Next Article in Journal
Chitosan-Based Ciprofloxacin Extended Release Systems: Combined Synthetic and Pharmacological (In Vitro and In Vivo) Studies
Next Article in Special Issue
Antifungal Effect and Inhibition of the Virulence Mechanism of D-Limonene against Candida parapsilosis
Previous Article in Journal
Phenotypic and Target-Directed Screening Yields New Acaricidal Alternatives for the Control of Ticks
Previous Article in Special Issue
Pharmacological and Clinical Efficacy of Picrorhiza kurroa and Its Secondary Metabolites: A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Importance of Kaempferol in the Treatment of Cancer through the Modulation of Cell Signalling Pathways

by
Malak Yahia Qattan
1,
Mohammad Idreesh Khan
2,
Shudayyed Hasham Alharbi
3,4,
Amit Kumar Verma
5,*,
Fatimah A. Al-Saeed
6,7,
Alduwish Manal Abduallah
8 and
Azza A. Al Areefy
9,10
1
Department of Health Sciences, College of Applied Studies and Community Service, King Saud University, KSA- 4545, Riyadh 11451, Saudi Arabia
2
Department of Clinical Nutrition, College of Applied Health Sciences in Ar Rass, Qassim University, Ar Rass 51921, Saudi Arabia
3
Pharmacy Department, Maternity and Children Hospital (MCH), Qassim Cluster, Ministry of Health, Buraydah 52384, Saudi Arabia
4
Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
5
Department of Biotechnology, Jamia Millia Islamia University, New Delhi 110025, India
6
Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
7
Research Centre for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
8
Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Alkarj 11942, Saudi Arabia
9
Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
10
Nutrition & Food Science Department, Faculty of Home Economics, Helwan University, P.O. Box 11795, Cairo 11281, Egypt
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(24), 8864; https://doi.org/10.3390/molecules27248864
Submission received: 9 November 2022 / Revised: 8 December 2022 / Accepted: 9 December 2022 / Published: 13 December 2022
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)

Abstract

:
Plant-derived flavonoids are considered natural nontoxic chemo-preventers and have been widely studied for cancer treatment in recent decades. Mostly all flavonoid compounds show significant anti-inflammatory, anticancer and antioxidant properties. Kaempferol (Kmp) is a well-studied compound and exhibits remarkable anticancer and antioxidant potential. Kmp can regulate various cancer-related processes and activities such as cell cycle, oxidative stress, apoptosis, proliferation, metastasis, and angiogenesis. The anti-cancer properties of Kmp primarily occur via modulation of apoptosis, MAPK/ERK1/2, P13K/Akt/mTOR, vascular endothelial growth factor (VEGF) signalling pathways. The anti-cancer property of Kmp has been recognized in several in-vivo and in-vitro studies which also includes numerous cell lines and animal models. This flavonoid possesses toxic activities against only cancer cells and have restricted toxicity on healthy cells. In this review, we present extensive research investigations about the therapeutic potential of Kmp in the management of different types of cancers. The anti-cancer properties of Kmp are discussed by concentration on its capability to target molecular-signalling pathway such as VEGF, STAT, p53, NF-κB and PI3K-AKT signalling pathways. The anti-cancer property of Kmf has gained a lot of attention, but the accurate action mechanism remains unclear. However, this natural compound has a great pharmacological capability and is now considered to be an alternative cancer treatment.

1. Introduction

The cancer prevalence and incidence of harmful disorders is increasing among developing and developed nations [1]. GLOBOCAN 2020 data reported 19.3 million new cancer cases and around 10 million cancer related deaths across the world [2]. Though the advancement in treatment and diagnostic methods and cancer-awareness programmes have caused incredible decrease in mortality of cancer in United States, but the prevalence of cancer is yet increasing unceasingly [1]. Intake of unhealthy diet and alcohol, stress, physical inactivity and smoking are the key factors responsible for cancer prevalence in under developed, developing and developed nations [3]. Chemo- and radio- therapy, targeted treatment, surgery, and biological therapy are widely used therapies used in the treatment of cancer from decades. KRAS inhibitors such as cetuximab in colorectal carcinoma, inhibitors of epidermal growth factor receptor such as fefitinib, osimertinib, afatinib and erlotinib in non-small-cell lung carcinoma (NSCLC), BCR-ABL tyrosine kinase inhibitors such as Imatinib mesylate in leukaemia, BRAF inhibitors such as encorafenib, dabrafenib and vemurafenib in melanomas, herceptin and tamoxifen in breast cancers have been extensively utilised in treatment of different cancer types [4]. Cancerous cells can escape mortality by achieving resistance to several therapeutic methods, and this can attenuate the projected outcomes of the cancer treatments [5,6,7]. Nowadays actively goes search of new natural compounds with anticancer potential as well. New and alternate therapeutic methods have been required to treat patients of cancer. Several in-vitro research in combination of ex-vivo research have demonstrated the anticancer impacts of natural substances like flavonoids [8,9,10,11]. In a study it was found that dietary intakes of flavonols, including kaempferol, quercetin, myricetin, and isorhamnetin may be associated with reduced risk of developing Alzheimer dementia [12]. Annona purpurea contains five acetogenins named annopurpuricins A–E, which are active against tumoural cell lines in a subnanomolar range. Fagopyrin is a naphthodianthrone found in Buckwheat (Fagopyrum esculentum L.) have antioxidant and anticancerogenic activity.
One of the major component in flavonoids among plants are quercetin (Qu), kaempferol (Kmp) [13,14] which have anti-oxidant and anti-inflammatory property [15]. Quercetin and kaempferol are widely distributed in fruit and vegetables. High concentrations of quercetin are found in a few foods such as onion, asparagus, and berries, and small quantities are found in many different fruit and vegetables. The richest plant sources of kaempferol (mg/100 g fresh weight) are green leafy vegetables, including spinach and kale, and herbs such as dill, chives, and tarragon [16]. Kmp has been overlooked by the researchers because of a very few in-vitro researches have evaluated the potential of Kmp in treatment of cancers. In very recent times, few studies reported that antioxidant property of flavonoids offered novel therapeutic approaches for chemotherapy in treatment of cancer. Some studies described that Kmp-stimulate activation of antioxidants that might play a vital function in NSCLC-H460 cancerous cell apoptosis [17]. Numerous researchers have proven the antioxidant, anticarcinogenic, antidiabetic, cardioprotective, neuroprotective and antimicrobial properties of kaempferol and its glycosides [18]. Additionally, several research stated that Kmp considerably prevents the cancerous growth in in-vitro conditions which leads to apoptosis in cancerous cells [19,20,21].
Many cancer-related molecules like matrix metallopeptidases (MMPs), proapoptotic and anti-apoptotic proteins, various growth factors, cyclins, and cyclin-dependent kinases (CDKs) have been demonstrated to be regulated by Kmp [22,23,24,25,26,27]. Moreover, Kmp also exhibits synergetic effect where it enhances the anti-tumor activities of several anticancer drugs.
Kmp is (3,4′,5,7-tetrahydroxyflavone) is a yellowish flavonoid compound which has 4 hydroxy-groups at -3, -4′, -5 and -7 positions (Figure 1) [28]. It can be present in several parts of plants like leaves, flowers, seeds, vegetables, and fruits [25,27,29]. Kmp and its glycosides have antidiabetic, anticancer, neuro protective, antioxidant, antitumor, anti-inflammatory, anti-microbial and cardio protective properties [30]. Epidemiologic researches exhibited that greater consumption of Kmp is linked with reduced occurrence of various cancers including colorectal, hepatic, pancreatic, urinary bladder, ovarian, and gastric cancers [28,29]. Due to several anticancer activities, intake of Kmp and associated uses are getting vast interest among researchers in the cancer treatment [31,32,33]. Inhibition of cancerous cells growth is generally attained by preventing cancerous cells proliferation via enhancing apoptosis [34,35,36]. Certainly, Kmp prevents growth of cancerous cells by initiating G2-M stage cell-cycle arrest, apoptosis, down-regulation of signalling pathways such as phosphatidylinositol 3-kinase-protein kinase B (PI3K-PKB), epithelial mesenchymal transition (EMT) markers’ expression such as SNAI1, E-cadherin and N-cadherin and (MMP2) markers [37,38]. Kmp also stimulates the activation of caspases such as caspase-9, caspase-7, caspase-3 and poly (ADP-ribose) polymerase (PARP) proteins which involve in initiation and execution of apoptosis [39], hence, inhibiting the reactive oxygen species (ROS) accumulation included in the development of cancer [40]. It has been also stated that Kmp can maintain normal viability of cells and prevent angiogenesis [40].
Due to effective role in cancer inhibition, extensive pharmacological properties and numerous health promoting benefits of Kmp, this current review article provides a collective compendium of wide-ranging studies examining the potential therapeutic role of Kmp in treatment of several cancer types. Since Kmp asserted to carry the anti-inflammatory, antioxidant and anti-tumor properties and capability to prevent the cancerous cells proliferation, it has been widely studied as a chemo preventive agent in numerous cancerous models and Kmp modulates several cell signalling molecules. Figure 2 graphical representation of literature review.

2. Major Mechanisms of Kaempferol (Kmp) in Management of Cancer

2.1. Inflammation

Inflammation is a biologically complex protective body’s reaction rising due to dangerous stimuli and damaged cells. Many diseases are characterized by inflammation including allergy, transplant rejection, preperfusion injury, hepatitis, glomerulonephritis, asthma, autoimmune disorders, celiac disease, intestinal inflammation and cancer [41]. Hence, inflammation is a biologically, self-protecting body’s reaction in times of problem which eliminates injured and damaged cells and starts the healing process [42]. It has been suggested that chronic inflammation is linked with the progression of several disorders such as neurodegeneration, cancer, and arthritis [43,44,45]. Kmp has been recognised as an effective inhibitor of pro-inflammatory molecules including vascular cell adhesion protein 1, prostaglandin-endoperoxide synthase (PTGS) and inducible nitric oxide synthase (NOSII) [46,47]. Anti-inflammatory effects of Kmp are mostly facilitated by downregulation of numerous sequence-specific DNA-binding factors like STAT, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which have the capability to encourage the pro-inflammatory cytokines activation [48]. A study analysed the anti-inflammatory property of Kmp in hepatic cell lines and found that Kmp reduced the PTGS, NOSII, C-reactive protein (CRP) expression by altering NF-κB signalling pathway [49]. The ability of Kmp to deal with inflammation is one of its critical and considerable features in cancer prevention (Table 1). When lipopolysaccharide-induced macrophages treated with Kmp, it resulted into downregulation of PTGS, NOSII and tumor necrosis factor-alpha (TNF-alpha) at translational- and transcriptional- levels through inhibiting sequence-specific DNA-binding factors such as Activator protein 1 (AP1) and NF-κB [50,51]. Furthermore, protein-kinase signalling cascades mechanism directed by interleukin-1 receptor-associated kinase (IRAK)-1, -4, Syk and Src which are generally take part in AP1 and NF-κB factors activation and could prevented by Kmp [52]. In diseases like Crohn’s disease or rheumatoid arthritis, uncontrolled inflammation can cause immune system arrest where immune system harms normal healthy cells. Chronic inflammation is associated with a susceptibility in development of cancer [53]. Stomach ulcer is linked with an increasing risk of peptic cancer and mesothelioma can be tracked back to irritation caused by asbestos. It has been reported that flavonoid (particularly Kmp) rich diet is correlated with decreased level of serum interleukin-6 which is an inflammatory cytokine [54]. In aldosterone induced human umbilical-vein endothelial cell (HUVEC), Kmp has been reported to downregulate the expression of ROS-dependent cytokines such as osteopontin which activates and stimulates NF-κB and p38-mitogen-activated protein kinases (p38-MAPK) signalling [47]. Hence, studies are recommending Kmp as a promising anti-inflammatory drug and it can be proposed for in-vivo trials.

2.2. Reactive Oxygen Species (ROS)

Metabolic pathways generate ROS in the body which are key resource of destructive oxidative stress [55,56]. Though humans have antioxidant enzymes as defence mechanisms which continuously neutralises ROS, but high ROS concentration causes infections, senescence, cerebrovascular accident, autoimmune disorders, cardiovascular arteriosclerosis, oxygen poisoning, Parkinson’s disorder and becomes lethal [55,57]. Studies suggested that flavonoids can be efficient secondary-metabolites against oxidative stress-related diseases [58]. Kmp increases the anti-oxidant enzymes expressions at high concertation and at low-concentration it scavenges hydroxyl (OH) radical and peroxonitrite radical [30]. The antioxidant property of Kmp is linked with its up regulatory effects on antioxidant-response element- (ARE) meditative anti-oxidative enzymes like superoxide dismutase, catalase and haem oxygenase in control of Nuclear factor erythroid 2-related factor 2 signalling pathway [59]. Kmp can be used in prevention of susceptibility of oxidation of low density lipoproteins (LDL) and aggregation of platelets [60]. Both Wahab et al. 2014 and Choe et al. 2012 studied antioxidant property of Kmp by extracting and purifying Kmp from Senna alata beans and Rhodiola sachalinensis roots respectively [61,62]. It has been observed that Kmp reduced the thiobarbituric-acid reactive substances and red blood corpuscles lysates and upregulated the level of enzymatic antioxidants such as superoxide dismutase, glutathione perxidases (GSHPx) and catalase when 1,2-dimethylehydrazine (DMH)-induced-colon cancer male Wistar-rats treated with Kmp [63]. Similarly, researchers studied hepatoprotective effects of Kmp by increasing in carbon tetrachloride (CCl4)-induced liver damage in rodents [64,65]. Kmp reduces the level of reactive oxygen and increases the survival of cell in oxidatively stressed HT 22 neuronal cells and reduces oxidative DNA-damage in isolated human lymphocytes [66].

2.3. Angiogenesis

Cancerous cells also need nutrients and oxygen to survive provided with networks of capillaries. Angiogenesis is linked with repair of damaged cells and reproductive development via formation of new capillaries which is mediated by growth molecules, endostatins, adhesion molecules etc. [67]. Main mediator in angiogenesis is VEGF and formation of new capillaries aimed to meet increasing requirements of the tumor [68]. Current studies have demonstrated the efficiency of Kmp in reducing angiogenesis of cancer in in-vitro and in-vivo by preventing secretion of VEGF in human cancerous cells [69,70]. A study reported that Kmp prevented VEGF secretion in MDA-MB-231 cancerous cells and decreased the concentration of VEGF-mRNA among ovarian cancerous cell lines [71]. Level of VEGF proteins was significantly influenced by Kmp, indicating action-mechanism involved in translation [67]. Kmp inhibits angiogenesis and expression of VEGF via ERK-NFkappaB-cMyc-p21 pathways [70]. Administered Kmp inhibited expression of NF-κB, c-Myc and phosphorylation of ERK and reduction of these encourages expression of p21 which antagonizes the release of VEGF [68]. Moreover, Kmp also affected regulators of VEGF. Kmp reduces the level of hypoxia inducible factor (HIF)-1 and inhibits phosphorylation of AKT signalling pathway and it blocks signalling mechanisms which involves in enhanced VEGF secretion [67]. Kmp also inhibits activity of estrogen related receptor alpha (ESRRA) by reducing its mRNA level. ESRRA is linked with oestrogen-activity and considered as a cancer promoter. Kmp is an opponent of VEGF and attacks production of VEGF from every path (Figure 3) [23].

