  PubMed  CAS  Google Scholar 

  99. Drake LE, Springer MZ, Poole LP, Kim CJ, Macleod KF. Expanding perspectives on the significance of mitophagy in cancer. Sem Cancer Biol. 2017;47:110–24.

    Article  CAS  Google Scholar 

  100. Vega RB, Huss JM, Kelly DP. The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor α in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes. Mol Cell Biol. 2000;20(5):1868–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Moradi SZ, Momtaz S, Bayrami Z, Farzaei MH, Abdollahi M. Nanoformulations of herbal extracts in treatment of neurodegenerative disorders. Front Bioeng Biotechnol. 2020;8:238.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Moradi SZ, Jalili F, Farhadian N, Joshi T, Wang M, Zou L, Cao H, Farzaei MH, Xiao J. Polyphenols and neurodegenerative diseases: focus on neuronal regeneration. Crit Rev Food Sci Nutr. 2022;62(13):3421–36.

    Article  PubMed  CAS  Google Scholar 

  103. Fakhri S, Moradi SZ, Abbaszadeh F, Faraji F, Amirian R, Sinha D, McMahon EG, Bishayee A. (2024) Targeting the key players of phenotypic plasticity in cancer cells by phytochemicals. Cancer Metastasis Rev:1–32.

  104. Fakhri S, Abbaszadeh F, Moradi SZ, Cao H, Khan H, Xiao J. (2022) Effects of Polyphenols on Oxidative Stress, Inflammation, and Interconnected Pathways during Spinal Cord Injury. Oxidative Medicine and Cellular Longevity, 2022.

  105. Sajadimajd S, Moradi SZ, Akbari V, Aghaz F, Farzaei MH. Nanoformulated herbal bioactives for the treatment of neurodegenerative disorders. Herbal bioactive-based drug Delivery systems. edn.: Elsevier; 2022. pp. 371–91.

  106. Kooshki L, Zarneshan SN, Fakhri S, Moradi SZ, Echeverria J. The pivotal role of JAK/STAT and IRS/PI3K signaling pathways in neurodegenerative diseases: mechanistic approaches to polyphenols and alkaloids. Phytomedicine. 2023;112:154686.

    Article  PubMed  CAS  Google Scholar 

  107. Ko JH, Sethi G, Um JY, Shanmugam MK, Arfuso F, Kumar AP, Bishayee A, Ahn KS. (2017) The role of Resveratrol in Cancer Therapy. Int J Mol Sci, 18(12).

  108. Bishayee A. Cancer prevention and treatment with resveratrol: from rodent studies to clinical trials. Cancer Prev Res (Philadelphia Pa). 2009;2(5):409–18.

    Article  CAS  Google Scholar 

  109. Liu Y, Tong L, Luo Y, Li X, Chen G, Wang Y. Resveratrol inhibits the proliferation and induces the apoptosis in ovarian cancer cells via inhibiting glycolysis and targeting AMPK/mTOR signaling pathway. J Cell Biochem. 2018;119(7):6162–72.

    Article  PubMed  CAS  Google Scholar 

  110. Park SW, Yoon S, Moon JS, Park BW, Kim KS. Resveratrol Downregulates Acetyl-CoA carboxylase α and fatty acid synthase by AMPK-mediated downregulation of mTOR in breast Cancer cells. Food Sci Biotechnol. 2008;17(5):1047–51.

    CAS  Google Scholar 

  111. Puissant A, Robert G, Fenouille N, Luciano F, Cassuto JP, Raynaud S, Auberger P. Resveratrol promotes autophagic cell death in chronic myelogenous leukemia cells via JNK-mediated p62/SQSTM1 expression and AMPK activation. Cancer Res. 2010;70(3):1042–52.

    Article  PubMed  CAS  Google Scholar 

  112. Saunier E, Antonio S, Regazzetti A, Auzeil N, Laprévote O, Shay JW, Coumoul X, Barouki R, Benelli C, Huc L, et al. Resveratrol reverses the Warburg effect by targeting the pyruvate dehydrogenase complex in colon cancer cells. Sci Rep. 2017;7(1):6945.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Wang ZY, Zhang L, Ni ZH, Sun J, Gao H, Cheng ZA, Xu JH, Yin PH. Resveratrol induces AMPK-dependent MDR1 inhibition in colorectal cancer HCT116/L-OHP cells by preventing activation of NF-κB signaling and suppressing cAMP-responsive element transcriptional activity. Tumor Biology. 2015;36(12):9499–510.

    Article  PubMed  CAS  Google Scholar 

  114. Selvaraj S, Sun Y, Sukumaran P, Singh BB. Resveratrol activates autophagic cell death in prostate cancer cells via downregulation of STIM1 and the mTOR pathway. Mol Carcinog. 2016;55(5):818–31.

    Article  PubMed  CAS  Google Scholar 

  115. Xing J, Wang Z, Xu H, Liu C, Wei Z, Zhao L, Ren L. Pak2 inhibition promotes resveratrol-mediated glioblastoma A172 cell apoptosis via modulating the AMPK-YAP signaling pathway. J Cell Physiol. 2020;235(10):6563–73.

    Article  PubMed  CAS  Google Scholar 

  116. Fakhri S, Piri S, Moradi SZ, Khan H. Phytochemicals targeting oxidative stress, interconnected neuroinflammatory, and neuroapoptotic pathways following radiation. Curr Neuropharmacol. 2022;20(5):836.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Fakhri S, Moradi SZ, Farzaei MH, Bishayee A. Modulation of dysregulated cancer metabolism by plant secondary metabolites: a mechanistic review. Seminars in cancer biology: 2022. Elsevier; 2022. pp. 276–305.

  118. Fakhri S, Gravandi MM, Abdian S, Moradi SZ, Echeverría J. Quercetin derivatives in combating spinal cord Injury: a mechanistic and systematic review. Life. 2022;12(12):1960.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Khan F, Niaz K, Maqbool F, Ismail Hassan F, Abdollahi M, Nagulapalli Venkata KC, Nabavi SM, Bishayee A. (2016) Molecular targets underlying the Anticancer effects of Quercetin: an update. Nutrients, 8(9).

  120. Guo H, Ding H, Tang X, Liang M, Li S, Zhang J, Cao J. Quercetin induces pro-apoptotic autophagy via SIRT1/AMPK signaling pathway in human lung cancer cell lines A549 and H1299 in vitro. Thorac Cancer. 2021;12(9):1415–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Jung JH, Lee JO, Kim JH, Lee SK, You GY, Park SH, Park JM, Kim EK, Suh PG, An JK, et al. Quercetin suppresses HeLa cell viability via AMPK-induced HSP70 and EGFR down-regulation. J Cell Physiol. 2010;223(2):408–14.

