Short abstract
Hesperidin belongs to flavanones class of flavonoids and is known to possess broad-spectrum applicability to prevent dreadful diseases such as cardiovascular disease, neurodegeneration, and cancer. The reported anticancer effects of hesperidin have been found to be associated with its anti-oxidant and anti-inflammatory activities. Hesperidin interacts with numerous recognized cellular targets and inhibits cancer cell proliferation by inducing apoptosis and cell cycle arrest. In addition, evidence has suggested its promising role in inhibiting tumor cell metastasis, angiogenesis, and chemoresistance. The present mini-review highlights the ongoing development to identify hesperidin targets in cancer. Furthermore, the potential of nano technology-based hesperidin combinations and delivery systems will also be discussed. Overall, this review highlights all the possible molecular targets affected by hesperidin in tumor cells on a single platform.
Impact statement
Experimental findings from numerous studies have demonstrated the anticancer effects of hesperidin (Hesp) to be associated with anti-oxidant and anti-inflammatory activities along with its potential role in inhibiting the tumor cell metastasis and angiogenesis. Additionally, Hesp can also reverse drug resistance of cancer cells, which make it a promising candidate to be used in combination with existing anti-cancer drugs. This review will be helpful for upcoming researchers and scientific community to find out complete capsular package about cancer drug targets of Hesp and its role in modulating various important hallmarks of cancer.
Keywords: Hesperidin, anticancer, synergistic effect, nanotechnology
Introduction
Citrus fruits, such as oranges, tangerines, lemons, and limes, are well known for their health-promoting and chemopreventive properties.1 One of the most important bioactive components which elicits promising anti-cancer potential is a flavonoid hesperidin (Hesp), also known as hesperetin 7-rutinoside.2 Hesp was first isolated from orange peels in 1828 and was one of the two compounds formerly erroneously named as “vitamin P.”3 This polyphenolic glycoside can be abundantly found in various citrus fruits. For instance, sweet oranges contain about 200–600 mg/l of Hesp, whereas the total amount of this molecule varies within 50–850 mg/l in clementines, 8.1–460 mg/l in mandarins, 38–410 mg/l in lime and lemon juice, and 20–170 mg/l in grapefruit juice.1 Although the bioavailability of dietary flavonoids is known to be limited, the low micromolar levels of Hesp (around 1 µM range) have been detected in the blood serum for 5–7 h after the intake of citrus juice,3,4 probably high enough to exert its health-promoting activities in the human body.
Hesp belongs to the flavanone family of bioflavonoids. Differently from some other common groups of flavonoids, such as flavones, flavonols or isoflavones, flavanones lack the double bond between the positions 2 and 3 in the C ring of general flavonoid structure.1 Hesp has been shown to exert a broad range of pharmacological activities, being a potent anti-oxidant, anti-inflammatory, anti-atherosclerotic, cardioprotective, neuroprotective, anti-allergic, anti-viral, anti-microbial, and anti-cancer compound.1,2,4,5 Its role in protection against malignant transformation and progression has been described in multiple preclinical studies, acting through diverse cellular signalling pathways.5,6 Indeed, Hesp can affect diverse molecular targets involved in survival, division, and death mechanisms of tumor cells.5,6
Cancer is the second leading cause of death worldwide, just behind cardiovascular diseases.7 In fact, there was an estimated 18.1 million new cancer cases and 9.6 million cancer deaths in 2018 around the world.8 Due to the continuously increasing global prevalence of malignancies, novel efficient therapeutics and treatment strategies are highly needed. Application of safe natural compounds with strong anticancer properties, such as Hesp, may open new avenues in cancer treatment.9–11 Therefore, different anti-tumor mechanisms of this attractive dietary bioflavonoid, i.e. antioxidant, anti-inflammatory, anti-proliferative, anti-angiogenic, anti-invasive, anti-metastatic and, pro-apoptotic properties, are summarized in this review. The dataset presented in this article may be valuable for initiating clinical trials in patients suffering from different cancers, either alone or in combination with traditional therapies.