2.4. Signal Transduction

Numerous interleukin-6 related signalling pathways have been linked and found with increased migration, invasion, and proliferation of several tumor cells. Interleukin-6 binds with interleukin-6 specific binding receptor-α and activates the dimerization of signal-transducer receptor called glycoprotein 130 and causes its phosphorylation, followed by Janus tyrosin kinase (JAK) activation [72]. These incidents cause the activation of several signal-transduction pathways, like signal-transducer and activator of transcription (STAT), PI-3 kinase signalling pathways [72]. In all these pathways, STAT3 signalling pathway is the mostly analysed/studied cytokine-signalling pathway [73,74]. STAT3 is a member of STAT-family of transcription-factors and plays an important role in cancer related inflammation. STAT3 is often de-regulated in several kinds of cancer and function as an onco-gene in tumorigenesis [75]. STAT3 activation causes expression of down-stream genes which regulate main cell responses (includes survival of cancerous cell, cell invasion and proliferation) like BCL2, cyclin-D1 and MMP-2 [76]. STAT3 plays important role in tumorigenesis and in progression of cancer which allow STAT3 to arise as a promising molecule target in the treatment of cancer. Basu et al. (2020) observed that at high concentration, Kmp prevented interleukin-6 induced-phosphorylation of STAT3 [77]. A study conducted by Yang et al. (2019) concluded that Kmp inhibits STAT3 signalling pathway [78].
Phosphatidylinositide-3-Kinase (PI3K) is an important signal-transducing enzyme which regulates cell differentiation, survival, angiogenesis, proliferation, and apoptosis [79,80]. It is vital for AKT activation and has an important role in pathological as well as physiological signalling processes. Due to the repeated activation of PI3K-AKT mechanism in cancer, it is a key drug target [81,82,83,84,85]. PI3K is a lipid-kinase which causes phosphorylation PIP2–PIP3 and it is the PDK and AKT activation site. Family of PI3K has three different classes viz, class I, II and III and these classes are different in distribution of tissue, in function, preference of substrate, activation pathway and structure [86,87,88]. PI3K-dependent AKT activation results into multi step method which involve both phosphorylation as well as translocation [89]. Activation of AKT includes the phosphorylation of two residues: serine 473 (Ser473) at carboxy-terminal and threonine 308 (Thr308) on activation loop. Ser473 is phosphorylated by PDK2 while PDK1 phosphorylated by Thr308 [90,91]. PDK1 is an important kinase needed for normal development in mammals [92]. AKT has three isoforms: AKT-1, AKT-2 and AKT-3 based on their different biological activities and distribution of tissue. AKT-1 plays a vital role in angiogenesis and cell survival regulation [86,93,94]. PI3K activation is counter-production to apoptotic pathway and due to this, several drugs related to cancer treatment concentrate on inhibition of this pathway. Chin et al. (2018) reported that Kmp in dosage-dependent manner significantly reduced the mTOR and AKT phosphorylation and level of PI3K protein [95]. Another study reported that Kmp repressed the growth of colorectal cancerous cells by preventing the activation of PI3K-AKT signalling pathways [96].
Some studies reported the apoptosis inducing properties of Kmp which can be partly accredited to its impacts on pathway of MAPK. In A-549 and MCF-7 cell lines, initiation of MAPK pathway is a key factor in Kmp-induced apoptosis. Moreover, Kmp-mediated activation of MAPK can block DNA damage which leads to transformation of cell. Kmp presence increases the expression of haemoxygenase-1 gene (HO-1), which triggers the rise in antioxidant ability of cells [97]. Treatment of Kmp significantly increased the viability of cells in response to oxidative stress, which involves unstable free radicals susceptible to damage DNA. Thus, Kmp-induced MAPK induction defends healthy cells from converting into cancer cells. RSK2 is a major suppressor of apoptosis, it downregulates the BAD, a protein which promotes apoptosis and upregulates the Bcl-2 level [98]. It has been observed that Kmp directly binds to RSK2 protein particularly at lysine-100 (Lys) and valine-82 (Val) positions, which plays an important role in RSK2 functioning [99]. Thus, Kmp paralyzes the RSK2. Obviously, treatment dropped Bcl level and increase concentration of tumor suppressor protein such as p53 and BAD [98]. Moreover, Kmp has also been reported to interrupt activity of Src-kinase [100]. MAPK is activated by Src in pro growth situation, which activates the COX-2 protein, and occurrence of COX-2 is a cautionary marker for skin tumor [101]. MAPK-ERK pathway is modified at various crucial sites by Kmp (Figure 4).
Hence, Kmp affects STAT3, PI3K signalling and MAPK pathway and exhibits significant potential in manipulation of cell-signalling pathways in apoptosis initiation and leaves normal cells alone.

2.5. Cell Cycle

A cell cycle is repeating series of events which involves copying of contents of cell and following division. Cells are continuously subject to DNA mutation that is harmful for cells but hardly results in cells production which can avoid the normal restrictions and flourish as pathologic tumors [102]. The development and progression of cancer is often associated with disruption or dysregulation of normal cell-cycle progression. Cells react to damage in DNA by stopping cell cycle progress and / or by enduring apoptosis [102].
Several flavonoids and natural chemo preventers including Kmp have been observed to precisely regulate numerous proteins which are involved in cellular homeostasis and cell cycle, whose de-regulation may play a role in carcinogenesis [103,104]. The ability of Kmp to induce cell cycle arrest have been observed in several cell cycles like in a study conducted by Gao et al. (2018) found that Kmp treatment induces G2-M phase cell cycle arrest through checkpoint kinase2 (CHK2) in ovarian cancerous cells [105] and Xu et al. (2008) in their study observed that Kmp induces G2-M phase cell cycle arrest in cervical cancerous cells [106], it has been reported that Kmp therapy can lead to G0-G1 cell cycle arrest in human esophageal squamous carcinoma Eca-109 cells [107]. Kmp treatment increased the level of p53 in MDA-MB-453 breast cancerous cells [108]. Furthermore, gene c-Myc is usually overexpressed in cancerous cells which leads to uncontrolled cell proliferation [109]. Studies showed that enhanced c-Myc level antagonized mRNA concentration of CDKN1A [110], administration of Kmp in combination with cisplatin reduces mRNA concentration of c-Myc and increases mRNA concentration of CDKN1A in ovarian cancerous cells. Cisplatin alone cannot kill cancerous cells, however, in combination with Kmp, they initiate apoptotic pathway via hindering c-Myc expressions in cancerous cells [111]. p53 is famous tumor suppressor protein generally indicated as ‘guardian of genome’ [111]. Repairing of damaged DNA is generally regulated by p53 [111]. Luo et al. (2011) observed that Kmp prevented phosphorylation of AKT signalling but upregulated the p53 expression and induced apoptosis in ovarian cancerous cells (Figure 5) [98]. Kmp is a useful flavonoid with genuine ability in disrupting growth of cancer and deserves more study into its impact on the cell cycle. A versatile chemoprophylactic molecule, kmp appears to play a role in each part of growth of cancer. Indeed, there persist a host of kmp-sensitive genes awaiting to be studied [112]. Kmp can efficiently prevent the proliferation and activation of mice T-lymphocytes in response to ConA, and can arrest cell cyle at G2/M and S phases [113].

2.6. Remodeling Tumor Metabolism

Metabolic remodeling is a phenomenon of the occurrence and development of tumors. It provides energy and material to the cells for survival and proliferation and prepares cells to survive in the harsh microenvironment [114]. Kaempferol inhibit both growth and migration of glioma cells, even when kaempferol was loaded to mucoadhesive nanoemulsion (KPF-MNE) or kaempferol-loaded nanoemulsion (KPF-NE) [27].
Table 1. Major mechanism of action of Kaempferol (Kmp) in cancer management.
Table 1. Major mechanism of action of Kaempferol (Kmp) in cancer management.
Major MechanismOutcome of the StudyRefs
InflammationKmp has been recognised as an effective inhibitor of pro-inflammatory molecules including vascular cell adhesion protein 1, prostaglandin-endoperoxide synthase (PTGS) and inducible nitric oxide synthase (NOSII)[46,47]
InflammationAnti-inflammatory effects of Kmp are mostly facilitated by downregulation of numerous sequence-specific DNA-binding factors like STAT, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) which have the capability to encourage the pro-inflammatory cytokines activation[48]
Reactive Oxygen Species (ROS)The ant-oxidant property of Kmp is linked with its up regulatory effects on antioxidant-response element- (ARE) mediative anti-oxidative enzymes like superoxide dismutase, catalase, and haem oxygenase in control of nuclear factor erythroid 2-related factor 2 signalling pathway[59]
Reactive Oxygen Species (ROS)Kmp reduced the thiobarbituric-acid reactive substances and red blood corpuscles lysates and upregulated the level of enzymatic antioxidants such as superoxide dismutase, glutathione perxidases (GSHPx) and catalase when 1,2-dimethylehydrazine (DMH)-induced-colon cancer male Wistar-rats treated with Kmp[63]
AngiogenesisKmp prevented VEGF secretion in MDA-MB-231 cancerous cells and decreased the concentration of VEGF-mRNA among ovarian cancerous cell lines[71]
AngiogenesisAdministered Kmp inhibited expression of NF-κB, c-Myc and phosphorylation of ERK and reduction of these encourages expression of p21 which antagonizes the release of VEGF[70,71]
Signal transducer and activator of transcription 3 (STAT3)At high concentration, Kmp prevented interleukin-6 induced-phosphorylation of STAT3[77]
Phosphatidylinositide-3-kinases (PI3K)-AKT pathways (PI3K-AKT)Kmp repressed the growth of colorectal cancerous cells by preventing the activation of PI3K-AKT signalling pathways[96]
Cell cycleKmp treatment induces G2-M phase cell cycle arrest through checkpoint kinase2 (CHK2) in ovarian cancerous cells or it has been shown that Kmp therapy can lead to G0-G1 cell cycle arrest in human esophageal squamous carcinoma Eca-109 cells[105,106,107]
Cell cycleAdministration of Kmp in combination with cisplatin reduces mRNA concentration of c-Myc and increases mRNA concentration of CDKN1A in ovarian cancerous cells[111]

3. Role of Kaempferol in Prevention and Inhibition of Various Types of Cancer

3.1. Hepatic Cancer

Hepatocellular carcinoma (HCC) is highly encountered hepatic cancer in adults [115]. It has been reported that Kmp in a dosage-dependent manner substantially prevent proliferation of liver cancerous cells such as Huh-7, SKHEP-1 and Hep.G2. Kmp plays an important role in inhibition and prevention of cancer through modulating various biological activities (Table 2). Additionally, 2-acetylaminofluorene and N-Nitrosodiethylamine-stimulated hepatocellular carcinoma from mice treated with combination of Kmp and luteolin prevented cancerous cells growth and caused apoptosis [116,117]. Kmp initiates apoptosis and triggers G2-M stage cell cycle arrest, hence, inhibiting invasion and migration of cancerous cells. Kmp can discharge cytochrome-c by generating ROS stimulates initiation of mitochondria swelling and loss of MtMP and increase the caspase 3 levels [115,116,117]. Kmp also increases the expression of non-receptor tyrosine-protein kinase (TYK-2), Janus kinase-1 (JAK-1), microtubule associated protein-1A-1B light chain-3 (MAPILC3), STAT1-2, autophagy related genes -5, -7 and -12, beclin-1 and phosphatase and tensin homolog (PTEN) and reduced the expression of cytokine signalling-3 (SOCS-3), PI3K-AKT-mTOR, miRNA-21, signal transducer and activator of transcription-3 (STAT-3), phosphorylated-mTOR signalling pathways and HIF1 in HCC [116,117,118,119].

3.2. Lung Cancer

Lung cancer is one of the most diagnosed cancers globally with an average survival rate of 5 years in most of the countries [120]. Lung cancer mainly is of two types- adenocarcinomas and NSCLC [121]. Harmful diet such as high intake of salt, low intake of fruits and vegetables, exposure to chemical carcinogens and smoking tobacco are the main risk factors linked with lung cancer [122]. Several flavonoids role in lung cancer have been examined by researchers [123,124] and in a cell line based study, it has been reported that Kmp inhibited the NSCLC, A549 cancerous cells [125,126,127,128], reduced formation of colonies and caused apoptosis [129]. Kmp significantly inhibited the migration of cells, suppressed epithelial mesenchymal transition and regained E-cadherin loss [130]. Kmp upregulated the expression level of Fas, transcription of miRNA-340, caspase 3, 7, 8, 9 and Bax and down-regulated the expressions of PI3K-AKT, Extracellular signal-regulated kinase signalling pathways, Mitogen-activated protein kinase (MEK)-1/2, MMP-2, B-cell lymphoma-extra-large and Bcl 2 which are involved in apoptotic pathways [17,125,126,127,128,129]. A study showed that Kmp reduced the number of metastasis and sub-cutaneous xenograft’s volume in comparison with control groups in lung-metastasis models [129].

3.3. Prostate Cancer

Prostate cancer (PCa) is among the male population is prominent cause of death worldwide and there is a requirement of efficient therapy for this disorder [131]. Kmp in dose-dependent manner prevents the proliferation of PCa cells [131], through up-regulation of expression levels of PARP and caspase-3, -9, -8 proteins [131,132]. Colony stimulating factor-2 activates the immune system of host and enable the immune surveillance of host through dendritic cells (DCs), hence, indicating a potential therapeutic in PCa treatment [132]. It has been observed that Kmp induces colony stimulation factor-2 release in PC3 cancerous cells and increases the DCs chemotaxis by activating protein kinase C, phosphor lipase C and MEK-1/2 [129]. Apparently, the PCa cells transcriptome is significantly influenced by treatment of Kmp as it downregulated the expression of androgen-receptor genes [133]. While orally consumed Kmp in mice did not demonstrate substantial toxicity and considerably enhanced survival and reduce the prostate cancer xenografts growth among athymic mouse group [133].

3.4. Oral Cancer

Oral cancer is the 6th common cancer around the globe [134]. In-vitro researches demonstrated the anti-proliferative effect of Kmp on oral squamous cell cancer (SCC) cells such as SCC-4, -25, -QLL1, -1483, oesophageal squamous cell carcinoma such as Eca109 cells, oral cavity tumor cells such as PCI13 and pharyngeal squamous carcinoma cells such as FaDu and inhibited cell invasion and migration, formation of clones and caused apoptosis [107,134,135,136]. Kmp triggered G0-G1 stage cell-cycle arrest and down-regulated the expression level of Bcl2, MMP2, hexokinase2 (HK2) and c-Jun and enhanced activation of EGFR, phosphorylation of ERK-1/2, glucose-uptake and up-regulated the expression of proteins PARP, caspase-9, -3 and Bax [107,134,135,136]. The anti-cancer properties of Kmp were verified in mouse xenograft models which revealed the capability of Kmp to substantially inhibit the tumor growth in combination with reduction in activity of EFGR and expression level of HK2 among cancerous tissues [135].