    Article  PubMed  CAS  Google Scholar 

  122. Kim HJ, Kim SK, Kim BS, Lee SH, Park YS, Park BK, Kim SJ, Kim J, Choi C, Kim JS, et al. Apoptotic effect of quercetin on HT-29 colon cancer cells via the AMPK signaling pathway. J Agric Food Chem. 2010;58(15):8643–50.

    Article  PubMed  CAS  Google Scholar 

  123. Kim HS, Wannatung T, Lee S, Yang WK, Chung SH, Lim JS, Choe W, Kang I, Kim SS, Ha J. Quercetin enhances hypoxia-mediated apoptosis via direct inhibition of AMPK activity in HCT116 colon cancer. Apoptosis. 2012;17(9):938–49.

    Article  PubMed  CAS  Google Scholar 

  124. Rivera Rivera A, Castillo-Pichardo L, Gerena Y, Dharmawardhane S. Anti-breast Cancer potential of Quercetin via the Akt/AMPK/Mammalian target of Rapamycin (mTOR) Signaling Cascade. PLoS ONE. 2016;11(6):e0157251.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Fakhri S, Pesce M, Patruno A, Moradi SZ, Iranpanah A, Farzaei MH, Sobarzo-Sánchez E. Attenuation of Nrf2/Keap1/ARE in Alzheimer’s Disease by Plant secondary metabolites: a mechanistic review. Molecules. 2020;25(21):4926.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Shui L, Wang W, Xie M, Ye B, Li X, Liu Y, Zheng M. Isoquercitrin induces apoptosis and autophagy in hepatocellular carcinoma cells via AMPK/mTOR/p70S6K signaling pathway. Aging. 2020;12(23):24318–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Fakhri S, Moradi SZ, Yarmohammadi A, Narimani F, Wallace CE, Bishayee A. Modulation of TLR/NF-κB/NLRP signaling by bioactive phytocompounds: a promising strategy to augment cancer chemotherapy and immunotherapy. Front Oncol. 2022;12:834072.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Fakhri S, Moradi SZ, Nouri Z, Cao H, Wang H, Khan H, Xiao J. (2022) Modulation of integrin receptor by polyphenols: downstream Nrf2-Keap1/ARE and associated cross-talk mediators in cardiovascular diseases. Crit Rev Food Sci Nutr:1–25.

  129. Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. (2023) Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev:1–74.

  130. Fakhri S, Moradi SZ, Faraji F, Farhadi T, Hesami O, Iranpanah A, Webber K, Bishayee A. Current advances in nanoformulations of therapeutic agents targeting tumor microenvironment to overcome drug resistance. Cancer Metastasis Rev. 2023;42(3):959–1020.

    Article  PubMed  Google Scholar 

  131. Bianchi G, Ravera S, Traverso C, Amaro A, Piaggio F, Emionite L, Bachetti T, Pfeffer U, Raffaghello L. Curcumin induces a fatal energetic impairment in tumor cells in vitro and in vivo by inhibiting ATP-synthase activity. Carcinogenesis. 2018;39(9):1141–50.

    Article  PubMed  CAS  Google Scholar 

  132. Salucci S, Bavelloni A, Stella AB, Fabbri F, Vannini I, Piazzi M, Volkava K, Scotlandi K, Martinelli G, Faenza I et al. (2023) The cytotoxic effect of Curcumin in Rhabdomyosarcoma is Associated with the modulation of AMPK, AKT/mTOR, STAT, and p53 signaling. Nutrients, 15(3).

  133. Zhang YJ, Xiang H, Liu JS, Li D, Fang ZY, Zhang H. Study on the mechanism of AMPK signaling pathway and its effect on apoptosis of human hepatocellular carcinoma SMMC-7721 cells by curcumin. Eur Rev Med Pharmacol Sci. 2017;21(5):1144–50.

    PubMed  Google Scholar 

  134. Fakhri S, Moradi SZ, Ash-Rafzadeh A, Bishayee A. Targeting cellular senescence in cancer by plant secondary metabolites: a systematic review. Pharmacol Res. 2022;177:105961.

    Article  PubMed  CAS  Google Scholar 

  135. Fakhri S, Iranpanah A, Gravandi MM, Moradi SZ, Ranjbari M, Majnooni MB, Echeverría J, Qi Y, Wang M, Liao P. Natural products attenuate PI3K/Akt/mTOR signaling pathway: a promising strategy in regulating neurodegeneration. Phytomedicine. 2021;91:153664.

    Article  PubMed  CAS  Google Scholar 

  136. Fakhri S, Darvish E, Narimani F, Moradi SZ, Abbaszadeh F, Khan H. The regulatory role of non-coding RNAs and their interactions with phytochemicals in neurodegenerative diseases: a systematic review. Brief Funct Genomics. 2023;22(2):143–60.

    Article  PubMed  Google Scholar 

  137. Fakhri S, Abdian S, Zarneshan SN, Moradi SZ, Farzaei MH, Abdollahi M. (2022) Nanoparticles in combating neuronal dysregulated signaling pathways: recent approaches to the nanoformulations of phytochemicals and synthetic drugs against neurodegenerative diseases. Int J Nanomed:299–331.

  138. Aggarwal V, Tuli HS, Tania M, Srivastava S, Ritzer EE, Pandey A, Aggarwal D, Barwal TS, Jain A, Kaur G, et al. Molecular mechanisms of action of epigallocatechin gallate in cancer: recent trends and advancement. Sem Cancer Biol. 2022;80:256–75.

    Article  CAS  Google Scholar 

  139. Chen BH, Hsieh CH, Tsai SY, Wang CY, Wang CC. Anticancer effects of epigallocatechin-3-gallate nanoemulsion on lung cancer cells through the activation of AMP-activated protein kinase signaling pathway. Sci Rep. 2020;10(1):5163.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Hwang JT, Ha J, Park IJ, Lee SK, Baik HW, Kim YM, Park OJ. Apoptotic effect of EGCG in HT-29 colon cancer cells via AMPK signal pathway. Cancer Lett. 2007;247(1):115–21.

    Article  PubMed  CAS  Google Scholar 

  141. Wang Y, Pan H, chen D, Guo D, Wang X. (2021) Targeting at cancer energy metabolism and lipid droplet formation as new treatment strategies for epigallocatechin-3-gallate (EGCG) in colorectal cancer cells. J Funct Foods, 83.