Chemistry and synthetic preview of hesperidin
On the chemical perspective, hesperidin structure harbors an aglycone known as methyl eriodictyol (hesperetin) bonded to rutinose.12 Hesp contains a glycoside moiety which is a disaccharide (glucose and rhamnose) which is present in two isomeric forms, i.e. neohesperidose and rutinose.12 Neohesperidose is chemically known as 2-O-alpha-l-Rhamnopyranosyl-d-glucopyranose. It is reported to be present in citrus fruits in the form of hesperetin 7-O-neohesperidoside.13 Rutinose on the other hand, a disaccharide, usually obtained from herbal sources is chemically 6-O-(-l-Rhamnosyl)-d-glucose or 6-O-(-l-Rhamnopyranosyl)-d-glucopyranose.12,14 These disaccharide moieties are responsible for the bitterness of citrus bioflavonoids. Of the disaccharides, the taste of citrus fruits is tasteless because of the presence of rutinosides moiety, while the neohesperidosides moiety is responsible for the bitter taste. Hesp is commonly found in neohesperidosides in the form of grapefruit (bitter) and in rutinoside in the form of orange (non-bitter).12,14,15 In the chemical Hesp skeleton, hesperetin (aglycone structure) is bonded to glucose and rhamnose is further bonded through glucose moiety to this structure.12
In hesperetin, a bioflavonoid, aglycone structure is (S)-2,3-dihydro-5,7-dihydroxy-2–(3-hydroxy-4-methoxyphenyl)-4H-1-benzopyran-4-one (Figure 1).12 Hesp is produced from the alkaline hydrolysis of hesperetenic acid and phloroglucinol, which during acid hydrolysis gets converted into hesperetin, l-rhamnose, and d-glucose.12,16 The biological activity of Hesp is due to the presence of hydroxyl moieties in both the heterocyclic and aromatic rings.12,16 Further, there is a close relationship between the presence and number of hydroxyl moieties with the potential antioxidant capacity of hesperidin.12,16,17 Additionally, Hesp also harbors neuro-protective actions and has the capability of penetrating the blood–brain barrier. 12,16,17
Figure 1.
The chemical structures and three-dimensional (3D) conformers of Hesperidin and Hesperetin.
Hesperidin: Molecular mechanisms of action
Apoptotic and cell cycle arrest
Apoptosis induction and cell cycle arrest are among the most important mechanisms of Hesp action against cancer cells. Studies revealed that the pro-apoptotic action of Hesp is related to different kinase pathways. For example, suppression of phosphatidylinositol-4,5-bisphosphate 3-kinase subunit/ AKT Serine/threonine kinase/ inhibitor of kappa light polypeptide gene enhancer in B-cells (PI3K/Akt/IKK) signalling pathway in NALM-6 human pre-B cells by Hesp was reported to induce apoptosis (Figure 2).18 In breast cancer cell line, MCF-7, Hesp induced apoptotic events like phosphatidyl-serine externalization, DNA fragmentation, caspase-7 activation, and PARP (Poly (ADP-ribose)polymerase) cleavage, which are associated with the activation of caspase-9, loss of mitochondrial membrane potential, release of cytochrome c, and an increase Bax:Bcl-2 ratio. Further experiments revealed that Hesp induced apoptosis by accumulating reactive oxygen species (ROS) and activation of apoptosis signal regulating kinase 1/ Jun N-terminal kinase (ASK1/JNK) pathway.19 Besides these, the most important mechanism of apoptotic effect of Hesp is via generation of ROS. As reported by Zhang et al.,20 high ROS levels, along with adenosine triphosphate (ATP) and calcium are responsible for the induction of apoptosis by Hesp in hepatocellular carcinoma cells via the activation of mitochondrial pathway. Similarly, in gastric cancer cells and esophageal cancer cells, Hesp induces apoptosis by increasing ROS and activating mitochondrial pathway.21,22
Figure 2.
Role of hesperidin on apoptosis and cell cycle. Hesperidin can generate reactive oxygen species (ROS) in cancer cells and activate mitochondrial pathways (by upregulating caspases) and inhibit kinases, which can induce apoptosis. Also by regulating cell cycle-related proteins, hesperidin can arrest cancer cell cycle in G0/G1 phase and G2/M phase. (A color version of this figure is available in the online journal.)