3.5. Gastric Cancer

Several studies observed the anti-proliferative activities of Kmp in SGC7901 and MKN29 stomach cancerous and promoted the G2-M stage cell cycle arrest, cell death and autophagy in these cancer cell lines [137,138]. Caused autophagic-cell death was associated with the up-regulation of PARP, IRE1-CHOP/JNK, signalling pathways, caspase-9, -3 and Bax and down-regulation expression level of Bcl2, phosphorylated-ERK, phosphorylated-AKT, CDK-1, cyclin-B1, p62, prostaglandin-endoperoxide synthase 2 [137,138].

3.6. Breast Cancer

Breast cancer is one of the most prevalent cancers among females in the world with significantly high mortality rate. Despite latest progress in early detection and therapeutic strategies, prevalence and mortality rate increasing continuously [139]. At concentration in micro molars (μM), Kmp efficiently prevents the breast cancerous cells growth such as MCF7, MDA-MB231 [140,141,142]. In addition, Kmp significantly prevents the bisphenol-A (and endocrine disrupting chemical) and triclosan-stimulated antiapoptotic activities [143], which initiates apoptosis, G2-M phase cell cycle arrest and DNA-fragmentation at sub G0 stage. Kmp reduces the level of antiapoptotic proteins including cyclin-A, -B, -E, -D1, CDK-1, phospho-AKT, Bcl2, Serine/threonine-protein kinase PLK1, phospho-MEK-1/2 and cathepsin-D [108,140,141,144,145,146,147] and enhances the level of proapoptotic proteins and enzymes including caspase-7, -9, -3, phospho-ATM, PARP, BAX, p53 [144,145]. It has been observed that Kmp reduced invasion and migration of cells among triple negative breast (TNB) cancerous cells in comparison with healthy cells [142]. These results described that Kmp downregulates RhoA protein and activates Rac-1 among TNB cancer cells and also activates HER-2-silence in SKBR-3 cells and ER-PR-silence in non TNB cells [142], and this indicates that the anti-proliferative effect of Kmp is initiated through estrogen receptor (ER)-dependent pathway which facilitates cell processes such as proliferation, development and differentiation [148]. Additionally, Kmp substantially triggers MAPK-cascades, and these are vital signalling pathways play an important role in regulation of differentiation, proliferation, and survival in healthy cells. Certainly, Kmp also initiates ERK along with ELK-1 and MEK-1 and reduces metastasis and EMT. After activation, MAPK signalling pathways causes activation of MMP-9 and -2, cathepsin-D and -B, AP1 which ultimately decreases invasion, adhesion, migration of cells [149,150,151,152].

3.7. Leukaemia

Acute promyelocytic leukaemia is a destructive disorder and characterised by defects in apoptotic pathway and growth of cells [153]. Kmp in dosage-dependent manner (12.5 to 100 μM) reduced the viability of cells among leukaemia cells such as NB-4 and HL60 [153,154]. Kmp downregulates the expression of proteins linked with phosphorylated-ATM, O6 methylguanine DNA methyltransferase (MGMT), p53, mediator of DNA damage checkpoint 1, phospho-ATR, DNA-dependent-protein kinase, DNA-repair mechanism, AKT, ATP Binding Cassette Subfamily C Member 1, Bcl2 genes expression and encourages G2/M stage cell cycle arrest and apoptosis. Kmp also upregulates the expression of phospho-p53, caspase-8, -3, cytochrome-c and phospho-H2AX [153,154,155,156]. The biomarker of cancer cell lines is not always directly referred to the anti-cancer event but a study observed that Kmp decreased the β-hexosaminidase release as a marker of de-granulation among leukemic cells such as RBL2H3 among mouse models [157] and enhanced the development of secretory granules in human leukaemia cells such as HMC1 [158]. A study on rat model of leukemia, found that kmp decreases the release of beta-hexosaminidase as a marker of degranulation in basophilic leukemia (RBL-2H3) cells, and increased the accumulation of mediators and the secretory granule development in human leukemic mast cells (HMC-1) [27].

3.8. Colon Cancer

Colon cancer is one of the most common cancers prevalent globally. The high incidences are often linked with western-style diet and intake of meat-dominant diet [159]. It has been stated that Kmp possesses cytotoxic effects on several colon cancerous cells such as HCT15, LS174T, HT29, SW40 and HCT-116 [160,161,162]. Several studies reported that Kmp in combination with 5Fluorouracil (5FU) among LS174T cells exhibited anti-proliferative effects [160]. Moreover, when Kmp combined to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resulted in apoptosis in colorectal cancerous cells via upregulation of death-receptor 5 (DR5) and receptors of TRAIL which increased the activity of TRAIL. Kmp causes G2-M stage cell cycle arrest and apoptosis and decreases invasion and migration of cells [160,161,162]. Kmp also prevented production of ROS and regulated the expression level of PI3K-AKT, JAK-STAT3, H2AX, MAPK, p-p38, PARP, caspase-7, -9, -3, -8, Bcl2, p21, p53-upregulated-modulator-of-apoptosis, ERK1/2, NF-κB and cytochrome-c release. Kmp decreased expression of heregulin-β (HRG-β), CDK-2, cyclin-B1, -E, -A, -D1, CDK-2, -4, CDC-25C, -2 and Gap junction alpha-1 protein. Kmp also increased the cleavage of PARP and repressed the retinoblastoma protein phosphorylation [63,160,161,162,163,164,165]. In a study it was found that by modulating miR-339-5p-hnRNPA1/PTBP1-PKM2 axis, kaempferol inhibits glycolysis and colon cancer growth, which reveals a new explanation for the molecular mechanism underlying kaempferol anti-tumor [166].

3.9. Brain Tumor

Glioblastoma (GB) is the most destructive and common form of brain tumor which is malignant and made from of connective tissue [167,168]. Several studies observed that Kmp prevented migration and growth of GB cells and kmp-loaded mucoadhesive nano emulsion also prevented the growth of glioma tumor cells [169,170,171]. Kmp can also induces apoptosis and generation of ROS by reducing concentration of thioredoxins, activity of superoxide dismutases and by increasing the level of Bcl2, caspase-8, -3, antiapoptotic proteins such as XIAP and survivin, PARP expression, proinflammatory cytokines such as Monocyte chemoattractant protein 1 (MCP-1/CCL2), IL-8, -6, decrease in AKT and ERK signalling pathways phosphorylation and de polarization of MtMP [169,170,171,172].

3.10. Pancreatic Cancer

Pancreatic cancer is one of the major causes of cancer related deaths around the globe and have worst prognosis [173]. Kmp in dosage-dependent manner prevents the pancreatic cancerous cells growth in PANC-1, MIA PaCa-2 and SNU213-pancreatic cancer cell line by causing apoptosis [173] and efficiently preventing ERK-1/2, EGFR-related AKT and Src signalling pathways and migration of cells [174]. Kmp can improve the repressive activities of regulatory T-cells by enhancing the expression levels of forkhead box P3 (FOXP3) [175,176].

3.11. Bladder Cancer

Bladder cancer is the highly prominent cancer of urinary tract [177]. Kmp inhibits growth of urinary bladder cancerous cells by encouraging apoptotic pathway and cell-cycle arrest [177,178,179,180]. It has been observed that Kmp can upregulate expression level of p38, phosphorylated-BRCA1, phosphorylated-ATM, Bax, p21, DNA methylation, Bid and p53 and downregulate the PTEN-PI3K-AKT signalling pathways, cyclin-D1, B-cell lymphoma-extra-large, MCL1, DNA-methyltransferase 3 beta and CDK-4 in bladder cancer cells [177,178,179,180]. These results have been supported by experiments conducted in sub-cutaneous xenografted mice model. Kmp substantially repressed the growth of tumor, invasion, and metastasis in xenografted models in comparison with untreated healthy controls. In these xenograft mice models, Kmp also downregulated the c-MET signalling pathways and growth-related markers and triggered upregulation of markers of apoptosis [178].

3.12. Osteosarcoma

Osteosarcoma is a bone cancer type which begins in the cells involved in bone formation. This cancer type is highly metastatic and infects soft and bone tissues proliferate to the lungs. It occurs mostly in long body bones like leg-bones and rarely occur in soft tissues outside the bones. It commonly happens in adolescents and young adults, but it can occur in older adults too [181]. Kmp in dosage-dependent manner prevents the growth of cancerous cells in bone cancerous cell lines such as HOB, 143B, U2OS and migration of U2OS cells with poorer toxicity in human fetal osteoblast cells [182,183]. Kmp can reduce JNK, p38 and ERK mitogen-activated protein kinase (MAPK) signalling pathways by down-regulating the MMP -9, -2, urokinase-type plasminogen activator (uPA) and activator protein-1 DNA binding activity [183]. Kmp substantially reduced the cell viability and number of viable cells and decreased the size of tumor in BALB/c-nu/nu rats transplanted with U2OS cells [182].

3.13. Cervical Cancer

Several studies observed that the Kmp inhibited cancerous cells growth in SiHa, KB-V1 and HeLa cervical cancerous cell lines in comparison with HFF cell line and healthy cells [112,184,185,186,187]. Kmp also triggered apoptotic pathway and G2-M stage cell cycle arrest associated with up-regulation of p53 with loss of MtMP and down-regulation of PI3K-AKT, NF-κB signalling pathways, P-glycoprotein, efflux of Rhodamine 123, Bcl2, cyclin-B1 and CDK-1 [106,112,184,185,186,187].

3.14. Renal Cancer

Renal cell cancer signifies the very common kidney cancer [188]. Kmp substantially prevents the growth of cancerous cells and initiates apoptotic pathway in renal cancerous cell lines such as 769-P and 786-O [189,190]. Kmp uses its anti-cancer activities via inhibiting invasion and migration of cells and enhancing the focal adhesion kinase activity [188]. Kmp upregulates the expression of p21, cyclin-B1 and cleavage of PARP and encourages the EGF receptor/p38 signalling pathway activation [189,190].

3.15. Ovarian Cancer

Studies utilizing human ovarian cancerous cell lines such as SK-OV-3, A2780, OVCAR 3, A2780-CP70 demonstrated that Kmp can prevent angiogenesis, proliferation, and growth of cancerous cells through reducing expression of VEGF [69]. Kmp could also encourages G2-M stage cell-cycle arrest and apoptotic pathway through up-regulation of Bax, p38, CDK1/CHK2-dependent CDC25C phosphorylation, p53, death receptor-5 and -4, gadd153, p21, ERK-1/2 and bad proteins and through down-regulation of HIF-1alpha [69,70,98,105,191,192].
Table 2. Kaempferol (Kmp) role in cancer management through modulating cell signalling pathways.
Table 2. Kaempferol (Kmp) role in cancer management through modulating cell signalling pathways.
Types of CancerMechanism/Outcome of the StudyRefs.
Hepatic cancerKmp in a dosage-dependent manner substantially prevent proliferation liver cancerous cells such as Huh-7, SKHEP-1 and Hep.G2[116,117]
Hepatic cancerAdditionally, 2-acetylaminofluorene and N-Nitrosodiethylamine-stimulated hepatocellular carcinoma from mice treated with combination of Kmp and luteolin prevented cancerous cells growth and caused apoptosis[116,117]
Lung cancerKmp inhibited the NSCLC A549 cancerous cells, reduced formation of colonies and caused apoptosis[125,126,127,128,129]
Lung cancerKmp reduced the number of metastasis and sub-cutaneous xenograft’s volume in comparison with control groups in lung-metastasis models [129]
Prostate cancerKmp in dose-dependent manner prevents the proliferation of prostate cancer cells, through up-regulation of expression levels of PARP and caspase-3, -9, -8 proteins[131,132]
Prostate cancerProstate cancerous cells transcriptome is significantly influenced by treatment of Kmp as it downregulated the expression of androgen-receptor genes [133]
Oral cancerIn-vitro research demonstrated the anti-proliferative effect of Kmp on oral squamous cell cancer (SCC) cells such as SCC-4, -25, -QLL1, -1483, oesophageal squamous cell carcinoma such as Eca109 cells, oral cavity tumor cells such as PCI13 and pharyngeal squamous carcinoma cells such as FaDu and inhibited cell invasion and migration, formation of clones and caused apoptosis [134,135,136]
Gastric cancerSeveral studies observed the anti-proliferative activities of Kmp in SGC7901 and MKN29 stomach cancerous and promoted the G2-M stage cell cycle arrest, cell death and autophagy in these cancer cell lines [137,138]
Breast cancerAt concentration in micro molars (μM), Kmp efficiently prevents the breast cancerous cells growth such as MCF7, MDA-MB231[140,141,142]
LeukaemiaKmp in dosage-dependent manner (12.5 to 100 μM) reduced the viability of cells among leukaemia cells such as NB-4 and HL60 [153,154]
LeukaemiaKmp decreased the β-hexosaminidase release as a marker of de-granulation among leukemic cells such as RBL2H3 among mouse models, and enhanced the development of secretory granules in human leukaemia cells such as HMC1 (The biomarker of cancer cell lines is not always directly referred to the anti-cancer event)[157,158]
Colon cancerwhen Kmp combined to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resulted in apoptosis in colorectal cancerous cells via upregulation of death-receptor 5 (DR5) and receptors of TRAIL which increased the activity of TRAIL [159]
Brain tumorKmp prevented migration and growth of GB cells and kmp-loaded mucoadhesive nano emulsion also prevented the growth of glioma tumor cells [169,170,171]
Pancreatic cancerKmp in dosage-dependent manner prevents the pancreatic cancerous cells growth in PANC-1, MIA PaCa-2, and SNU213-pancreatic cancer cell line by causing apoptosis and efficiently preventing ERK-1/2, EGFR-related AKT and Src signalling pathways and migration of cells [173,174]
Bladder cancerKmp inhibits growth of urinary bladder cancerous cells by encouraging apoptotic pathway and cell-cycle arrest [177,178,179,180]
OsteosarcomaKmp in dosage-dependent manner prevents the growth of cancerous cells in bone cancerous cell lines such as HOB, 143B, U2OS and migration of U2OS cells with poorer toxicity in human fetal osteoblast cells [182,183]
Cervical cancerKmp inhibited cancerous cells growth in SiHa, KB-V1 and HeLa cervical cancerous cell lines in comparison with HFF cell line and healthy cells [186,187]
Renal cancerKmp substantially prevents the growth of cancerous cells and initiates apoptotic pathway in renal cancerous cell lines such as 769-P and 786-O [189,190]
Renal cancerKmp uses its anti-cancer activities via inhibiting invasion and migration of cells and enhancing the focal adhesion kinase activity [188]
Ovarian cancerKmp can prevent angiogenesis, proliferation, and growth of ovarian cancerous cells through reducing expression of VEGF [69]