  142. Pandey P, Khan F, Upadhyay TK. Deciphering the modulatory role of apigenin targeting oncogenic pathways in human cancers. Chem Biol Drug Des. 2023;101(6):1446–58.

    Article  PubMed  CAS  Google Scholar 

  143. Lin SC, Chen MC, Liu S, Callahan VM, Bracci NR, Lehman CW, Dahal B, de la Fuente CL, Lin CC, Wang TT, et al. Phloretin inhibits Zika virus infection by interfering with cellular glucose utilisation. Int J Antimicrob Agents. 2019;54(1):80–4.

    Article  PubMed  CAS  Google Scholar 

  144. Kim TW, Lee HG. (2021) Apigenin Induces Autophagy and Cell Death by Targeting EZH2 under Hypoxia Conditions in Gastric Cancer Cells. Int J Mol Sci, 22(24).

  145. Wang KL, Yu YC, Hsia SM. (2021) Perspectives on the role of Isoliquiritigenin in Cancer. Cancers (Basel), 13(1).

  146. Tuli HS, Garg VK, Mehta JK, Kaur G, Mohapatra RK, Dhama K, Sak K, Kumar A, Varol M, Aggarwal D, et al. Licorice (Glycyrrhiza glabra L.)-Derived phytochemicals target multiple signaling pathways to Confer Oncopreventive and Oncotherapeutic effects. Onco Targets Ther. 2022;15:1419–48.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Zhao Y, Han Y, Wang Z, Chen T, Qian H, He J, Li J, Han B, Wang T. (2020) Rosmarinic acid protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurotoxicity in zebrafish embryos. Toxicol in Vitro, 65.

  148. Yu M, Pan Q, Li W, Du T, Huang F, Wu H, He Y, Wu X, Shi H. Isoliquiritigenin inhibits gastric cancer growth through suppressing GLUT4 mediated glucose uptake and inducing PDHK1/PGC-1α mediated energy metabolic collapse. Phytomedicine. 2023;121:155045.

    Article  PubMed  CAS  Google Scholar 

  149. Chaudhry GE, Zeenia, Sharifi-Rad J, Calina D. (2023) Hispidulin: a promising anticancer agent and mechanistic breakthrough for targeted cancer therapy. Naunyn-Schmiedeberg’s archives of pharmacology.

  150. Han M, Gao H, Ju P, Gao MQ, Yuan YP, Chen XH, Liu KL, Han YT, Han ZW. Hispidulin inhibits hepatocellular carcinoma growth and metastasis through AMPK and ERK signaling mediated activation of PPARγ. Biomed Pharmacother. 2018;103:272–83.

    Article  PubMed  CAS  Google Scholar 

  151. Kumar D, Shankar S, Srivastava RK. Rottlerin induces autophagy and apoptosis in prostate cancer stem cells via PI3K/Akt/mTOR signaling pathway. Cancer Lett. 2014;343(2):179–89.

    Article  PubMed  CAS  Google Scholar 

  152. Lu W, Lin C, Li Y. Rottlerin induces wnt co-receptor LRP6 degradation and suppresses both Wnt/β-catenin and mTORC1 signaling in prostate and breast cancer cells. Cell Signal. 2014;26(6):1303–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Iranpanah A, Kooshki L, Moradi SZ, Saso L, Fakhri S, Khan H. The exosome-mediated PI3K/Akt/mTOR signaling pathway in neurological diseases. Pharmaceutics. 2023;15(3):1006.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Ganguly R, Gupta A, Pandey AK. Role of baicalin as a potential therapeutic agent in hepatobiliary and gastrointestinal disorders: a review. World J Gastroenterol. 2022;28(26):3047–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Wang X, Xie L, Long J, Liu K, Lu J, Liang Y, Cao Y, Dai X, Li X. Therapeutic effect of baicalin on inflammatory bowel disease: a review. J Ethnopharmacol. 2022;283:114749.

    Article  PubMed  CAS  Google Scholar 

  156. Wang L, Feng T, Su Z, Pi C, Wei Y, Zhao L. Latest research progress on anticancer effect of baicalin and its aglycone baicalein. Arch Pharm Res. 2022;45(8):535–57.

    Article  PubMed  CAS  Google Scholar 

  157. Liang J, Zhou Y, Cheng X, Chen J, Cao H, Guo X, Zhang C, Zhuang Y, Hu G. (2023) Baicalin attenuates H(2)O(2)-Induced oxidative stress by regulating the AMPK/Nrf2 signaling pathway in IPEC-J2 cells. Int J Mol Sci, 24(11).

  158. You J, Cheng J, Yu B, Duan C, Peng J. Baicalin, a Chinese Herbal Medicine, inhibits the Proliferation and Migration of Human Non-small Cell Lung Carcinoma (NSCLC) cells, A549 and H1299, by activating the SIRT1/AMPK signaling pathway. Med Sci Monit. 2018;24:2126–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Aryal P, Kim K, Park PH, Ham S, Cho J, Song K. Baicalein induces autophagic cell death through AMPK/ULK1 activation and downregulation of mTORC1 complex components in human cancer cells. Febs j. 2014;281(20):4644–58.

    Article  PubMed  CAS  Google Scholar 

  160. Liu B, Ding L, Zhang L, Wang S, Wang Y, Wang B, Li L. Baicalein induces Autophagy and apoptosis through AMPK Pathway in Human Glioma cells. Am J Chin Med. 2019;47(6):1405–18.

    Article  PubMed  CAS  Google Scholar 

  161. Zheng F, Wu J, Zhao S, Luo Q, Tang Q, Yang L, Li L, Wu W, Hann SS. Baicalein increases the expression and reciprocal interplay of RUNX3 and FOXO3a through crosstalk of AMPKα and MEK/ERK1/2 signaling pathways in human non-small cell lung cancer cells. J Exp Clin Cancer Res. 2015;34(1):41.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Huang WW, Tsai SC, Peng SF, Lin MW, Chiang JH, Chiu YJ, Fushiya S, Tseng MT, Yang JS. Kaempferol induces autophagy through AMPK and AKT signaling molecules and causes G2/M arrest via downregulation of CDK1/cyclin B in SK-HEP-1 human hepatic cancer cells. Int J Oncol. 2013;42(6):2069–77.

    Article  PubMed  CAS  Google Scholar 

  163. Hwang JT, Park OJ, Lee YK, Sung MJ, Hur HJ, Kim MS, Ha JH, Kwon DY. Anti-tumor effect of luteolin is accompanied by AMP-activated protein kinase and nuclear factor-κB modulation in HepG2 hepatocarcinoma cells. Int J Mol Med. 2011;28(1):25–31.