Caspase-dependent apoptosis induction by Hesp in NCI-H358 and A549 non-small cell lung cancer (NSCLC) cells was also observed by Birsu et al.23 and Xia et al.24, while Hesp was also found to arrest cell cycle at G0/G1 phase via the downregulation of cyclinD1, and increasing the expression of p21 and p53. In addition, the endoplasmic reticulum stress pathway was also found to be involved in apoptosis induction in HeLa (immortal cervical cancer cells) by Hesp, along with arresting cell cycle at G0/G1 phase via the downregulation of cyclin D1, cyclin E1, and cyclin-dependent kinase 2 (Cdk2) at protein level25. The ROS-mediated apoptosis induction by hesp in human gall bladder carcinoma was also reported by Pandey et al.,26 but they found cell cycle arrest at G2/M phase. A recent25 evidence showed that Hesp is a potent inhibitor of calcium/calmodulin-dependent protein kinase IV (CAMKIV), and by inhibiting CAMKIV along with activating the caspase-3-dependent intrinsic pathway through the upregulation of pro-apoptotic protein, Bax (BCL2 associated X, apoptosis regulator), Hesp exerts its anti-apoptotic and anticancer activities.27
The in vitro activity of Hesp in inducing apoptosis and arresting cell cycle is also proved in vivo (Table 1). For example, in azoxymethane-induced mouse model of colon cancer, Hesp was found to alter the anti-apoptotic scenario by modulating Bax/Bcl-2 ratio, together with enhanced release of cytochrome-c and activation of caspase-3/9. Experimental studies revealed that Hesp initiates apoptosis by inhibiting constitutively activated Aurora-A-mediated PI3K/Akt/GSK-3β pathway, and mTOR (mammalian target of rapamycin) pathway coupled with the stimulation of autophagy in this colon cancer model.28 Hesp in combination with fistein has been reported to inhibit cellular proliferation via triggering programmed cell death in human K562 chronic myeloid leukemia (CML) cells through activation of caspase-3 and JAK/STAT (Janus Kinase/Signal transducer and activator of transcription 3) pathway and genes of JAK/STAT pathway have also been identified as candidates of CML therapy.29 In ferric nitrilotriacetate (Fe-NTA)-induced renal cancer model of Wistar rats, Hesp was found to induce apoptosis-related proteins caspase-3, caspase-9, Bax expression and downregulation of Bcl-2 (BCL2 apoptosis regulator), NF-κB (nuclear factor kappa B subunit), iNOS (inducible nitric oxide synthase), TNF-α (tumor necrosis factor-alpha), PCNA (proliferating cell nuclear antigen) expression, which were associated with Hesp anti-cancer activities in vivo.30 In xenograft model of colon cancerous mice, Hesp also showed mitochondrial pathway-mediated apoptosis induction, and cell cycle arrest at G2/M phase.31
Table 1.
A brief overview of the in vivo studies carried out using hesperidin.
Compound of interest | Model of study | Type of cancer | Treatment dose | Underlying mechanisms of observed results | Refs |
---|---|---|---|---|---|
Hesperidin | Diethylnitrosamine/CCl4-induced rats | Hepatocellular carcinoma | 11 mg/kg | ↓oxidative stress, inflammation, cell proliferation, TGF-β1/Smad3 signaling, and collagen deposition by activating Nrf2/ARE/HO-1 and PPARγ pathways | 32 |
Diethyl nitrosamine hepatocarcinogenesis-induced rat | Hepatocellular carcinoma | 1000, 500, and 250 ppm | Hypomethylating effect on the LINE-1 sequence (up to 47% hypomethylation at 12.5 mM) and on the ALU-M2 repetitive sequences (up to 32% at 6 mM) | 33 | |
Rats | Hepatocellular carcinoma | 150 mg/kg/day | ↓Wnt3a, β-catenin, Cyclin D1, and Wnt5a gene expressions | 34 | |
Female Sprague-Dawley rats | Breast cancer | 30 mg/kg/body weight | Elevation in glycoproteins, nucleic acids, lysosomal enzymes and also significant alterations in macromolecules in renal tissues of cancer bearing animals | 35 | |
Rats | Hepatocellular carcinoma | 50, 100, and 200 mg/kg/d | ↓Exosomal RAB11A messenger RNA and long noncoding RNA-RP11-583F2.2 along with the increase in exosomal miR-1298 | 36 | |
Wistar rats | Renal cancer | 100 and 200 mg/kg body weight | ↓PGE2, COX-2, VEGF, and improved renal function by ↓BUN, creatinine, and KIM-1 | 37 | |
Wistar rats | Renal cancer | 100 and 200 mg/kg b wt | Induce caspase-3, caspase-9, bax expression, and downregulate bcl-2, NFκB, iNOS, TNF-α, PCNA expression | 30 | |
Wistar rats | Hepatocellular carcinoma | 200 mg/kg body weight | ↓PI3K, Akt, CDK-2 protein expression | 38 | |
Mice | Lung cancer | 25 mg/kg body weight | ↓COX-2, MMP-2, and MMP-9 | 39 | |
Colon carcinoma (CT-26)–bearing mice | Colon cancer | 200 mg/kg | ↓WBC count caused by cyclophosphamide to reduce antitumor effect | 40 | |