4. Bioavailability of Kaempferol

The bioavailability of ingested natural materials is associated with their absorption scope and concentration [193,194]. Factors like lipophilicity, permeability, efflux, and uptake by transporters influence the amount of every compound which is taken up by mesentery and transmitted to hepatic tissue via cells of intestine [195,196,197]. Till now, lot of studies have been done describing the in-vitro effects of flavonoids such as Kmp. Although it is still debatable whether Kmp is efficient in helping actual cancerous patients. Less consumption of vegetables has been constantly connected with enhanced cancer risk. Kmp is poorly absorbed with very deprived oral bioavailability, and it is usually metabolized into glucuronide, methyl, or sulphate forms [198,199]. Kmp efflux has been reported to limit its role as an anti-cancer agent [26,199]. Numerous population-based studies have verified that Kmp rich diet decreases risk of cancer among smokers [176,200]. These results can be partly described by Kmp’s disruption of aryl-hydrocarbon receptor (AHR) signalling pathway. Human carcinogenic agents like substances found in smoke of cigarettes activated AHR signalling pathway in humans [201]. These carcinogens form complex with AHR, which translocated to nucleus and encourage carcinogenic genes expression. Kmp functions to prevent AHR and carcinogenic substance binding, thus preventing transformation of cell bring in by use of cigarettes [202]. On the other hand, uncertain outcomes have been reported by the studies concentrating on the non-smoking individuals. Several potential researches showed that in recent years, Kmp intake significantly decreased the cancer risk among American woman nurses [203]. This indicates that Kmp as low-cost, non-toxic dietary element is a promising candidate for the chemo prevention of ovarian cancer. In contrast, a few researchers have observed very restricted help for chemo prevention via flavonoid-rich diet [204], but some claim no link exists between content of flavonoid and risk of cancer [205]. Similar to all the substances, flavonoids are also administered orally, and they first pass metabolism via wall of intestine and liver [198]. Flavonoid is identified as a foreign substance by body, human cells have several pumps intended to guide these foreign substances outside of membranes and cells [206]. Kmp is inadequately absorbed in the blood and can’t make its path into the cells, where it can prevent functions of some proteins and influence signalling pathways. Due to these effluxes, anti-cancer effects of Kmp might not be felt by body [207].
But latest developments have reported hope to overcome these hurdles in bioavailability. Breast cancer-resistance protein/ATP-binding cassette super-family G member-2 (BCRP/ABCG-2) is a transporter protein which can remove host of toxic substances from cell comprising Qu, a different flavonoid which have promising future in cancer treatment. But is has been reported that Kmp has a greater affinity for BCRP/ABCG-2 as compared to Qu. Studies found that anti-cancer affinity of Kmp increased, in combination with other anti-cancer agents. For instance, Kmp and Qu combination substantially increases the anti-cancer effects of Qu via obstructing the Qu efflux which allow Qu to stay inside and influence the signalling pathways [26,189]. Hence, Kmp might probably be in combination with other components or flavonoids exhibit much greater affinity for BCRP/ABCG-2 and that would put Kmp in the cancerous cells to cause destruction. Additionally, Kmp has been observed to reduce level of mRNA of ATP Binding Cassette Subfamily C Member 6 (ABCC-6), another ATP binding cassette transport protein coding gene [108]. ABCC-6 is associated with the transport of several chemotherapeutic drugs such as cisplatin to the outside of cell [204]. It has been observed that Kmp administration considerable increase the cytotoxic efficiency of cisplatin among cancerous cells [110]. Instead of many pharmacological properties, Kmp usage in biomedical applications is less, because it has poor water solubility, poor permeability, instability of chemicals in water alkaline medium, extensive metabolic processing before entering the systemic circulation. Hence, it has been reported that Kmp can enhance the bioavailability of other components used in the cancer treatment. Recently researchers working on a new approach to tackle this issue, is the development of nanoparticles as regulated drug delivery systems for increasing the oral bioavailability of hydrophobic and lipophilic drugs such as KFP. In research it was found that encapsulation of Kmp in NPs provides a potential platform for oxidative stress induce liver injury [208]. Another study confirmed that kaempferol-coated AgNPs can induce a potential anti-cancer effect in HepG2 cells via oxidative stress-mediated apoptosis [209]. Transporters such as BCRP/ABCG-2 are a promising therapeutic research aim for enhancing the access of body to Kmp and other flavonoids [4].

5. Conclusions

Plant-derived substances have been widely studied for cancer therapy in recent time. Substances such as kaempferol have significant capabilities to prevent cancer such as anti-proliferation, anti-inflammation, cell cycle arrest and pro-oxidation and got attention as a promising cancer treatment. Various techniques and processes have been developed by researchers to study the capabilities of natural chemo preventers which improve the impact of other chemo-therapeutic treatment by reducing their toxicity and enhancing their effects. Kaempferol has demonstrated to substantially affecting several cancer-related mechanisms, pathways and exhibited inhibitory effect on various cancer types including breast, hepatic, colon, lung, prostate, bladder, ovarian, oral, gastric, renal cancers. The review thus presents cumulative compendium of extensive research investigating the potential therapeutic role of kmp, in treatment of various types of cancers. Kaempferol also linked with some limitations mainly related to limited research in various domains of cancer, poor-absorption, and poor bioavailability. Hence, based on various therapeutic benefits of kmp, it strongly supports the development of clinical trials, incorporation of new approaches like nano technology to its application which can significantly enhance the potential of quercetin as powerful therapeutic agent. It can open new horizons in effective utilization, wider applicability, and better bioavailability of kmp as a potent natural chemo preventer alone or in from of combination drug for better prevention and management of cancer.

Author Contributions

A.K.V.—Conceptualization; M.Y.Q., M.I.K., S.H.A., A.K.V., F.A.A.-S., A.M.A. and A.A.A.A. wrote, reviewed, and edited the manuscript, analyzed the data, provided resources, were responsible for data curation, and reviewed drafts of the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to the deanship of scientific research at King Khalid University, Saudi Arabia for supporting this work under the General Project number (G.R.P/64/43). Malak Yahia Qattan would also like to extend her appreciation to the Research Centre, College of Applied Sciences and Community Services and the Deanship of Scientific Research at King Saud University, Riyadh, Saudi Arabia for funding this study. Deanship of scientific research at King Saud University, Riyadh, Saudi Arabia for funding this work.

Conflicts of Interest

The authors declare that there are no conflict of interest.