    PubMed  CAS  Google Scholar 

  164. Cho AR, Park WY, Lee HJ, Sim DY, Im E, Park JE, Ahn CH, Shim BS, Kim SH. (2021) Antitumor effect of morusin via g1 arrest and antiglycolysis by ampk activation in hepatocellular cancer. Int J Mol Sci, 22(19).

  165. Zhang E, Yin S, Lu X, Ye L, Fan L, Hu H. (2018) Glycycoumarin sensitizes liver cancer cells to ABT-737 by targeting de novo lipogenesis and TOPK-survivin axis. Nutrients, 10(3).

  166. Jia Y, Wu C, Rivera-Piza A, Kim YJ, Lee JH, Lee SJ. (2022) Mechanism of action of cyanidin 3-O-Glucoside in Gluconeogenesis and oxidative stress-Induced Cancer Cell Senescence. Antioxidants, 11(4).

  167. Duan J, Li Y, Gao H, Yang D, He X, Fang Y, Zhou G. Phenolic compound ellagic acid inhibits mitochondrial respiration and tumor growth in lung cancer. Food Funct. 2020;11(7):6332–9.

    Article  PubMed  CAS  Google Scholar 

  168. Luo LX, Li Y, Liu ZQ, Fan XX, Duan FG, Li RZ, Yao XJ, Leung ELH, Liu L. (2017) Honokiol induces apoptosis, G1 arrest, and Autophagy in KRAS Mutant Lung Cancer cells. Front Pharmacol, 8.

  169. Mun JG, Han YH, Jeon HD, Yoon DH, Lee YG, Hong SH, Kee JY. Inhibitory effect of Gallotannin on Lung Metastasis of Metastatic Colorectal Cancer cells by inducing apoptosis, cell cycle arrest and autophagy. Am J Chin Med. 2021;49(06):1535–55.

    Article  PubMed  CAS  Google Scholar 

  170. Qu YQ, Song LL, Xu SW, Yu MSY, Kadioglu O, Michelangeli F, Law BYK, Efferth T, Lam CW, Wong VKW. Pomiferin targets SERCA, mTOR, and P-gp to induce autophagic cell death in apoptosis-resistant cancer cells, and reverses the MDR phenotype in cisplatin-resistant tumors in vivo. Pharmacol Res. 2023;191:106769.

    Article  PubMed  CAS  Google Scholar 

  171. Park GB, Jeong JY, Kim D. Ampelopsin-induced reactive oxygen species enhance the apoptosis of colon cancer cells by activating endoplasmic reticulum stress-mediated AMPK/MAPK/XAF1 signaling. Oncol Lett. 2017;14(6):7947–56.

    PubMed  PubMed Central  Google Scholar 

  172. Tang S, Cai S, Ji S, Yan X, Zhang W, Qiao X, Zhang H, Ye M, Yu S. Isoangustone A induces autophagic cell death in colorectal cancer cells by activating AMPK signaling. Fitoterapia. 2021;152:104935.

    Article  PubMed  CAS  Google Scholar 

  173. Zhao Y, Zhou Y, Wang M. Brosimone I, an isoprenoid-substituted flavonoid, induces cell cycle G(1) phase arrest and apoptosis through ROS-dependent endoplasmic reticulum stress in HCT116 human colon cancer cells. Food Funct. 2019;10(5):2729–38.

    Article  PubMed  CAS  Google Scholar 

  174. Li H, Chen C. Inhibition of autophagy enhances synergistic effects of Salidroside and anti-tumor agents against colorectal cancer. BMC Complement Altern Med. 2017;17(1):538.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Jia S, Xu X, Zhou S, Chen Y, Ding G, Cao L. Fisetin induces autophagy in pancreatic cancer cells via endoplasmic reticulum stress- and mitochondrial stress-dependent pathways. Cell Death Dis. 2019;10(2):142.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Park TH, Kim HS. Eupatilin suppresses pancreatic Cancer cells via glucose uptake inhibition, AMPK activation, and cell cycle arrest. Anticancer Res. 2022;42(1):483–91.

    Article  PubMed  CAS  Google Scholar 

  177. Ye TT, Su JD, Huang CH, Yu DL, Dai SJ, Huang XC, Chen BC, Zhou MT. Isoorientin induces apoptosis, decreases invasiveness, and downregulates VEGF secretion by activating AMPK signaling in pancreatic cancer cells. Oncotargets Therapy. 2016;9:7481–92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Yang T, Xiao Y, Liu S, Luo F, Tang D, Yu Y, Xie Y. Isorhamnetin induces cell cycle arrest and apoptosis by triggering DNA damage and regulating the AMPK/mTOR/p70S6K signaling pathway in doxorubicin-resistant breast cancer. Phytomedicine. 2023;114:154780.

    Article  PubMed  CAS  Google Scholar 

  179. Park C, Cha HJ, Choi EO, Lee H, Hwang-Bo H, Ji SY, Kim MY, Kim SY, Hong SH, Cheong J et al. (2019) Isorhamnetin induces cell cycle arrest and apoptosis Via reactive oxygen species-mediated AMP-Activated protein kinase signaling pathway activation in human bladder Cancer cells. Cancers, 11(10).

  180. Kong Y, Sun W, Wu P. Hyperoside exerts potent anticancer activity in skin cancer. Front Biosci (Landmark Ed). 2020;25(3):463–79.

    Article  PubMed  CAS  Google Scholar 

  181. Li F, Ma Z, Guan Z, Chen Y, Wu K, Guo P, Wang X, He D, Zeng J. Autophagy induction by silibinin positively contributes to its anti-metastatic capacity via AMPK/mTOR pathway in renal cell carcinoma. Int J Mol Sci. 2015;16(4):8415–29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Zhu HY, Huang ZX, Chen GQ, Sheng F, Zheng YS. Typhaneoside prevents acute myeloid leukemia (AML) through suppressing proliferation and inducing ferroptosis associated with autophagy. Biochem Biophys Res Commun. 2019;516(4):1265–71.

    Article  PubMed  CAS  Google Scholar 

  183. Fakhri S, Abdian S, Moradi SZ, Delgadillo BE, Fimognari C, Bishayee A. Marine compounds, mitochondria, and malignancy: a therapeutic Nexus. Mar Drugs. 2022;20(10):625.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for cancer prevention and therapy: current progress and future perspectives. Eur J Pharmacol. 2019;858:172472.