7, 12-Dimethybenz (a) anthracene-induced rats | Breast cancer | 30 mg/kg | Decline in lipid peroxidation and membrane bound marker enzyme AST, ALT, ALP, ACP, 5′ND, γ-GT | 41 | |
Benzo(a)pyrene-induced Swiss albino mice | Lung cancer | 50 mg/kg | ↓lipid peroxides, aryl hydrocarbon hydroxylase (AHH), gamma glutamyl transpeptidase (γ-GT), 5′-nucleotidase (5′-ND) and lactate dehydrogenase (LDH) | 42 |
Anti-angiogenesis and anti-metastasis
In view of the limitations of presently marketed monoclonal antibodies and synthetic compounds as anti-angiogenic agents pertaining to toxicity and high cost incurred, natural compounds are being extensively explored as potential anti-angiogenic agents.43,44 These natural products such as flavonoids (e.g. hesp) modulate tumor angiogenesis via targeting vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), basic fibroblast growth factor (bFGF), endothelial cell proliferation, migration, and metastasis owing to their anti-proliferative potential.5 The first excerpts of the anti-metastatic and anti-proliferative effect of hesperetin were documented in 2007 by Lentini et al.45 in B16-F10 metastatic murine melanoma cells in vitro and C57BL6/N mice in vivo. Thereafter, in 2009, Yeh et al.46 illustrated anti-metastatic potential of Hesp in vitro in HepG2 human hepatocellular carcinoma cells where hesperidin suppressed secreted cytosolic MMP-9 expression via inhibition of activator protein-1 (AP-1, JNK signaling pathway) and NF-κB (NF-κB signaling pathway). This was further supported by findings from other studies in 2010 which reported Hesp inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced cytosolic MMP-2 and MMP-9 and cyclooxygenase-2 (COX-2) expression via modulating NF-κB and AP-1 induced tumor cell invasion and metastasis in lung cancer and hepatocellular carcinoma (Figure 3).39,47 Later, in 2012, hesperetin was documented to inhibit TGF-β1 (transforming growth factor-β signaling pathway)-induced tumor migration and metastasis via phosphorylation of Smad3 (SMAD family member 3).48
Figure 3.
Cellular targets of hesperidin while acting as anti-angiogenic and anti-metastatic agent. (A color version of this figure is available in the online journal.)
In 2015, Kim 49 reported the inhibitory effect of Hesp on vascular formation in human umbilical vascular endothelial cells (HUVECS) and mouse embryonic stem cell (mES)-derived endothelial like cells via blocking AKT and mTOR signaling pathway which lead to inhibition of cell migration, suppression of micro-vessel sprouting, and tube formation in HUVECS. Zhao et al.50 documented the anti-angiogenic effect of Hesp (a component of Qingdu granule) in MCF-7 and HUVECs cells where Hesp inhibited migration and tube formation in human breast cancer cells via downregulation of NFATc3 (nuclear factor of activated T-cells) expression. In female BALB/c nude mice, a xenograft tumor model in vivo Hesp inhibited tumor growth, decreased vascular density, inhibited VEGF, and down-regulated NFATc3, VEGF, and VEGFR2 expression via NFAT signaling pathway.50 Further research in the domain of anti-metastatic potential of Hesp in pancreatic cancer has shown to target MKK3/6 and p38 intracellular signaling pathways.51 In A549 non-small cell lung cancer cells, Hesp was described to significantly inhibit tumor migration capability via targeting SDF-1α (stromal cell-derived factor 1) leading to downregulation of CXCR-4 (C-X-C chemokine receptor type 4), p-Akt, p-IκB (phosphorylated-I kappa B), and p-p65 expression (SDF-1/CXCR-4 signaling cascade).52 Hesp was also documented to inhibit cell migration and invasion in human osteosarcoma MG-63 cells via wound healing and matrigel assay and in vivo in male BALB/c xenograft mice model.31
Hesperetin administered in combination with platinum drugs in vitro in A549 lung adenocarcinoma cells and in in vivo in C57BL/6 mice inhibited tumor proliferation and migration via targeting UDP-glucuronosyltransferse (UGT) family 1 member A3 (UGT1A3) more significantly in comparison to single drug treatment regime53. In another study, combined administration of naringenin with hesperetin was shown to maximize anti-metastatic effect in Panc-1 human pancreatic cancer cells via downregulation of FAK (focal adhesion kinase) and p38 signaling pathway54. These in vitro (Table 2) and in vivo studies (Table 1) on the cumulative front document the potential anti-angiogenic and anti-metastatic potential of hesperidin which may be used as a promising anti-cancer strategy for the management of human cancers without eliciting toxic effects on surrounding normal cells.