References

  1. Kashyap, D.; Garg, V.K.; Tuli, H.S.; Yerer, M.B.; Sak, K.; Sharma, A.K.; Kumar, M.; Aggarwal, V.; Sandhu, S.S. Fisetin and Quercetin: Promising Flavonoids with Chemopreventive Potential. Biomolecules 2019, 9, 174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. UICC. GLOBOCAN 2020: New Global Cancer Data. Available online: https://www.uicc.org/news/globocan-2020-new-global-cancer-data (accessed on 27 September 2022).
  3. Soto, A.M.; Sonnenschein, C. Environmental causes of cancer: Endocrine disruptors as carcinogens. Nat. Rev. Endocrinol. 2010, 6, 363–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sawyers, C. Targeted cancer therapy. Nature 2004, 432, 294–297. [Google Scholar] [CrossRef] [PubMed]
  5. Housman, G.; Byler, S.; Heerboth, S.; Lapinska, K.; Longacre, M.; Snyder, N.; Sarkar, S. Drug resistance in cancer: An overview. Cancers 2014, 6, 1769–1792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef] [PubMed]
  7. Gottesman, M.M. Mechanisms of cancer drug resistance. Annu. Rev. Med. 2002, 53, 615–627. [Google Scholar] [CrossRef] [Green Version]
  8. Kashyap, D.; Sharma, A.; Tuli, H.S.; Sak, K.; Garg, V.K.; Buttar, H.S.; Setzer, W.N.; Sethi, G. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. J. Funct. Foods 2018, 48, 457–471. [Google Scholar] [CrossRef]
  9. Kashyap, D.; Sharma, A.; Sak, K.; Tuli, H.S.; Buttar, H.S.; Bishayee, A. Fisetin: A bioactive phytochemical with potential for cancer prevention and pharmacotherapy. Life Sci. 2018, 194, 75–87. [Google Scholar] [CrossRef]
  10. Kashyap, D.; Sharma, A.; Tuli, H.S.; Sak, K.; Punia, S.; Mukherjee, T.K. Kaempferol—A dietary anticancer molecule with multiple mechanisms of action: Recent trends and advancements. J. Funct. 2017, 30, 203–219. [Google Scholar] [CrossRef]
  11. Kashyap, D.; Mondal, R.; Tuli, H.S.; Kumar, G.; Sharma, A.K. Molecular targets of gambogic acid in cancer: Recent trends and advancements. Tumor Biol. 2016, 37, 12915–12925. [Google Scholar] [CrossRef]
  12. Holland, T.M.; Agarwal, P.; Wang, Y.; Leurgans, S.E.; Bennett, D.A.; Booth, S.L.; Morris, M.C. Dietary flavonols and risk of Alzheimer dementia. Neurology 2020, 94, e1749–e1756. [Google Scholar] [CrossRef] [PubMed]
  13. Miean, K.H.; Mohamed, S. Flavonoid (myricetin, quercetin, kaempferol, luteolin, and apigenin) content of edible tropical plants. J. Agric. Food Chem. 2001, 49, 3106–3112. [Google Scholar] [CrossRef] [PubMed]
  14. de Vries, J.H.; Hollman, P.C.; Meyboom, S.; Buysman, M.N.; Zock, P.L.; van Staveren, W.A.; Katan, M.B. Plasma concentrations and urinary excretion of the antioxidant flavonols quercetin and kaempferol as biomarkers for dietary intake. Am. J. Clin. Nutr. 1998, 68, 60–65. [Google Scholar] [CrossRef] [Green Version]
  15. Marfak, A.; Trouillas, P.; Allais, D.P.; Champavier, Y.; Calliste, C.A.; Duroux, J.L. Radiolysis of kaempferol in water/methanol mixtures. Evaluation of antioxidant activity of kaempferol and products formed. J. Agric. Food Chem. 2003, 51, 1270–1277. [Google Scholar] [CrossRef] [PubMed]
  16. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Leung, H.W.; Lin, C.J.; Hour, M.J.; Yang, W.H.; Wang, M.Y.; Lee, H.Z. Kaempferol induces apoptosis in human lung non-small carcinoma cells accompanied by an induction of antioxidant enzymes. Food Chem. Toxicol. 2007, 45, 2005–2013. [Google Scholar] [CrossRef]
  18. Cid-Ortega, S.; Monroy-Rivera, J.A. Extraction of Kaempferol and Its Glycosides Using Supercritical Fluids from Plant Sources: A Review. Food Technol. Biotechnol. 2018, 56, 480–493. [Google Scholar] [CrossRef]
  19. Bestwick, C.S.; Milne, L.; Duthie, S.J. Kaempferol induced inhibition of HL-60 cell growth results from a heterogeneous response, dominated by cell cycle alterations. Chem.-Biol. Interact. 2007, 170, 76–85. [Google Scholar] [CrossRef]
  20. Casagrande, F.; Darbon, J.M. Effects of structurally related flavonoids on cell cycle progression of human melanoma cells: Regulation of cyclin-dependent kinases CDK2 and CDK1. Biochem. Pharmacol. 2001, 61, 1205–1215. [Google Scholar] [CrossRef]
  21. Knowles, L.M.; Zigrossi, D.A.; Tauber, R.A.; Hightower, C.; Milner, J.A. Flavonoids suppress androgen-independent human prostate tumor proliferation. Nutr. Cancer 2000, 38, 116–122. [Google Scholar] [CrossRef]
  22. Shanmugam, M.K.; Lee, J.H.; Chai, E.Z.; Kanchi, M.M.; Kar, S.; Arfuso, F.; Dharmarajan, A.; Kumar, A.P.; Ramar, P.S.; Looi, C.Y.; et al. Cancer prevention and therapy through the modulation of transcription factors by bioactive natural compounds. Semin. Cancer Biol. 2016, 40–41, 35–47. [Google Scholar] [CrossRef]
  23. Shanmugam, M.K.; Kannaiyan, R.; Sethi, G. Targeting cell signaling and apoptotic pathways by dietary agents: Role in the prevention and treatment of cancer. Nutr. Cancer 2011, 63, 161–173. [Google Scholar] [CrossRef] [PubMed]
  24. van Ginkel, P.R.; Yan, M.B.; Bhattacharya, S.; Polans, A.S.; Kenealey, J.D. Natural products induce a G protein-mediated calcium pathway activating p53 in cancer cells. Toxicol. Appl. Pharmacol. 2015, 288, 453–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Rajendran, P.; Rengarajan, T.; Nandakumar, N.; Palaniswami, R.; Nishigaki, Y.; Nishigaki, I. Kaempferol, a potential cytostatic and cure for inflammatory disorders. Eur. J. Med. Chem. 2014, 86, 103–112. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, A.Y.; Chen, Y.C. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Aslam Gondal, T.; Saeed, F.; Imran, A.; Shahbaz, M.; Tsouh Fokou, P.V.; Umair Arshad, M.; Khan, H.; et al. Kaempferol: A Key Emphasis to Its Anticancer Potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [Green Version]
  28. Li, H.; Ji, H.S.; Kang, J.H.; Shin, D.H.; Park, H.Y.; Choi, M.S.; Lee, C.H.; Lee, I.K.; Yun, B.S.; Jeong, T.S. Soy Leaf Extract Containing Kaempferol Glycosides and Pheophorbides Improves Glucose Homeostasis by Enhancing Pancreatic β-Cell Function and Suppressing Hepatic Lipid Accumulation in db/db Mice. J. Agric. Food Chem. 2015, 63, 7198–7210. [Google Scholar] [CrossRef]
  29. Sharifi-Rad, M.; Fokou, P.V.T.; Sharopov, F.; Martorell, M.; Ademiluyi, A.O.; Rajkovic, J.; Salehi, B.; Martins, N.; Iriti, M.; Sharifi-Rad, J. Antiulcer Agents: From Plant Extracts to Phytochemicals in Healing Promotion. Molecules 2018, 23, 1751. [Google Scholar] [CrossRef] [Green Version]
  30. Calderón-Montaño, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  31. Pei, J.; Chen, A.; Zhao, L.; Cao, F.; Ding, G.; Xiao, W. One-Pot Synthesis of Hyperoside by a Three-Enzyme Cascade Using a UDP-Galactose Regeneration System. J. Agric. Food Chem. 2017, 65, 6042–6048. [Google Scholar] [CrossRef]
  32. Neuhouser, M.L. Dietary flavonoids and cancer risk: Evidence from human population studies. Nutr. Cancer 2004, 50, 1–7. [Google Scholar] [CrossRef] [PubMed]
  33. Weng, C.J.; Yen, G.C. Flavonoids, a ubiquitous dietary phenolic subclass, exert extensive in vitro anti-invasive and in vivo anti-metastatic activities. Cancer Metastasis Rev. 2012, 31, 323–351. [Google Scholar] [CrossRef] [PubMed]
  34. Elsharkawy, E.R. Isolation of phytoconstituents and evaluation of anticancer and Antioxidant potential of Launaea mucronata (Forssk.) Muschl. subsp. Pak. J. Pharm. Sci. 2017, 30, 399–405. [Google Scholar] [PubMed]
  35. Yi, X.; Zuo, J.; Tan, C.; Xian, S.; Luo, C.; Chen, S.; Yu, L.; Luo, Y. Kaempferol, a flavonoid compound from gynura medica induced apoptosis and growth inhibition in mcf-7 breast cancer cell. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 210–215. [Google Scholar] [CrossRef] [Green Version]
  36. Mishra, A.P.; Salehi, B.; Sharifi-Rad, M.; Pezzani, R.; Kobarfard, F.; Sharifi-Rad, J.; Nigam, M. Programmed Cell Death, from a Cancer Perspective: An Overview. Mol. Diagn. Ther. 2018, 22, 281–295. [Google Scholar] [CrossRef]
  37. Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Atif, M.; Peters, D.G.; et al. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 33, 263–275. [Google Scholar] [CrossRef]
  38. Marfe, G.; Tafani, M.; Indelicato, M.; Sinibaldi-Salimei, P.; Reali, V.; Pucci, B.; Fini, M.; Russo, M.A. Kaempferol induces apoptosis in two different cell lines via Akt inactivation, Bax and SIRT3 activation, and mitochondrial dysfunction. J. Cell. Biochem. 2009, 106, 643–650. [Google Scholar] [CrossRef]
  39. Kim, K.Y.; Jang, W.Y.; Lee, J.Y.; Jun, D.Y.; Ko, J.Y.; Yun, Y.H.; Kim, Y.H. Kaempferol Activates G2-Checkpoint of the Cell Cycle Resulting in G2-Arrest and Mitochondria-Dependent Apoptosis in Human Acute Leukemia Jurkat T Cells. J. Microbiol. Biotechnol. 2016, 26, 287–294. [Google Scholar] [CrossRef]
  40. Kim, B.; Jung, J.W.; Jung, J.; Han, Y.; Suh, D.H.; Kim, H.S.; Dhanasekaran, D.N.; Song, Y.S. PGC1α induced by reactive oxygen species contributes to chemoresistance of ovarian cancer cells. Oncotarget 2017, 8, 60299–60311. [Google Scholar] [CrossRef] [Green Version]
  41. Ferrero-Miliani, L.; Nielsen, O.H.; Andersen, P.S.; Girardin, S.E. Chronic inflammation: Importance of NOD2 and NALP3 in interleukin-1beta generation. Clin. Exp. Immunol. 2007, 147, 227–235. [Google Scholar] [CrossRef]
  42. Warren, C.A.; Paulhill, K.J.; Davidson, L.A.; Lupton, J.R.; Taddeo, S.S.; Hong, M.Y.; Carroll, R.J.; Chapkin, R.S.; Turner, N.D. Quercetin may suppress rat aberrant crypt foci formation by suppressing inflammatory mediators that influence proliferation and apoptosis. J. Nutr. 2009, 139, 101–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Tuli, H.S.; Chaudhary, P.; Beniwal, V.; Sharma, A.K. Microbial pigments as natural color sources: Current trends and future perspectives. J. Food Sci. Technol. 2015, 52, 4669–4678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Tuli, H.S.; Kashyap, D.; Sharma, A.K.; Sandhu, S.S. Molecular aspects of melatonin (MLT)-mediated therapeutic effects. Life Sci. 2015, 135, 147–157. [Google Scholar] [CrossRef] [PubMed]
  45. Tuli, H.S.; Kashyap, D.; Bedi, S.K.; Kumar, P.; Kumar, G.; Sandhu, S.S. Molecular aspects of metal oxide nanoparticle (MO-NPs) mediated pharmacological effects. Life Sci. 2015, 143, 71–79. [Google Scholar] [CrossRef]
  46. Kong, L.; Luo, C.; Li, X.; Zhou, Y.; He, H. The anti-inflammatory effect of kaempferol on early atherosclerosis in high cholesterol fed rabbits. Lipids Health Dis. 2013, 12, 115. [Google Scholar] [CrossRef] [Green Version]
  47. Liu, Z.K.; Xiao, H.B.; Fang, J. Anti-inflammatory properties of kaempferol via its inhibition of aldosterone signaling and aldosterone-induced gene expression. Can. J. Physiol. Pharmacol. 2014, 92, 117–123. [Google Scholar] [CrossRef]
  48. Hämäläinen, M.; Nieminen, R.; Vuorela, P.; Heinonen, M.; Moilanen, E. Anti-inflammatory effects of flavonoids: Genistein, kaempferol, quercetin, and daidzein inhibit STAT-1 and NF-kappaB activations, whereas flavone, isorhamnetin, naringenin, and pelargonidin inhibit only NF-kappaB activation along with their inhibitory effect on iNOS expression and NO production in activated macrophages. Mediat. Inflamm. 2007, 2007, 45673. [Google Scholar]
  49. García-Mediavilla, V.; Crespo, I.; Collado, P.S.; Esteller, A.; Sánchez-Campos, S.; Tuñón, M.J.; González-Gallego, J. The anti-inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappaB pathway in Chang Liver cells. Eur. J. Pharmacol. 2007, 557, 221–229. [Google Scholar] [CrossRef]
  50. Kim, K.; Kim, S.; Moh, S.H.; Kang, H. Kaempferol inhibits vascular smooth muscle cell migration by modulating BMP-mediated miR-21 expression. Mol. Cell. Biochem. 2015, 407, 143–149. [Google Scholar] [CrossRef]
  51. Kim, S.H.; Park, J.G.; Lee, J.; Yang, W.S.; Park, G.W.; Kim, H.G.; Yi, Y.S.; Baek, K.S.; Sung, N.Y.; Hossen, M.J.; et al. The dietary flavonoid Kaempferol mediates anti-inflammatory responses via the Src, Syk, IRAK1, and IRAK4 molecular targets. Mediat. Inflamm. 2015, 2015, 904142. [Google Scholar] [CrossRef] [Green Version]
  52. Kadioglu, O.; Nass, J.; Saeed, M.E.; Schuler, B.; Efferth, T. Kaempferol Is an Anti-Inflammatory Compound with Activity towards NF-κB Pathway Proteins. Anticancer Res. 2015, 35, 2645–2650. [Google Scholar] [PubMed]
  53. Rakoff-Nahoum, S. Why cancer and inflammation? Yale J. Biol. Med. 2006, 79, 123–130. [Google Scholar] [PubMed]
  54. Bobe, G.; Albert, P.S.; Sansbury, L.B.; Lanza, E.; Schatzkin, A.; Colburn, N.H.; Cross, A.J. Interleukin-6 as a potential indicator for prevention of high-risk adenoma recurrence by dietary flavonols in the polyp prevention trial. Cancer Prev. Res. 2010, 3, 764–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Adegoke, O.; Forbes, P.B. Challenges and advances in quantum dot fluorescent probes to detect reactive oxygen and nitrogen species: A review. Anal. Chim. Acta 2015, 862, 1–13. [Google Scholar] [CrossRef] [Green Version]
  56. Hazra, B.; Das Sarma, M.; Sanyal, U. Separation methods of quinonoid constituents of plants used in Oriental traditional medicines. J. Chromatogr. B 2004, 812, 259–275. [Google Scholar] [CrossRef]
  57. Melo, P.S.; Massarioli, A.P.; Denny, C.; dos Santos, L.F.; Franchin, M.; Pereira, G.E.; Vieira, T.M.; Rosalen, P.L.; de Alencar, S.M. Winery by-products: Extraction optimization, phenolic composition and cytotoxic evaluation to act as a new source of scavenging of reactive oxygen species. Food Chem. 2015, 181, 160–169. [Google Scholar] [CrossRef] [Green Version]
  58. Yoshida, T.; Konishi, M.; Horinaka, M.; Yasuda, T.; Goda, A.E.; Taniguchi, H.; Yano, K.; Wakada, M.; Sakai, T. Kaempferol sensitizes colon cancer cells to TRAIL-induced apoptosis. Biochem. Biophys. Res. Commun. 2008, 375, 129–133. [Google Scholar] [CrossRef]