    Article  PubMed  CAS  Google Scholar 

  185. Gao Y, Nie K, Wang H, Dong H, Tang Y. Research progress on antidepressant effects and mechanisms of berberine. Front Pharmacol. 2024;15:1331440.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Khezri MR, Mohammadipanah S, Ghasemnejad-Berenji M. The pharmacological effects of Berberine and its therapeutic potential in different diseases: role of the phosphatidylinositol 3-kinase/AKT signaling pathway. Phytother Res. 2024;38(1):349–67.

    Article  PubMed  CAS  Google Scholar 

  187. Goel A. Current understanding and future prospects on Berberine for anticancer therapy. Chem Biol Drug Des. 2023;102(1):177–200.

    Article  PubMed  CAS  Google Scholar 

  188. Gu S, Song X, Xie R, Ouyang C, Xie L, Li Q, Su T, Xu M, Xu T, Huang D, et al. Berberine inhibits cancer cells growth by suppressing fatty acid synthesis and biogenesis of extracellular vesicles. Life Sci. 2020;257:118122.

    Article  PubMed  CAS  Google Scholar 

  189. Li W, Hua B, Saud SM, Lin H, Hou W, Matter MS, Jia L, Colburn NH, Young MR. Berberine regulates AMP-activated protein kinase signaling pathways and inhibits colon tumorigenesis in mice. Mol Carcinog. 2015;54(10):1096–109.

    Article  PubMed  CAS  Google Scholar 

  190. Park JJ, Seo SM, Kim EJ, Lee YJ, Ko YG, Ha J, Lee M. Berberine inhibits human colon cancer cell migration via AMP-activated protein kinase-mediated downregulation of integrin β1 signaling. Biochem Biophys Res Commun. 2012;426(4):461–7.

    Article  PubMed  CAS  Google Scholar 

  191. Wang J, Qi Q, Feng Z, Zhang X, Huang B, Chen A, Prestegarden L, Li X, Wang J. Berberine induces autophagy in glioblastoma by targeting the AMPK/mTOR/ULK1-pathway. Oncotarget. 2016;7(41):66944–58.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Yang X, Huang N. Berberine induces selective apoptosis through the AMPK–mediated mitochondrial/caspase pathway in hepatocellular carcinoma. Mol Med Rep. 2013;8(2):505–10.

    Article  PubMed  Google Scholar 

  193. Ming M, Sinnett-Smith J, Wang J, Soares HP, Young SH, Eibl G, Rozengurt E. Dose-dependent AMPK-Dependent and independent mechanisms of Berberine and Metformin Inhibition of mTORC1, ERK, DNA synthesis and proliferation in pancreatic Cancer cells. PLoS ONE. 2014;9(12):e114573.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Kim HS, Kim MJ, Kim EJ, Yang Y, Lee MS, Lim JS. Berberine-induced AMPK activation inhibits the metastatic potential of melanoma cells via reduction of ERK activity and COX-2 protein expression. Biochem Pharmacol. 2012;83(3):385–94.

    Article  PubMed  CAS  Google Scholar 

  195. Hu Q, Li L, Zou X, Xu L, Yi P. Berberine attenuated Proliferation, Invasion and Migration by targeting the AMPK/HNF4α/WNT5A pathway in gastric carcinoma. Front Pharmacol. 2018;9:1150.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  196. Hu S, Yin J, Yan S, Hu P, Huang J, Zhang G, Wang F, Tong Q, Zhang Y. Chaetocochin J, an epipolythiodioxopiperazine alkaloid, induces apoptosis and autophagy in colorectal cancer via AMPK and PI3K/AKT/mTOR pathways. Bioorg Chem. 2021;109:104693.

    Article  PubMed  CAS  Google Scholar 

  197. Bao X, Liu Y, Huang J, Yin S, Sheng H, Han X, Chen Q, Wang T, Chen S, Qiu Y, et al. Stachydrine hydrochloride inhibits hepatocellular carcinoma progression via LIF/AMPK axis. Phytomedicine. 2022;100:154066.

    Article  PubMed  Google Scholar 

  198. Kim SY, Hwangbo H, Kim MY, Ji SY, Lee H, Kim GY, Kwon CY, Leem SH, Hong SH, Cheong J, et al. Coptisine induces autophagic cell death through down-regulation of PI3K/Akt/mTOR signaling pathway and up-regulation of ROS-mediated mitochondrial dysfunction in hepatocellular carcinoma Hep3B cells. Arch Biochem Biophys. 2021;697:108688.

    Article  PubMed  CAS  Google Scholar 

  199. Xiang X, Tian Y, Hu J, Xiong R, Bautista M, Deng L, Yue Q, Li Y, Kuang W, Li J et al. (2021) Fangchinoline exerts anticancer effects on colorectal cancer by inducing autophagy via regulation AMPK/mTOR/ULK1 pathway. Biochem Pharmacol, 186.

  200. Chen B, Song Y, Zhan Y, Zhou S, Ke J, Ao W, Zhang Y, Liang Q, He M, Li S et al. (2022) Fangchinoline inhibits non-small cell lung cancer metastasis by reversing epithelial-mesenchymal transition and suppressing the cytosolic ROS-related Akt-mTOR signaling pathway. Cancer Lett, 543.

  201. Yu HI, Shen HC, Chen SH, Lim YP, Chuang HH, Tai TS, Kung FP, Lu CH, Hou CY, Lee YR. (2019) Autophagy modulation in human thyroid Cancer cells following Aloperine Treatment. Int J Mol Sci, 20(21).

  202. Wang F, Cao M, Fan M, Wu H, Huang W, Zhang Y, Hu Z, Jin X. AMPK-mTOR-ULK1 axis activation-dependent autophagy promotes hydroxycamptothecin-induced apoptosis in human bladder cancer cells. J Cell Physiol. 2020;235(5):4302–15.

    Article  PubMed  CAS  Google Scholar 

  203. Song CF, Hu YH, Mang ZG, Ye Z, Chen HD, Jing DS, Fan GX, Ji SR, Yu XJ, Xu XW, et al. Hernandezine induces autophagic cell death in human pancreatic cancer cells via activation of the ROS/AMPK signaling pathway. Acta Pharmacol Sin. 2023;44(4):865–76.

    Article  PubMed  CAS  Google Scholar 

  204. Si Y, Wang J, Liu XW, Zhou T, Xiang YC, Zhang T, Wang XH, Feng TT, Xu L, Yu QQ et al. (2020) Ethoxysanguinarine, a Novel Direct Activator of AMP-Activated protein kinase, induces autophagy and exhibits therapeutic potential in breast Cancer cells. Front Pharmacol, 10.

  205. Pal HC, Prasad R, Katiyar SK. (2017) Cryptolepine inhibits melanoma cell growth through coordinated changes in mitochondrial biogenesis, dynamics and metabolic tumor suppressor AMPKα1/2-LKB1. Sci Rep, 7.

  206. Li H, Zhang C, Zhang M, Yao Q, Yang H, Fan L, Zheng N. (2020) Angustoline inhibited esophageal tumors through regulating LKB1/AMPK/ELAVL1/LPACT2 pathway and phospholipid remodeling. Front Oncol, 10.

  207. Ge D, Tao HR, Fang L, Kong XQ, Han LN, Li N, Xu YX, Li LY, Yu M, Zhang H. 11-Methoxytabersonine induces necroptosis with autophagy through AMPK/mTOR and JNK pathways in Human Lung Cancer cells. Chem Pharm Bull (Tokyo). 2020;68(3):244–50.

    Article  PubMed  CAS  Google Scholar 

  208. Wróblewska-Łuczka P, Cabaj J, Bargieł J, Łuszczki JJ. Anticancer effect of terpenes: focus on malignant melanoma. Pharmacol Rep. 2023;75(5):1115–25.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Huang M, Lu J-J, Huang M-Q, Bao J-L, Chen X-P, Wang Y-T. Terpenoids: natural products for cancer therapy. Expert Opin Investig Drugs. 2012;21(12):1801–18.

    Article  PubMed  CAS  Google Scholar 

  210. Salminen A, Lehtonen M, Suuronen T, Kaarniranta K, Huuskonen J. Terpenoids: natural inhibitors of NF-κB signaling with anti-inflammatory and anticancer potential. Cell Mol Life Sci. 2008;65:2979–99.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Zhong ZF, Tan W, Qiang WW, Scofield VL, Tian K, Wang CM, Qiang WA, Wang YT. Furanodiene alters mitochondrial function in doxorubicin-resistant MCF-7 human breast cancer cells in an AMPK-dependent manner. Mol Biosyst. 2016;12(5):1626–37.

    Article  PubMed  CAS  Google Scholar 

  212. Zhao S, Wu J, Zheng F, Tang Q, Yang L, Li L, Wu W, Hann SS. β-elemene inhibited expression of DNA methyltransferase 1 through activation of ERK1/2 and AMPKα signalling pathways in human lung cancer cells: the role of Sp1. J Cell Mol Med. 2015;19(3):630–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Shanmugam MK, Dai X, Kumar AP, Tan BK, Sethi G, Bishayee A. Ursolic acid in cancer prevention and treatment: molecular targets, pharmacokinetics and clinical studies. Biochem Pharmacol. 2013;85(11):1579–87.

    Article  PubMed  CAS  Google Scholar 

  214. Yie Y, Zhao S, Tang Q, Zheng F, Wu J, Yang L, Deng S, Hann SS. Ursolic acid inhibited growth of hepatocellular carcinoma HepG2 cells through AMPKα-mediated reduction of DNA methyltransferase 1. Mol Cell Biochem. 2015;402(1–2):63–74.

    Article  PubMed  CAS  Google Scholar 

  215. Lewinska A, Adamczyk-Grochala J, Kwasniewicz E, Deregowska A, Wnuk M. Ursolic acid-mediated changes in glycolytic pathway promote cytotoxic autophagy and apoptosis in phenotypically different breast cancer cells. Apoptosis. 2017;22(6):800–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Zhao F, Huang W, Zhang Z, Mao L, Han Y, Yan J, Lei M. Triptolide induces protective autophagy through activation of the CaMKKβ-AMPK signaling pathway in prostate cancer cells. Oncotarget. 2016;7(5):5366–82.

    Article  PubMed  Google Scholar 

  217. Ren T, Tang YJ, Wang MF, Wang HS, Liu Y, Qian X, Chang C, Chen MW. Triptolide induces apoptosis through the calcium/calmodulin–dependent protein kinase kinaseβ/AMP–activated protein kinase signaling pathway in non–small cell lung cancer cells. Oncol Rep. 2020;44(5):2288–96.

    PubMed  CAS  Google Scholar 

  218. Kang JI, Hong JY, Lee HJ, Bae SY, Jung C, Park HJ, Lee SK. (2015) Anti-tumor activity of yuanhuacine by regulating AMPK/mTOR Signaling pathway and actin cytoskeleton organization in non- small cell lung cancer cells. PLoS ONE, 10(12).

  219. Younis T, Khan MI, Khan MR, Rasul A, Majid M, Adhami VM, Mukhtar H. Nummularic acid, a triterpenoid, from the medicinal plant Fraxinus xanthoxyloides, induces energy crisis to suppress growth of prostate cancer cells. Mol Carcinog. 2018;57(10):1267–77.

    Article  PubMed  CAS  Google Scholar 

  220. Wang SR, Ma K, Zhou CQ, Wang Y, Hu GH, Chen LC, Li Z, Hu CF, Xu Q, Zhu HX et al. (2019) LKB1 and YAP phosphorylation play important roles in Celastrol-induced β-catenin degradation in colorectal cancer. Therapeutic Adv Med Oncol, 11.

  221. Kim JH, Lee JO, Lee SK, Kim N, You GY, Moon JW, Sha J, Kim SJ, Park SH, Kim HS. Celastrol suppresses breast cancer MCF-7 cell viability via the AMP-activated protein kinase (AMPK)-induced p53-polo like kinase 2 (PLK-2) pathway. Cell Signal. 2013;25(4):805–13.

    Article  PubMed  CAS  Google Scholar 

  222. Liu T, Li Y, Sun J, Tian G, Shi Z. Gitogenin suppresses lung cancer progression by inducing apoptosis and autophagy initiation through the activation of AMPK signaling. Int Immunopharmacol. 2022;111:108806.

    Article  PubMed  CAS  Google Scholar 

  223. Liu J, Zheng L, Wu N, Ma L, Zhong J, Liu G, Lin X. Oleanolic acid induces metabolic adaptation in cancer cells by activating the AMP-activated protein kinase pathway. J Agric Food Chem. 2014;62(24):5528–37.

    Article  PubMed  CAS  Google Scholar 

  224. Chen L, Fang W, Liu J, Qi X, Zhao L, Wang Y, Liu Y, Kong D, Sun X, Li X, et al. Poricoic acid A (PAA) inhibits T-cell acute lymphoblastic leukemia through inducing autophagic cell death and ferroptosis. Biochem Biophys Res Commun. 2022;608:108–15.