  59. Saw, C.L.; Guo, Y.; Yang, A.Y.; Paredes-Gonzalez, X.; Ramirez, C.; Pung, D.; Kong, A.N. The berry constituents quercetin, kaempferol, and pterostilbene synergistically attenuate reactive oxygen species: Involvement of the Nrf2-ARE signaling pathway. Food Chem. Toxicol. 2014, 72, 303–311. [Google Scholar] [CrossRef]
  60. Kowalski, J.; Samojedny, A.; Paul, M.; Pietsz, G.; Wilczok, T. Effect of kaempferol on the production and gene expression of monocyte chemoattractant protein-1 in J774.2 macrophages. Pharmacol. Rep. 2005, 57, 107–112. [Google Scholar]
  61. Wahab, A.; Begum, S. Luteolin and kaempferol from Cassia alata, antimicrobial and antioxidant activity of its methanolic extracts. FUUAST J. Biol. 2014, 4, 1–5. [Google Scholar]
  62. Choe, K.I.; Kwon, J.H.; Park, K.H.; Oh, M.H.; Kim, M.H.; Kim, H.H.; Cho, S.H.; Chung, E.K.; Ha, S.Y.; Lee, M.W. The antioxidant and anti-inflammatory effects of phenolic compounds isolated from the root of Rhodiola sachalinensis A. BOR. Molecules 2012, 17, 11484–11494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Nirmala, P.; Ramanathan, M. Effect of kaempferol on lipid peroxidation and antioxidant status in 1,2-dimethyl hydrazine induced colorectal carcinoma in rats. Eur. J. Pharmacol. 2011, 654, 75–79. [Google Scholar] [CrossRef]
  64. Wang, M.; Sun, J.; Jiang, Z.; Xie, W.; Zhang, X. Hepatoprotective effect of kaempferol against alcoholic liver injury in mice. Am. J. Chin. Med. 2015, 43, 241–254. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Y.; Tang, C.; Zhang, H. Hepatoprotective effects of kaempferol 3-O-rutinoside and kaempferol 3-O-glucoside from Carthamus tinctorius L. on CCl4-induced oxidative liver injury in mice. J. Food Drug Anal. 2015, 23, 310–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Bestwick, C.S.; Milne, L.; Pirie, L.; Duthie, S.J. The effect of short-term kaempferol exposure on reactive oxygen levels and integrity of human (HL-60) leukaemic cells. Biochim. Biophys. Acta 2005, 1740, 340–349. [Google Scholar] [CrossRef] [Green Version]
  67. Folkman, J. Angiogenesis: An organizing principle for drug discovery? Nat. Rev. Drug Discov. 2007, 6, 273–286. [Google Scholar] [CrossRef]
  68. Ferrara, N. Vascular endothelial growth factor as a target for anticancer therapy. Oncologist 2004, 9 (Suppl. S1), 2–10. [Google Scholar] [CrossRef]
  69. Luo, H.; Rankin, G.O.; Liu, L.; Daddysman, M.K.; Jiang, B.H.; Chen, Y.C. Kaempferol inhibits angiogenesis and VEGF expression through both HIF dependent and independent pathways in human ovarian cancer cells. Nutr. Cancer 2009, 61, 554–563. [Google Scholar] [CrossRef]
  70. Luo, H.; Rankin, G.O.; Juliano, N.; Jiang, B.H.; Chen, Y.C. Kaempferol inhibits VEGF expression and in vitro angiogenesis through a novel ERK-NFκB-cMyc-p21 pathway. Food Chem. 2012, 130, 321–328. [Google Scholar] [CrossRef] [Green Version]
  71. Schindler, R.; Mentlein, R. Flavonoids and vitamin E reduce the release of the angiogenic peptide vascular endothelial growth factor from human tumor cells. J. Nutr. 2006, 136, 1477–1482. [Google Scholar] [CrossRef] [Green Version]
  72. Hirano, T.; Nakajima, K.; Hibi, M. Signaling mechanisms through gp130: A model of the cytokine system. Cytokine Growth Factor Rev. 1997, 8, 241–252. [Google Scholar] [CrossRef]
  73. Liu, Q.; Li, G.; Li, R.; Shen, J.; He, Q.; Deng, L.; Zhang, C.; Zhang, J. IL-6 promotion of glioblastoma cell invasion and angiogenesis in U251 and T98G cell lines. J. Neuro-Oncol. 2010, 100, 165–176. [Google Scholar] [CrossRef] [PubMed]
  74. Xie, T.X.; Wei, D.; Liu, M.; Gao, A.C.; Ali-Osman, F.; Sawaya, R.; Huang, S. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene 2004, 23, 3550–3560. [Google Scholar] [CrossRef] [PubMed]
  75. Michaud-Levesque, J.; Bousquet-Gagnon, N.; Béliveau, R. Quercetin abrogates IL-6/STAT3 signaling and inhibits glioblastoma cell line growth and migration. Exp. Cell Res. 2012, 318, 925–935. [Google Scholar] [CrossRef] [PubMed]
  76. Alvarez, J.V.; Frank, D.A. Genome-wide analysis of STAT target genes: Elucidating the mechanism of STAT-mediated oncogenesis. Cancer Biol. Ther. 2004, 3, 1045–1050. [Google Scholar] [CrossRef] [Green Version]
  77. Basu, A.; Das, A.S.; Sharma, M.; Pathak, M.P.; Chattopadhyay, P.; Biswas, K.; Mukhopadhyay, R. STAT3 and NF-κB are common targets for kaempferol-mediated attenuation of COX-2 expression in IL-6-induced macrophages and carrageenan-induced mouse paw edema. Biochem. Biophys. Rep. 2017, 12, 54–61. [Google Scholar] [CrossRef]
  78. Yang, S.; Si, L.; Jia, Y.; Jian, W.; Yu, Q.; Wang, M.; Lin, R. Kaempferol exerts anti-proliferative effects on human ovarian cancer cells by inducing apoptosis, G0/G1 cell cycle arrest and modulation of MEK/ERK and STAT3 pathways. J. Buon 2019, 24, 975–981. [Google Scholar]
  79. Kok, K.; Geering, B.; Vanhaesebroeck, B. Regulation of phosphoinositide 3-kinase expression in health and disease. Trends Biochem. Sci. 2009, 34, 115–127. [Google Scholar] [CrossRef]
  80. Vogt, P.K.; Hart, J.R.; Gymnopoulos, M.; Jiang, H.; Kang, S.; Bader, A.G.; Zhao, L.; Denley, A. Phosphatidylinositol 3-kinase: The oncoprotein. Curr. Top. Microbiol. Immunol. 2010, 347, 79–104. [Google Scholar]
  81. Arcaro, A.; Guerreiro, A.S. The phosphoinositide 3-kinase pathway in human cancer: Genetic alterations and therapeutic implications. Curr. Genom. 2007, 8, 271–306. [Google Scholar] [CrossRef]
  82. Brown, K.K.; Toker, A. The phosphoinositide 3-kinase pathway and therapy resistance in cancer. F1000prime Rep. 2015, 7, 13. [Google Scholar] [CrossRef]
  83. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [Green Version]
  84. Marone, R.; Cmiljanovic, V.; Giese, B.; Wymann, M.P. Targeting phosphoinositide 3-kinase: Moving towards therapy. Biochim. Biophys. Acta 2008, 1784, 159–185. [Google Scholar] [CrossRef]
  85. Maurya, A.K.; Vinayak, M. Abstract A07: Decline in the growth of murine T-cell lymphoma via modulation of PI3K signaling pathway: Key role of quercetin and PI-103. Mol. Cancer Ther. 2015, 14 (Suppl. S7), A07. [Google Scholar] [CrossRef]
  86. Maurya, A.K.; Vinayak, M. Quercetin regresses Dalton’s lymphoma growth via suppression of PI3K/AKT signaling leading to upregulation of p53 and decrease in energy metabolism. Nutr. Cancer 2015, 67, 354–363. [Google Scholar] [CrossRef]
  87. Maurya, A.K.; Vinayak, M. Anticarcinogenic action of quercetin by downregulation of phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC) via induction of p53 in hepatocellular carcinoma (HepG2) cell line. Mol. Biol. Rep. 2015, 42, 1419–1429. [Google Scholar] [CrossRef]
  88. Vanhaesebroeck, B.; Guillermet-Guibert, J.; Graupera, M.; Bilanges, B. The emerging mechanisms of isoform-specific PI3K signalling. Nat. Reviews. Mol. Cell Biol. 2010, 11, 329–341. [Google Scholar] [CrossRef]
  89. Dibble, C.C.; Manning, B.D. A molecular link between AKT regulation and chemotherapeutic response. Cancer Cell 2009, 16, 178–180. [Google Scholar] [CrossRef] [Green Version]
  90. Chan, C.H.; Jo, U.; Kohrman, A.; Rezaeian, A.H.; Chou, P.C.; Logothetis, C.; Lin, H.K. Posttranslational regulation of Akt in human cancer. Cell Biosci. 2014, 4, 59. [Google Scholar] [CrossRef] [Green Version]
  91. Liao, Y.; Hung, M.C. Physiological regulation of Akt activity and stability. Am. J. Transl. Res. 2010, 2, 19–42. [Google Scholar]
  92. Fyffe, C.; Falasca, M. 3-Phosphoinositide-dependent protein kinase-1 as an emerging target in the management of breast cancer. Cancer Manag. Res. 2013, 5, 271–280. [Google Scholar]
  93. Osaki, M.; Oshimura, M.; Ito, H. PI3K-Akt pathway: Its functions and alterations in human cancer. Apoptosis 2004, 9, 667–676. [Google Scholar] [CrossRef]
  94. Testa, J.R.; Bellacosa, A. AKT plays a central role in tumorigenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 10983–10985. [Google Scholar] [CrossRef] [Green Version]
  95. Chin, H.K.; Horng, C.T.; Liu, Y.S.; Lu, C.C.; Su, C.Y.; Chen, P.S.; Chiu, H.Y.; Tsai, F.J.; Shieh, P.C.; Yang, J.S. Kaempferol inhibits angiogenic ability by targeting VEGF receptor-2 and downregulating the PI3K/AKT, MEK and ERK pathways in VEGF-stimulated human umbilical vein endothelial cells. Oncol. Rep. 2018, 39, 2351–2357. [Google Scholar] [CrossRef]
  96. Li, Q.; Wei, L.; Lin, S.; Chen, Y.; Lin, J.; Peng, J. Synergistic effect of kaempferol and 5-fluorouracil on the growth of colorectal cancer cells by regulating the PI3K/Akt signaling pathway. Mol. Med. Rep. 2019, 20, 728–734. [Google Scholar] [CrossRef]
  97. Hong, J.T.; Yen, J.H.; Wang, L.; Lo, Y.H.; Chen, Z.T.; Wu, M.J. Regulation of heme oxygenase-1 expression and MAPK pathways in response to kaempferol and rhamnocitrin in PC12 cells. Toxicol. Appl. Pharmacol. 2009, 237, 59–68. [Google Scholar] [CrossRef]
  98. Luo, H.; Rankin, G.O.; Li, Z.; Depriest, L.; Chen, Y.C. Kaempferol induces apoptosis in ovarian cancer cells through activating p53 in the intrinsic pathway. Food Chem. 2011, 128, 513–519. [Google Scholar] [CrossRef]
  99. Cho, Y.Y.; Yao, K.; Pugliese, A.; Malakhova, M.L.; Bode, A.M.; Dong, Z. A regulatory mechanism for RSK2 NH2-terminal kinase activity. Cancer Res. 2009, 69, 4398–4406. [Google Scholar] [CrossRef] [Green Version]
  100. Lee, K.M.; Lee, K.W.; Jung, S.K.; Lee, E.J.; Heo, Y.S.; Bode, A.M.; Lubet, R.A.; Lee, H.J.; Dong, Z. Kaempferol inhibits UVB-induced COX-2 expression by suppressing Src kinase activity. Biochem. Pharmacol. 2010, 80, 2042–2049. [Google Scholar] [CrossRef] [Green Version]
  101. Athar, M.; An, K.P.; Morel, K.D.; Kim, A.L.; Aszterbaum, M.; Longley, J.; Epstein, E.H., Jr.; Bickers, D.R. Ultraviolet B(UVB)-induced cox-2 expression in murine skin: An immunohistochemical study. Biochem. Biophys. Res. Commun. 2001, 280, 1042–1047. [Google Scholar] [CrossRef]
  102. Meeran, S.M.; Katiyar, S.K. Cell cycle control as a basis for cancer chemoprevention through dietary agents. Front. Biosci. 2008, 13, 2191–2202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Gescher, A.; Pastorino, U.; Plummer, S.M.; Manson, M.M. Suppression of tumour development by substances derived from the diet—Mechanisms and clinical implications. Br. J. Clin. Pharmacol. 1998, 45, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Milner, J.A.; McDonald, S.S.; Anderson, D.E.; Greenwald, P. Molecular targets for nutrients involved with cancer prevention. Nutr. Cancer 2001, 41, 1–16. [Google Scholar]
  105. Gao, Y.; Yin, J.; Rankin, G.O.; Chen, Y.C. Kaempferol Induces G2/M Cell Cycle Arrest via Checkpoint Kinase 2 and Promotes Apoptosis via Death Receptors in Human Ovarian Carcinoma A2780/CP70 Cells. Molecules 2018, 23, 1095. [Google Scholar] [CrossRef] [Green Version]
  106. Xu, W.; Liu, J.; Li, C.; Wu, H.Z.; Liu, Y.W. Kaempferol-7-O-beta-D-glucoside (KG) isolated from Smilax china L. rhizome induces G2/M phase arrest and apoptosis on HeLa cells in a p53-independent manner. Cancer Lett. 2008, 264, 229–240. [Google Scholar] [CrossRef]
  107. Li, R.; Mei, J.; Liu, G.J. Kaempferol-induced apoptosis of human esophageal squamous carcinoma Eca-109 cells and the mechanism. J. South. Med. Univ. 2011, 31, 1440–1442. [Google Scholar]
  108. Choi, E.J.; Ahn, W.S. Kaempferol induced the apoptosis via cell cycle arrest in human breast cancer MDA-MB-453 cells. Nutr. Res. Pract. 2008, 2, 322–325. [Google Scholar] [CrossRef] [Green Version]
  109. Jung, P.; Menssen, A.; Mayr, D.; Hermeking, H. AP4 encodes a c-MYC-inducible repressor of p21. Proc. Natl. Acad. Sci. USA 2008, 105, 15046–15051. [Google Scholar] [CrossRef] [Green Version]
  110. Luo, H.; Daddysman, M.K.; Rankin, G.O.; Jiang, B.H.; Chen, Y.C. Kaempferol enhances cisplatin’s effect on ovarian cancer cells through promoting apoptosis caused by down regulation of cMyc. Cancer Cell Int. 2010, 10, 16. [Google Scholar] [CrossRef]
  111. Ito, K.; Nakazato, T.; Yamato, K.; Miyakawa, Y.; Yamada, T.; Hozumi, N.; Segawa, K.; Ikeda, Y.; Kizaki, M. Induction of apoptosis in leukemic cells by homovanillic acid derivative, capsaicin, through oxidative stress: Implication of phosphorylation of p53 at Ser-15 residue by reactive oxygen species. Cancer Res. 2004, 64, 1071–1078. [Google Scholar] [CrossRef] [Green Version]
  112. Kashafi, E.; Moradzadeh, M.; Mohamadkhani, A.; Erfanian, S. Kaempferol increases apoptosis in human cervical cancer HeLa cells via PI3K/AKT and telomerase pathways. Biomed. Pharmacother. 2017, 89, 573–577. [Google Scholar] [CrossRef] [PubMed]
  113. Mu, J.J.; Zeng, Y.Y.; Huang, X.Y.; Zhao, X.H.; Song, B. Effects of Kaempferol on activation, proliferation and cell cycle of mouse T lymphocytes in vitro. Chin. J. Cell. Mol. Immunol. 2009, 25, 1106–1108. [Google Scholar]
  114. Chang, L.; Fang, S.; Gu, W. The Molecular Mechanism of Metabolic Remodeling in Lung Cancer. J. Cancer 2020, 11, 1403–1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Seydi, E.; Salimi, A.; Rasekh, H.R.; Mohsenifar, Z.; Pourahmad, J. Selective Cytotoxicity of Luteolin and Kaempferol on Cancerous Hepatocytes Obtained from Rat Model of Hepatocellular Carcinoma: Involvement of ROS-Mediated Mitochondrial Targeting. Nutr. Cancer 2018, 70, 594–604. [Google Scholar] [CrossRef] [PubMed]
  116. Zhu, G.; Liu, X.; Li, H.; Yan, Y.; Hong, X.; Lin, Z. Kaempferol inhibits proliferation, migration, and invasion of liver cancer HepG2 cells by down-regulation of microRNA-21. Int. J. Immunopathol. Pharmacol. 2018, 32, 2058738418814341. [Google Scholar] [CrossRef] [Green Version]
  117. Mylonis, I.; Lakka, A.; Tsakalof, A.; Simos, G. The dietary flavonoid kaempferol effectively inhibits HIF-1 activity and hepatoma cancer cell viability under hypoxic conditions. Biochem. Biophys. Res. Commun. 2010, 398, 74–78. [Google Scholar] [CrossRef]
  118. Huang, W.W.; Tsai, S.C.; Peng, S.F.; Lin, M.W.; Chiang, J.H.; Chiu, Y.J.; Fushiya, S.; Tseng, M.T.; Yang, J.S. 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, 2069–2077. [Google Scholar] [CrossRef] [Green Version]
  119. Wonganan, O.; He, Y.J.; Shen, X.F.; Wongkrajang, K.; Suksamrarn, A.; Zhang, G.L.; Wang, F. 6-Hydroxy-3-O-methyl-kaempferol 6-O-glucopyranoside potentiates the anti-proliferative effect of interferon α/β by promoting activation of the JAK/STAT signaling by inhibiting SOCS3 in hepatocellular carcinoma cells. Toxicol. Appl. Pharmacol. 2017, 336, 31–39. [Google Scholar] [CrossRef]
  120. Allemani, C.; Weir, H.K.; Carreira, H.; Harewood, R.; Spika, D.; Wang, X.S.; Bannon, F.; Ahn, J.V.; Johnson, C.J.; Bonaventure, A.; et al. Global surveillance of cancer survival 1995–2009: Analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries (CONCORD-2). Lancet 2015, 385, 977–1010. [Google Scholar] [CrossRef] [Green Version]
  121. Herbst, R.S.; Heymach, J.V.; Lippman, S.M. Lung cancer. N. Engl. J. Med. 2008, 359, 1367–1380. [Google Scholar] [CrossRef]
  122. Shivappa, N.; Wang, R.; Hébert, J.R.; Jin, A.; Koh, W.P.; Yuan, J.M. Association between inflammatory potential of diet and risk of lung cancer among smokers in a prospective study in Singapore. Eur. J. Nutr. 2019, 58, 2755–2766. [Google Scholar] [CrossRef] [PubMed]
  123. Zanoaga, O.; Braicu, C.; Jurj, A.; Rusu, A.; Buiga, R.; Berindan-Neagoe, I. Progress in Research on the Role of Flavonoids in Lung Cancer. Int. J. Mol. Sci. 2019, 20, 4291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Collins, I.; Workman, P. New approaches to molecular cancer therapeutics. Nat. Chem. Biol. 2006, 2, 689–700. [Google Scholar] [CrossRef] [PubMed]
  125. Nguyen, T.T.; Tran, E.; Ong, C.K.; Lee, S.K.; Do, P.T.; Huynh, T.T.; Nguyen, T.H.; Lee, J.J.; Tan, Y.; Ong, C.S.; et al. Kaempferol-induced growth inhibition and apoptosis in A549 lung cancer cells is mediated by activation of MEK-MAPK. J. Cell. Physiol. 2003, 197, 110–121. [Google Scholar] [CrossRef] [PubMed]
  126. Sonoki, H.; Tanimae, A.; Endo, S.; Matsunaga, T.; Furuta, T.; Ichihara, K.; Ikari, A. Kaempherol and Luteolin Decrease Claudin-2 Expression Mediated by Inhibition of STAT3 in Lung Adenocarcinoma A549 Cells. Nutrients 2017, 9, 597. [Google Scholar] [CrossRef]
  127. Boadi, W.Y.; Lo, A. Effects of Quercetin, Kaempferol, and Exogenous Glutathione on Phospho- and Total-AKT in 3T3-L1 Preadipocytes. J. Diet. Suppl. 2018, 15, 814–826. [Google Scholar] [CrossRef]
  128. Han, X.; Liu, C.F.; Gao, N.; Zhao, J.; Xu, J. RETRACTED: Kaempferol suppresses proliferation but increases apoptosis and autophagy by up-regulating microRNA-340 in human lung cancer cells. Biomed. Pharmacother. 2018, 108, 809–816. [Google Scholar] [CrossRef]
  129. Qin, Y.; Cui, W.; Yang, X.; Tong, B. Kaempferol inhibits the growth and metastasis of cholangiocarcinoma in vitro and in vivo. Acta Biochim. Biophys. Sin. 2016, 48, 238–245. [Google Scholar] [CrossRef] [Green Version]
  130. Jo, E.; Park, S.J.; Choi, Y.S.; Jeon, W.K.; Kim, B.C. Kaempferol Suppresses Transforming Growth Factor-β1-Induced Epithelial-to-Mesenchymal Transition and Migration of A549 Lung Cancer Cells by Inhibiting Akt1-Mediated Phosphorylation of Smad3 at Threonine-179. Neoplasia 2015, 17, 525–537. [Google Scholar] [CrossRef] [Green Version]
  131. Halimah, E.; Diantini, A.; Destiani, D.P.; Pradipta, I.S.; Sastramihardja, H.S.; Lestari, K.; Subarnas, A.; Abdulah, R.; Koyama, H. Induction of caspase cascade pathway by kaempferol-3-O-rhamnoside in LNCaP prostate cancer cell lines. Biomed. Rep. 2015, 3, 115–117. [Google Scholar] [CrossRef]
  132. Bandyopadhyay, S.; Romero, J.R.; Chattopadhyay, N. Kaempferol and quercetin stimulate granulocyte-macrophage colony-stimulating factor secretion in human prostate cancer cells. Mol. Cell. Endocrinol. 2008, 287, 57–64. [Google Scholar] [CrossRef] [PubMed]
  133. Mamouni, K.; Zhang, S.; Li, X.; Chen, Y.; Yang, Y.; Kim, J.; Bartlett, M.G.; Coleman, I.M.; Nelson, P.S.; Kucuk, O.; et al. A Novel Flavonoid Composition Targets Androgen Receptor Signaling and Inhibits Prostate Cancer Growth in Preclinical Models. Neoplasia 2018, 20, 789–799. [Google Scholar] [CrossRef] [PubMed]
  134. Lin, C.W.; Chen, P.N.; Chen, M.K.; Yang, W.E.; Tang, C.H.; Yang, S.F.; Hsieh, Y.S. Kaempferol reduces matrix metalloproteinase-2 expression by down-regulating ERK1/2 and the activator protein-1 signaling pathways in oral cancer cells. PLoS ONE 2013, 8, e80883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Yao, S.; Wang, X.; Li, C.; Zhao, T.; Jin, H.; Fang, W. Kaempferol inhibits cell proliferation and glycolysis in esophagus squamous cell carcinoma via targeting EGFR signaling pathway. Tumour Biol. 2016, 37, 10247–10256. [Google Scholar] [CrossRef]
  136. Kang, J.W.; Kim, J.H.; Song, K.; Kim, S.H.; Yoon, J.H.; Kim, K.S. Kaempferol and quercetin, components of Ginkgo biloba extract (EGb 761), induce caspase-3-dependent apoptosis in oral cavity cancer cells. Phytother. Res. 2010, 24 (Suppl. S1), S77–S82. [Google Scholar] [CrossRef]
  137. Song, H.; Bao, J.; Wei, Y.; Chen, Y.; Mao, X.; Li, J.; Yang, Z.; Xue, Y. Kaempferol inhibits gastric cancer tumor growth: An in vitro and in vivo study. Oncol. Rep. 2015, 33, 868–874. [Google Scholar] [CrossRef] [Green Version]
  138. Kim, T.W.; Lee, S.Y.; Kim, M.; Cheon, C.; Ko, S.G. Kaempferol induces autophagic cell death via IRE1-JNK-CHOP pathway and inhibition of G9a in gastric cancer cells. Cell Death Dis. 2018, 9, 875. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, R.; Yang, L.; Li, S.; Ye, D.; Yang, L.; Liu, Q.; Zhao, Z.; Cai, Q.; Tan, J.; Li, X. Quercetin Inhibits Breast Cancer Stem Cells via Downregulation of Aldehyde Dehydrogenase 1A1 (ALDH1A1), Chemokine Receptor Type 4 (CXCR4), Mucin 1 (MUC1), and Epithelial Cell Adhesion Molecule (EpCAM). Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 412–420. [Google Scholar] [CrossRef] [Green Version]
  140. Azevedo, C.; Correia-Branco, A.; Araújo, J.R.; Guimarães, J.T.; Keating, E.; Martel, F. The chemopreventive effect of the dietary compound kaempferol on the MCF-7 human breast cancer cell line is dependent on inhibition of glucose cellular uptake. Nutr. Cancer 2015, 67, 504–513. [Google Scholar] [CrossRef]
  141. Zhu, L.; Xue, L. Kaempferol Suppresses Proliferation and Induces Cell Cycle Arrest, Apoptosis, and DNA Damage in Breast Cancer Cells. Oncol. Res. 2019, 27, 629–634. [Google Scholar] [CrossRef]
  142. Li, S.; Yan, T.; Deng, R.; Jiang, X.; Xiong, H.; Wang, Y.; Yu, Q.; Wang, X.; Chen, C.; Zhu, Y. Low dose of kaempferol suppresses the migration and invasion of triple-negative breast cancer cells by downregulating the activities of RhoA and Rac1. OncoTargets Ther. 2017, 10, 4809–4819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Lee, G.A.; Choi, K.C.; Hwang, K.A. Treatment with Phytoestrogens Reversed Triclosan and Bisphenol A-Induced Anti-Apoptosis in Breast Cancer Cells. Biomol. Ther. 2018, 26, 503–511. [Google Scholar] [CrossRef] [PubMed]
  144. Diantini, A.; Subarnas, A.; Lestari, K.; Halimah, E.; Susilawati, Y.; Supriyatna, S.; Julaeha, E.; Achmad, T.H.; Suradji, E.W.; Yamazaki, C.; et al. Kaempferol-3-O-rhamnoside isolated from the leaves of Schima wallichii Korth. inhibits MCF-7 breast cancer cell proliferation through activation of the caspase cascade pathway. Oncol. Lett. 2012, 3, 1069–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Tsiklauri, L.; An, G.; Ruszaj, D.M.; Alaniya, M.; Kemertelidze, E.; Morris, M.E. Simultaneous determination of the flavonoids robinin and kaempferol in human breast cancer cells by liquid chromatography-tandem mass spectrometry. J. Pharm. Biomed. Anal. 2011, 55, 109–113. [Google Scholar] [CrossRef]
  146. Kim, S.H.; Hwang, K.A.; Choi, K.C. Treatment with kaempferol suppresses breast cancer cell growth caused by estrogen and triclosan in cellular and xenograft breast cancer models. J. Nutr. Biochem. 2016, 28, 70–82. [Google Scholar] [CrossRef]
  147. Kang, G.Y.; Lee, E.R.; Kim, J.H.; Jung, J.W.; Lim, J.; Kim, S.K.; Cho, S.G.; Kim, K.P. Downregulation of PLK-1 expression in kaempferol-induced apoptosis of MCF-7 cells. Eur. J. Pharmacol. 2009, 611, 17–21. [Google Scholar] [CrossRef]
  148. Oh, S.M.; Kim, Y.P.; Chung, K.H. Biphasic effects of kaempferol on the estrogenicity in human breast cancer cells. Arch. Pharmacal Res. 2006, 29, 354–362. [Google Scholar] [CrossRef]
  149. Kim, B.W.; Lee, E.R.; Min, H.M.; Jeong, H.S.; Ahn, J.Y.; Kim, J.H.; Choi, H.Y.; Choi, H.; Kim, E.Y.; Park, S.P.; et al. Sustained ERK activation is involved in the kaempferol-induced apoptosis of breast cancer cells and is more evident under 3-D culture condition. Cancer Biol. Ther. 2008, 7, 1080–1089. [Google Scholar] [CrossRef] [Green Version]
  150. Lee, G.A.; Choi, K.C.; Hwang, K.A. Kaempferol, a phytoestrogen, suppressed triclosan-induced epithelial-mesenchymal transition and metastatic-related behaviors of MCF-7 breast cancer cells. Environ. Toxicol. Pharmacol. 2017, 49, 48–57. [Google Scholar] [CrossRef]
  151. Zheng, L.; Zhu, L.; Zhao, M.; Shi, J.; Li, Y.; Yu, J.; Jiang, H.; Wu, J.; Tong, Y.; Liu, Y.; et al. In Vivo Exposure of Kaempferol Is Driven by Phase II Metabolic Enzymes and Efflux Transporters. AAPS J. 2016, 18, 1289–1299. [Google Scholar] [CrossRef] [PubMed]
  152. Li, C.; Zhao, Y.; Yang, D.; Yu, Y.; Guo, H.; Zhao, Z.; Zhang, B.; Yin, X. Inhibitory effects of kaempferol on the invasion of human breast carcinoma cells by downregulating the expression and activity of matrix metalloproteinase-9. Biochem. Cell Biol. 2015, 93, 16–27. [Google Scholar] [CrossRef] [PubMed]
  153. Moradzadeh, M.; Tabarraei, A.; Sadeghnia, H.R.; Ghorbani, A.; Mohamadkhani, A.; Erfanian, S.; Sahebkar, A. Kaempferol increases apoptosis in human acute promyelocytic leukemia cells and inhibits multidrug resistance genes. J. Cell. Biochem. 2018, 119, 2288–2297. [Google Scholar] [CrossRef] [PubMed]
  154. Wu, L.Y.; Lu, H.F.; Chou, Y.C.; Shih, Y.L.; Bau, D.T.; Chen, J.C.; Hsu, S.C.; Chung, J.G. Kaempferol induces DNA damage and inhibits DNA repair associated protein expressions in human promyelocytic leukemia HL-60 cells. Am. J. Chin. Med. 2015, 43, 365–382. [Google Scholar] [CrossRef] [PubMed]
  155. Benyahia, S.; Benayache, S.; Benayache, F.; Quintana, J.; López, M.; León, F.; Hernández, J.C.; Estévez, F.; Bermejo, J. Isolation from Eucalyptus occidentalis and identification of a new kaempferol derivative that induces apoptosis in human myeloid leukemia cells. J. Nat. Prod. 2004, 67, 527–531. [Google Scholar] [CrossRef] [PubMed]
  156. Chen, D.; Daniel, K.G.; Chen, M.S.; Kuhn, D.J.; Landis-Piwowar, K.R.; Dou, Q.P. Dietary flavonoids as proteasome inhibitors and apoptosis inducers in human leukemia cells. Biochem. Pharmacol. 2005, 69, 1421–1432. [Google Scholar] [CrossRef] [PubMed]
  157. Xu, F.; Matsuda, H.; Hata, H.; Sugawara, K.; Nakamura, S.; Yoshikawa, M. Structures of new flavonoids and benzofuran-type stilbene and degranulation inhibitors of rat basophilic leukemia cells from the Brazilian herbal medicine Cissus sicyoides. Chem. Pharm. Bull. 2009, 57, 1089–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Alexandrakis, M.; Letourneau, R.; Kempuraj, D.; Kandere-Grzybowska, K.; Huang, M.; Christodoulou, S.; Boucher, W.; Seretakis, D.; Theoharides, T.C. Flavones inhibit proliferation and increase mediator content in human leukemic mast cells (HMC-1). Eur. J. Haematol. 2003, 71, 448–454. [Google Scholar] [CrossRef] [Green Version]
  159. Boateng, J.; Verghese, M.; Shackelford, L.; Walker, L.T.; Khatiwada, J.; Ogutu, S.; Williams, D.S.; Jones, J.; Guyton, M.; Asiamah, D.; et al. Selected fruits reduce azoxymethane (AOM)-induced aberrant crypt foci (ACF) in Fisher 344 male rats. Food Chem. Toxicol. 2007, 45, 725–732. [Google Scholar] [CrossRef]
  160. Riahi-Chebbi, I.; Souid, S.; Othman, H.; Haoues, M.; Karoui, H.; Morel, A.; Srairi-Abid, N.; Essafi, M.; Essafi-Benkhadir, K. The Phenolic compound Kaempferol overcomes 5-fluorouracil resistance in human resistant LS174 colon cancer cells. Sci. Rep. 2019, 9, 195. [Google Scholar] [CrossRef] [Green Version]
  161. Choi, J.B.; Kim, J.H.; Lee, H.; Pak, J.N.; Shim, B.S.; Kim, S.H. Reactive Oxygen Species and p53 Mediated Activation of p38 and Caspases is Critically Involved in Kaempferol Induced Apoptosis in Colorectal Cancer Cells. J. Agric. Food Chem. 2018, 66, 9960–9967. [Google Scholar] [CrossRef]
  162. Lee, H.S.; Cho, H.J.; Yu, R.; Lee, K.W.; Chun, H.S.; Park, J.H. Mechanisms underlying apoptosis-inducing effects of Kaempferol in HT-29 human colon cancer cells. Int. J. Mol. Sci. 2014, 15, 2722–2737. [Google Scholar] [CrossRef] [PubMed]
  163. Deepa, M.; Sureshkumar, T.; Satheeshkumar, P.K.; Priya, S. Antioxidant rich Morus alba leaf extract induces apoptosis in human colon and breast cancer cells by the downregulation of nitric oxide produced by inducible nitric oxide synthase. Nutr. Cancer 2013, 65, 305–310. [Google Scholar] [CrossRef] [PubMed]
  164. Li, W.; Du, B.; Wang, T.; Wang, S.; Zhang, J. Kaempferol induces apoptosis in human HCT116 colon cancer cells via the Ataxia-Telangiectasia Mutated-p53 pathway with the involvement of p53 Upregulated Modulator of Apoptosis. Chem.-Biol. Interact. 2009, 177, 121–127. [Google Scholar] [CrossRef] [PubMed]