    Article  PubMed  CAS  Google Scholar 

  225. Akhtar N, Syed DN, Khan MI, Adhami VM, Mirza B, Mukhtar H. The pentacyclic triterpenoid, plectranthoic acid, a novel activator of AMPK induces apoptotic death in prostate cancer cells. Oncotarget. 2016;7(4):3819–31.

    Article  PubMed  Google Scholar 

  226. Zhou X, Kang J, Zhang L, Cheng Y. Osthole inhibits malignant phenotypes and induces ferroptosis in KRAS-mutant colorectal cancer cells via suppressing AMPK/Akt signaling. Cancer Chemother Pharmacol. 2023;92(2):119–34.

    Article  PubMed  CAS  Google Scholar 

  227. Huang H, Xue J, Xie T, Xie ML. Osthole increases the radiosensitivity of hepatoma cells by inhibiting GSK-3β/AMPK/mTOR pathway-controlled glycolysis. Naunyn Schmiedebergs Arch Pharmacol. 2023;396(4):683–92.

    Article  PubMed  CAS  Google Scholar 

  228. Huang H, Xue J, Xie ML, Xie T. (2023) Osthole inhibits GSK-3β/AMPK/mTOR pathway-controlled glycolysis and increases radiosensitivity of subcutaneous transplanted hepatocellular carcinoma in nude mice. Strahlenther Onkol.

  229. Gao J, Song L, Xia H, Peng L, Wen Z. (2020) 6′-O-galloylpaeoniflorin regulates proliferation and metastasis of non-small cell lung cancer through AMPK/miR-299-5p/ATF2 axis. Respir Res, 21(1).

  230. Li X, Tang X, Su J, Xu G, Zhao L, Qi Q. Involvement of E-cadherin/AMPK/mTOR axis in LKB1-induced sensitivity of non-small cell lung cancer to gambogic acid. Biochem Pharmacol. 2019;169:113635.

    Article  PubMed  CAS  Google Scholar 

  231. Li YM, Liu H, Liu XX, Xiao B, Zhang MH, Luo YL, Li MC, Yang JQ. (2022) Gracillin shows potential efficacy against Non-small Cell Lung Cancer through inhibiting the mTOR pathway. Front Oncol, 12.

  232. Witayateeraporn W, Nguyen HM, Ho DV, Nguyen HT, Chanvorachote P, Vinayanuwattikun C, Pongrakhananon V. (2022) Aspiletrein A induces apoptosis cell death via increasing reactive Oxygen species Generation and AMPK activation in Non-small-cell Lung Cancer cells. Int J Mol Sci, 23(16).

  233. Zhu L, Wang Y, Lv W, Wu X, Sheng H, He C, Hu J. (2021) Schizandrin A can inhibit non–small cell lung cancer cell proliferation by inducing cell cycle arrest, apoptosis and autophagy. Int J Mol Med, 48(6).

  234. Ma J, Duan W, Han S, Lei J, Xu Q, Chen X, Jiang Z, Nan L, Li J, Chen K, et al. Ginkgolic acid suppresses the development of pancreatic cancer by inhibiting pathways driving lipogenesis. Oncotarget. 2015;6(25):20993–1003.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Xie G, Sun L, Li Y, Chen B, Wang C. Periplocin inhibits the growth of pancreatic cancer by inducing apoptosis via AMPK-mTOR signaling. Cancer Med. 2021;10(1):325–36.

    Article  PubMed  CAS  Google Scholar 

  236. Lai SL, Mustafa MR, Wong PF. Panduratin A induces protective autophagy in melanoma via the AMPK and mTOR pathway. Phytomedicine. 2018;42:144–51.

    Article  PubMed  CAS  Google Scholar 

  237. Qiu Y, Li C, Zhang B, Gu Y. (2022) Bixin Prevents Colorectal Cancer Development through AMPK-Activated Endoplasmic Reticulum Stress. BioMed Res Int, 2022.

  238. Shin EJ, Choi HK, Sung MJ, Park JH, Chung MY, Chung S, Hwang JT. Anti-tumour effects of beta-sitosterol are mediated by AMPK/PTEN/HSP90 axis in AGS human gastric adenocarcinoma cells and xenograft mouse models. Biochem Pharmacol. 2018;152:60–70.

    Article  PubMed  CAS  Google Scholar 

  239. Taş İ, Varlı M, Son Y, Han J, Kwak D, Yang Y, Zhou R, Gamage CDB, Pulat S, Park SY, et al. Physciosporin suppresses mitochondrial respiration, aerobic glycolysis, and tumorigenesis in breast cancer. Phytomedicine. 2021;91:153674.

    Article  PubMed  Google Scholar 

  240. Verrelli D, Dallera L, Stendardo M, Monzani S, Pasqualato S, Giorgio M, Pallavi R. (2022) Hydroxycitric Acid inhibits chronic myelogenous leukemia growth through activation of AMPK and mTOR Pathway. Nutrients, 14(13).

  241. Zhao W, Peng F, Shu M, Liu H, Hou X, Wang X, Ye J, Zhao B, Wang K, Zhong C, et al. Isogambogenic acid inhibits the growth of Glioma through activation of the AMPK-mTOR pathway. Cell Physiol Biochemistry: Int J Experimental Cell Physiol Biochem Pharmacol. 2017;44(4):1381–95.

    Article  CAS  Google Scholar 

  242. Solanki R, Jodha B, Prabina KE, Aggarwal N, Patel S. (2022) Recent advances in phytochemical based nano-drug delivery systems to combat breast cancer: a review. J Drug Deliv Sci Technol:103832.

  243. Jeetah R, Bhaw-Luximon A, Jhurry D. Nanopharmaceutics: phytochemical-based controlled or sustained drug-delivery systems for cancer treatment. J Biomed Nanotechnol. 2014;10(9):1810–40.

    Article  PubMed  CAS  Google Scholar 

  244. Kashyap D, Tuli HS, Yerer MB, Sharma A, Sak K, Srivastava S, Pandey A, Garg VK, Sethi G, Bishayee A. Natural product-based nanoformulations for cancer therapy: opportunities and challenges. Sem Cancer Biol. 2021;69:5–23.

    Article  CAS  Google Scholar 

  245. Lagoa R, Silva J, Rodrigues JR, Bishayee A. Advances in phytochemical delivery systems for improved anticancer activity. Biotechnol Adv. 2020;38:107382.

    Article  PubMed  CAS  Google Scholar 

  246. Sinha D, Roy S, Saha P, Chatterjee N, Bishayee A. (2021) Trends in Research on Exosomes in Cancer Progression and Anticancer Therapy. Cancers (Basel), 13(2).