  165. Nakamura, Y.; Chang, C.C.; Mori, T.; Sato, K.; Ohtsuki, K.; Upham, B.L.; Trosko, J.E. Augmentation of differentiation and gap junction function by kaempferol in partially differentiated colon cancer cells. Carcinogenesis 2005, 26, 665–671. [Google Scholar] [CrossRef] [PubMed]
  166. Wu, H.; Cui, M.; Li, C.; Li, H.; Dai, Y.; Cui, K.; Li, Z. Kaempferol reverses aerobic glycolysis via miR-339-5p-mediated PKM alternative splicing in colon cancer cells. J. Agric. Food Chem. 2021, 69, 3060–3068. [Google Scholar] [CrossRef]
  167. Ostrom, Q.T.; Gittleman, H.; Fulop, J.; Liu, M.; Blanda, R.; Kromer, C.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008–2012. Neuro-Oncology 2015, 17 (Suppl. S4), iv1–iv62. [Google Scholar] [CrossRef]
  168. Afshari, A.R.; Jalili-Nik, M.; Soukhtanloo, M.; Ghorbani, A.; Sadeghnia, H.R.; Mollazadeh, H.; Karimi Roshan, M.; Rahmani, F.; Sabri, H.; Vahedi, M.M.; et al. Auraptene-induced cytotoxicity mechanisms in human malignant glioblastoma (U87) cells: Role of reactive oxygen species (ROS). EXCLI J. 2019, 18, 576–590. [Google Scholar]
  169. Sharma, V.; Joseph, C.; Ghosh, S.; Agarwal, A.; Mishra, M.K.; Sen, E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol. Cancer Ther. 2007, 6, 2544–2553. [Google Scholar] [CrossRef] [Green Version]
  170. Jeong, J.C.; Kim, M.S.; Kim, T.H.; Kim, Y.K. Kaempferol induces cell death through ERK and Akt-dependent down-regulation of XIAP and survivin in human glioma cells. Neurochem. Res. 2009, 34, 991–1001. [Google Scholar] [CrossRef]
  171. Colombo, M.; Figueiró, F.; de Fraga Dias, A.; Teixeira, H.F.; Battastini, A.M.O.; Koester, L.S. Kaempferol-loaded mucoadhesive nanoemulsion for intranasal administration reduces glioma growth in vitro. Int. J. Pharm. 2018, 543, 214–223. [Google Scholar] [CrossRef]
  172. Siegelin, M.D.; Reuss, D.E.; Habel, A.; Herold-Mende, C.; von Deimling, A. The flavonoid kaempferol sensitizes human glioma cells to TRAIL-mediated apoptosis by proteasomal degradation of survivin. Mol. Cancer Ther. 2008, 7, 3566–3574. [Google Scholar] [CrossRef] [PubMed]
  173. Zhang, Y.; Chen, A.Y.; Li, M.; Chen, C.; Yao, Q. Ginkgo biloba extract kaempferol inhibits cell proliferation and induces apoptosis in pancreatic cancer cells. J. Surg. Res. 2008, 148, 17–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Lee, J.; Kim, J.H. Kaempferol Inhibits Pancreatic Cancer Cell Growth and Migration through the Blockade of EGFR-Related Pathway In Vitro. PLoS ONE 2016, 11, e0155264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Lin, F.; Luo, X.; Tsun, A.; Li, Z.; Li, D.; Li, B. Kaempferol enhances the suppressive function of Treg cells by inhibiting FOXP3 phosphorylation. Int. Immunopharmacol. 2015, 28, 859–865. [Google Scholar] [CrossRef] [PubMed]
  176. Nöthlings, U.; Murphy, S.P.; Wilkens, L.R.; Boeing, H.; Schulze, M.B.; Bueno-de-Mesquita, H.B.; Michaud, D.S.; Roddam, A.; Rohrmann, S.; Tjønneland, A.; et al. A food pattern that is predictive of flavonol intake and risk of pancreatic cancer. Am. J. Clin. Nutr. 2008, 88, 1653–1662. [Google Scholar] [CrossRef] [Green Version]
  177. Wu, P.; Meng, X.; Zheng, H.; Zeng, Q.; Chen, T.; Wang, W.; Zhang, X.; Su, J. Kaempferol Attenuates ROS-Induced Hemolysis and the Molecular Mechanism of Its Induction of Apoptosis on Bladder Cancer. Molecules 2018, 23, 2592. [Google Scholar] [CrossRef] [Green Version]
  178. Dang, Q.; Song, W.; Xu, D.; Ma, Y.; Li, F.; Zeng, J.; Zhu, G.; Wang, X.; Chang, L.S.; He, D.; et al. Kaempferol suppresses bladder cancer tumor growth by inhibiting cell proliferation and inducing apoptosis. Mol. Carcinog. 2015, 54, 831–840. [Google Scholar] [CrossRef]
  179. Garcia, R.; Gonzalez, C.A.; Agudo, A.; Riboli, E. High intake of specific carotenoids and flavonoids does not reduce the risk of bladder cancer. Nutr. Cancer 1999, 35, 212–214. [Google Scholar] [CrossRef]
  180. Xie, F.; Su, M.; Qiu, W.; Zhang, M.; Guo, Z.; Su, B.; Liu, J.; Li, X.; Zhou, L. Kaempferol promotes apoptosis in human bladder cancer cells by inducing the tumor suppressor, PTEN. Int. J. Mol. Sci. 2013, 14, 21215–21226. [Google Scholar] [CrossRef] [Green Version]
  181. Meyers, P.A.; Gorlick, R. Osteosarcoma. Pediatr. Clin. North Am. 1997, 44, 973–989. [Google Scholar] [CrossRef]
  182. Huang, W.W.; Chiu, Y.J.; Fan, M.J.; Lu, H.F.; Yeh, H.F.; Li, K.H.; Chen, P.Y.; Chung, J.G.; Yang, J.S. Kaempferol induced apoptosis via endoplasmic reticulum stress and mitochondria-dependent pathway in human osteosarcoma U-2 OS cells. Mol. Nutr. Food Res. 2010, 54, 1585–1595. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, H.J.; Lin, C.M.; Lee, C.Y.; Shih, N.C.; Peng, S.F.; Tsuzuki, M.; Amagaya, S.; Huang, W.W.; Yang, J.S. Kaempferol suppresses cell metastasis via inhibition of the ERK-p38-JNK and AP-1 signaling pathways in U-2 OS human osteosarcoma cells. Oncol. Rep. 2013, 30, 925–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Afroze, N.; Pramodh, S.; Almutary, A.G.; Rizvi, T.A.; Rais, N.; Raina, R.; Faiyazuddin, M.; Alnuqaydan, A.M.; Hussain, A. Kaempferol Regresses Carcinogenesis through a Molecular Cross Talk Involved in Proliferation, Apoptosis and Inflammation on Human Cervical Cancer Cells, HeLa. Appl. Sci. 2022, 12, 3155. [Google Scholar] [CrossRef]
  185. Liao, W.; Chen, L.; Ma, X.; Jiao, R.; Li, X.; Wang, Y. Protective effects of kaempferol against reactive oxygen species-induced hemolysis and its antiproliferative activity on human cancer cells. Eur. J. Med. Chem. 2016, 114, 24–32. [Google Scholar] [CrossRef]
  186. Limtrakul, P.; Khantamat, O.; Pintha, K. Inhibition of P-glycoprotein function and expression by kaempferol and quercetin. J. Chemother. 2005, 17, 86–95. [Google Scholar] [CrossRef]
  187. Tu, L.Y.; Bai, H.H.; Cai, J.Y.; Deng, S.P. The mechanism of kaempferol induced apoptosis and inhibited proliferation in human cervical cancer SiHa cell: From macro to nano. Scanning 2016, 38, 644–653. [Google Scholar] [CrossRef] [Green Version]
  188. Hung, T.W.; Chen, P.N.; Wu, H.C.; Wu, S.W.; Tsai, P.Y.; Hsieh, Y.S.; Chang, H.R. Kaempferol Inhibits the Invasion and Migration of Renal Cancer Cells through the Downregulation of AKT and FAK Pathways. Int. J. Med. Sci. 2017, 14, 984–993. [Google Scholar] [CrossRef] [Green Version]
  189. An, G.; Gallegos, J.; Morris, M.E. The bioflavonoid kaempferol is an Abcg2 substrate and inhibits Abcg2-mediated quercetin efflux. Drug Metab. Dispos. 2011, 39, 426–432. [Google Scholar] [CrossRef] [Green Version]
  190. Song, W.; Dang, Q.; Xu, D.; Chen, Y.; Zhu, G.; Wu, K.; Zeng, J.; Long, Q.; Wang, X.; He, D.; et al. Kaempferol induces cell cycle arrest and apoptosis in renal cell carcinoma through EGFR/p38 signaling. Oncol. Rep. 2014, 31, 1350–1356. [Google Scholar] [CrossRef] [Green Version]
  191. Zhao, Y.; Tian, B.; Wang, Y.; Ding, H. Kaempferol Sensitizes Human Ovarian Cancer Cells-OVCAR-3 and SKOV-3 to Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL)-Induced Apoptosis via JNK/ERK-CHOP Pathway and Up-Regulation of Death Receptors 4 and 5. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2017, 23, 5096–5105. [Google Scholar] [CrossRef] [Green Version]
  192. Xu, G.; Li, B.; Wang, T.; Wan, J.; Zhang, Y.; Huang, J.; Shen, Y. Enhancing the anti-ovarian cancer activity of quercetin using a self-assembling micelle and thermosensitive hydrogel drug delivery system. RSC Adv. 2018, 8, 21229–21242. [Google Scholar] [CrossRef] [PubMed]
  193. Amidon, G.L.; Lennernäs, H.; Shah, V.P.; Crison, J.R. A theoretical basis for a biopharmaceutic drug classification: The correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm. Res. 1995, 12, 413–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Cutler, D. Assessment of rate and extent of drug absorption. Pharmacol. Ther. 1981, 14, 123–160. [Google Scholar] [CrossRef] [PubMed]
  195. Chakraborty, S.; Shukla, D.; Mishra, B.; Singh, S. Lipid—An emerging platform for oral delivery of drugs with poor bioavailability. Eur. J. Pharm. Biopharm. 2009, 73, 1–15. [Google Scholar] [CrossRef] [PubMed]
  196. Wang, J.; Skolnik, S. Mitigating permeability-mediated risks in drug discovery. Expert Opin. Drug Metab. Toxicol. 2010, 6, 171–187. [Google Scholar] [CrossRef]
  197. Bohets, H.; Annaert, P.; Mannens, G.; Van Beijsterveldt, L.; Anciaux, K.; Verboven, P.; Meuldermans, W.; Lavrijsen, K. Strategies for absorption screening in drug discovery and development. Curr. Top. Med. Chem. 2001, 1, 367–383. [Google Scholar] [CrossRef] [Green Version]
  198. Barve, A.; Chen, C.; Hebbar, V.; Desiderio, J.; Saw, C.L.; Kong, A.N. Metabolism, oral bioavailability and pharmacokinetics of chemopreventive kaempferol in rats. Biopharm. Drug Dispos. 2009, 30, 356–365. [Google Scholar] [CrossRef] [Green Version]
  199. Li, C.; Li, X.; Choi, J.S. Enhanced bioavailability of etoposide after oral or intravenous administration of etoposide with kaempferol in rats. Arch. Pharmacal Res. 2009, 32, 133–138. [Google Scholar] [CrossRef]
  200. Bobe, G.; Weinstein, S.J.; Albanes, D.; Hirvonen, T.; Ashby, J.; Taylor, P.R.; Virtamo, J.; Stolzenberg-Solomon, R.Z. Flavonoid intake and risk of pancreatic cancer in male smokers (Finland). Cancer Epidemiol. Biomark. Prev. 2008, 17, 553–562. [Google Scholar] [CrossRef] [Green Version]
  201. Denison, M.S.; Nagy, S.R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharmacol. Toxicol. 2003, 43, 309–334. [Google Scholar] [CrossRef]
  202. Puppala, D.; Gairola, C.G.; Swanson, H.I. Identification of kaempferol as an inhibitor of cigarette smoke-induced activation of the aryl hydrocarbon receptor and cell transformation. Carcinogenesis 2007, 28, 639–647. [Google Scholar] [CrossRef] [PubMed]
  203. Gates, M.A.; Tworoger, S.S.; Hecht, J.L.; De Vivo, I.; Rosner, B.; Hankinson, S.E. A prospective study of dietary flavonoid intake and incidence of epithelial ovarian cancer. Int. J. Cancer 2007, 121, 2225–2232. [Google Scholar] [CrossRef] [PubMed]
  204. Gates, M.A.; Vitonis, A.F.; Tworoger, S.S.; Rosner, B.; Titus-Ernstoff, L.; Hankinson, S.E.; Cramer, D.W. Flavonoid intake and ovarian cancer risk in a population-based case-control study. Int. J. Cancer 2009, 124, 1918–1925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Wang, L.; Lee, I.M.; Zhang, S.M.; Blumberg, J.B.; Buring, J.E.; Sesso, H.D. Dietary intake of selected flavonols, flavones, and flavonoid-rich foods and risk of cancer in middle-aged and older women. Am. J. Clin. Nutr. 2009, 89, 905–912. [Google Scholar] [CrossRef] [Green Version]
  206. Schinkel, A.H.; Jonker, J.W. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: An overview. Adv. Drug Deliv. Rev. 2003, 55, 3–29. [Google Scholar] [CrossRef]
  207. Zhou, S.F.; Wang, L.L.; Di, Y.M.; Xue, C.C.; Duan, W.; Li, C.G.; Li, Y. Substrates and inhibitors of human multidrug resistance associated proteins and the implications in drug development. Curr. Med. Chem. 2008, 15, 1981–2039. [Google Scholar] [CrossRef]
  208. Kazmi, I.; Al-Abbasi, F.A.; Afzal, M.; Altayb, H.N.; Nadeem, M.S.; Gupta, G. Formulation and Evaluation of Kaempferol Loaded Nanoparticles against Experimentally Induced Hepatocellular Carcinoma: In Vitro and In Vivo Studies. Pharmaceutics 2021, 13, 2086. [Google Scholar] [CrossRef]
  209. Alyami, N.M.; Alyami, H.M.; Almeer, R. Using green biosynthesized kaempferol-coated sliver nanoparticles to inhibit cancer cells growth: An in vitro study using hepatocellular carcinoma (HepG2). Cancer Nanotechnol. 2022, 13, 26. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of Kaempferol [26].
Figure 1. Chemical structure of Kaempferol [26].
Molecules 27 08864 g001
Figure 2. Graphical representation of literature review.
Figure 2. Graphical representation of literature review.
Molecules 27 08864 g002
Figure 3. Kaempferol effects on Angiogenesis. HIF-1: hypoxia inducible factor-1; VEGF: vascular endothelial growth factor; ESRRA: estrogen related receptor alpha. Dotted lines signify earlier processes that have decreased due to Kmp [23].
Figure 3. Kaempferol effects on Angiogenesis. HIF-1: hypoxia inducible factor-1; VEGF: vascular endothelial growth factor; ESRRA: estrogen related receptor alpha. Dotted lines signify earlier processes that have decreased due to Kmp [23].
Molecules 27 08864 g003
Figure 4. Effect of Kmp on MAPK pathway [26].
Figure 4. Effect of Kmp on MAPK pathway [26].
Molecules 27 08864 g004
Figure 5. Effect of Kaempferol on the Cell Cycle [112].
Figure 5. Effect of Kaempferol on the Cell Cycle [112].
Molecules 27 08864 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Qattan, M.Y.; Khan, M.I.; Alharbi, S.H.; Verma, A.K.; Al-Saeed, F.A.; Abduallah, A.M.; Al Areefy, A.A. Therapeutic Importance of Kaempferol in the Treatment of Cancer through the Modulation of Cell Signalling Pathways. Molecules 2022, 27, 8864. https://doi.org/10.3390/molecules27248864

AMA Style

Qattan MY, Khan MI, Alharbi SH, Verma AK, Al-Saeed FA, Abduallah AM, Al Areefy AA. Therapeutic Importance of Kaempferol in the Treatment of Cancer through the Modulation of Cell Signalling Pathways. Molecules. 2022; 27(24):8864. https://doi.org/10.3390/molecules27248864

Chicago/Turabian Style

Qattan, Malak Yahia, Mohammad Idreesh Khan, Shudayyed Hasham Alharbi, Amit Kumar Verma, Fatimah A. Al-Saeed, Alduwish Manal Abduallah, and Azza A. Al Areefy. 2022. "Therapeutic Importance of Kaempferol in the Treatment of Cancer through the Modulation of Cell Signalling Pathways" Molecules 27, no. 24: 8864. https://doi.org/10.3390/molecules27248864

Article Metrics

Back to TopTop