  247. Maleki Dizaj S, Alipour M, Dalir Abdolahinia E, Ahmadian E, Eftekhari A, Forouhandeh H, Rahbar Saadat Y, Sharifi S, Zununi Vahed S. Curcumin nanoformulations: beneficial nanomedicine against cancer. Phytother Res. 2022;36(3):1156–81.

    Article  PubMed  CAS  Google Scholar 

  248. Massironi A, Marzorati S, Marinelli A, Toccaceli M, Gazzotti S, Ortenzi MA, Maggioni D, Petroni K, Verotta L. Synthesis and characterization of curcumin-loaded nanoparticles of poly (glycerol sebacate): a novel highly stable anticancer system. Molecules. 2022;27(20):6997.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  249. Vieira IRS, Conte-Junior CA. (2022) Nano-delivery systems for food bioactive compounds in cancer: Prevention, therapy, and clinical applications. Crit Rev Food Sci Nutr:1–26.

  250. Yaghoubi F, Motlagh NSH, Naghib SM, Haghiralsadat F, Jaliani HZ, Moradi A. A functionalized graphene oxide with improved cytocompatibility for stimuli-responsive co-delivery of curcumin and doxorubicin in cancer treatment. Sci Rep. 2022;12(1):1959.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  251. (!!!. INVALID CITATION !!!).

  252. Hashemzehi M, Behnam-Rassouli R, Hassanian SM, Moradi‐Binabaj M, Moradi‐Marjaneh R, Rahmani F, Fiuji H, Jamili M, Mirahmadi M, Boromand N. Phytosomal‐curcumin antagonizes cell growth and migration, induced by thrombin through AMP‐Kinase in breast cancer. J Cell Biochem. 2018;119(7):5996–6007.

    Article  PubMed  CAS  Google Scholar 

  253. Tong W, Wang Q, Sun D, Suo J. Curcumin suppresses colon cancer cell invasion via AMPK-induced inhibition of NF-κB, uPA activator and MMP9. Oncol Lett. 2016;12(5):4139–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  254. Granja A, Pinheiro M, Reis S. Epigallocatechin gallate nanodelivery systems for cancer therapy. Nutrients. 2016;8(5):307.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Farabegoli F, Pinheiro M. Epigallocatechin-3-Gallate delivery in lipid-based nanoparticles: potentiality and perspectives for future applications in cancer chemoprevention and therapy. Front Pharmacol. 2022;13:809706.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  256. Li K, Teng C, Min Q. Advanced nanovehicles-enabled delivery systems of epigallocatechin gallate for cancer therapy. Front Chem. 2020;8:573297.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  257. Saha S, Prajapati DG, Ratrey P, Mishra A. Co-delivery nanosystem of Epigallocatechin Gallate and Rutin for anticancer and antibacterial activities. J Drug Deliv Sci Technol. 2022;70:103191.

    Article  CAS  Google Scholar 

  258. Lee DG, Lee M, Go EB, Chung N. Resveratrol-loaded gold nanoparticles enhance caspase-mediated apoptosis in PANC-1 pancreatic cells via mitochondrial intrinsic apoptotic pathway. Cancer Nanotechnol. 2022;13(1):1–19.

    Article  Google Scholar 

  259. Ferreira M, Costa D, Sousa Â. Flavonoids-based delivery systems towards cancer therapies. Bioengineering. 2022;9(5):197.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Peng B, Zhang S-Y, Chan KI, Zhong Z-F, Wang Y-T. Novel anti-cancer products targeting AMPK: natural herbal medicine against breast cancer. Molecules. 2023;28(2):740.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  261. Wang H, Zheng Y, Sun Q, Zhang Z, Zhao M, Peng C, Shi S. Ginsenosides emerging as both bifunctional drugs and nanocarriers for enhanced antitumor therapies. J Nanobiotechnol. 2021;19:1–40.

    Article  Google Scholar 

  262. Jin Y, Huynh DTN, Nguyen TLL, Jeon H, Heo K-S. Therapeutic effects of ginsenosides on breast cancer growth and metastasis. Arch Pharm Res. 2020;43:773–87.

    Article  PubMed  CAS  Google Scholar 

  263. Hong C, Wang D, Liang J, Guo Y, Zhu Y, Xia J, Qin J, Zhan H, Wang J. Novel ginsenoside-based multifunctional liposomal delivery system for combination therapy of gastric cancer. Theranostics. 2019;9(15):4437.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  264. Wang H, Zheng Y, Sun Q, Zhang Z, Zhao M, Peng C, Shi S. Ginsenosides emerging as both bifunctional drugs and nanocarriers for enhanced antitumor therapies. J Nanobiotechnol. 2021;19(1):322.

    Article  CAS  Google Scholar 

  265. Jeon H, Jin Y, Myung CS, Heo KS. Ginsenoside-Rg2 exerts anti-cancer effects through ROS-mediated AMPK activation associated mitochondrial damage and oxidation in MCF-7 cells. Arch Pharm Res. 2021;44(7):702–12.

    Article  PubMed  CAS  Google Scholar 

  266. Jeon H, Huynh DTN, Baek N, Nguyen TLL, Heo KS. Ginsenoside-Rg2 affects cell growth via regulating ROS-mediated AMPK activation and cell cycle in MCF-7 cells. Phytomedicine. 2021;85:153549.

    Article  PubMed  CAS  Google Scholar 

  267. Law BYK, Michelangeli F, Qu YQ, Xu SW, Han Y, Mok SWF, Dias I, Javed MU, Chan WK, Xue WW, et al. Neferine induces autophagy-dependent cell death in apoptosis-resistant cancers via ryanodine receptor and ca(2+)-dependent mechanism. Sci Rep. 2019;9(1):20034.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

SF: Conceptualization, Methodology, Data Curation, Writing – Original Draft, and Writing – Review & Editing; SZM: Methodology, Data Curation, and Writing – Original Draft; SYM, SP (Sarina Piri), BSV, SP (Sana Piri), and MRK: Writing – Original Draft; AB (Ankur Bishayee): Writing – Original Draft and Writing – Review & Editing; NC: Writing – Review & Editing; AB (Anupam Bishayee): Conceptualization, Writing – Original Draft, Writing – Review & Editing; Supervision, Project administration. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Sajad Fakhri or Anupam Bishayee.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fakhri, S., Moradi, S.Z., Moradi, S.Y. et al. Phytochemicals regulate cancer metabolism through modulation of the AMPK/PGC-1α signaling pathway. BMC Cancer 24, 1079 (2024). https://doi.org/10.1186/s12885-024-12715-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12715-7

Keywords