Table 2.
A brief overview of the in vitro studies carried out using hesperidin to study effect on cellular processes.
Effect | Underlying mechanism | Concentration | Cell line | Cancer type | Refs |
---|---|---|---|---|---|
Apoptosis | ↑Caspase-9, -8, and -3 activities, Bax, Bak, and tBid protein levels ↓Bcl-xL protein | 150.43 ± 12.32 μM | HepG2 | Hepatic cancer | 55 |
↑caspase-3 and ↓mitochondrial membrane potential | 50 μM | A549 and NCI-H358 | Lung cancer | 23 | |
cleavages of Bid, caspase-3, and PARP, upregulation of Bax, and down-regulation of Bcl-xl | 152.3 μM | MSTO-211H | Human malignant pleural mesothelioma | 56 | |
↑caspase-3 cleavage expression through ADD153’CHOP’GRP78 and cytochrome c signaling pathways | 10 μM | A2780 | Ovarian Cancer | 57 | |
↑expression pf PPARγ and p53 accumulation, ↓NF-κB | 10 μM | NALM-6 | Lymphoblastic leukemia | 58 | |
↑ROS, nuclear condensation, and activation of Caspase-3 | 143.39 mM | Primary GBC | Gall bladder carcinoma | 26 | |
Cell cycle arrest | G2/M phase cell cycle arrest | 33.5, 23.8 and 17.6 µM, respectively, at 24, 48 and 72 h | MG-63 | Cervical cancer | 31 |
G0/G1 arrest by ↓cyclinD1, cyclinE1, and cyclin-dependent kinase 2 at the protein level. | 0, 40, 80, and 160 μM | HeLa, HT-29 | Cervical cancer and Colon cancer | ||
G0/G1 arrest by ↓cyclin D1 and ↑p21 , p53 | 75–125 μg/mL | A549 | Lung cancer | 24 | |
Anti-metastatic | ↓acetaldehyde-activated NF-κB and activator protein 1 (AP-1) activity results in ↓MMP-9 expression | 50 μM | HepG2 | Hepatocellular carcinoma | 46 |
↓MMP-9 via NF-kappaB an AP-1 signaling pathway | 50 μM | HepG2 | Hepatocellular carcinoma | 47 | |
14-fold increase in loss of MMP via FGF and NF-κB signal transduction pathways | 50 μM | A549 | Lung cancer | 23 | |
inhibit migratory and invasive capability by SDF-1/CXCR-4 signaling cascade | 25–65.5 µg/mL | A549 | Lung cancer | 52 | |
Anti-angiogenic | inhibits vascular formation by blocking the AKT/mTOR signaling pathways | 100 μM | HUVECs | Nil | 49 |
Anti-oxidant | Cytotoxicity mediated through antioxidant properties | IC50 recorded at b10 μg/mL | MCF-7, HEp-2, HeLa and HepG-2 | Breast, larynx, cervix and liver carcinoma | 59 |
Anti-oxidant and anti-inflammatory effects
Inflammation is a complex physiological and biological process in which body fights toward the harmful stimuli such as undigested particles, chemical irritants, damaged cells, moreover viral, bacterial and parasitic infections by overexpression of various cytokines, chemokines, and pro-inflammatory mediators, including TNF-α, COX-2, IL-1β (interleukin-1β), IL-6, IL-8, iNOS, NO (nitric oxide), prostaglandins, and eicosanoids.60–65 Apart from the vigorous dependence of the expression level of pro-inflammatory mediators on the activation of different signaling pathways that can be regulated by various factors such as MAPKs (mitogen-activated protein kinases), NF-κB, ICAM-1 (intercellular adhesion molecule-1), and VCAM-1 (vascular cell adhesion molecule-1), there is a tight relationship between inflammation and production of ROS and RNS (reactive nitrogen species).66–71 Moreover, the mounting evidences indicate that both inflammation and oxidative stress drive carcinogenesis by inactivation of tumor suppressor genes, activation of oncogenes, and disruption of various cellular signaling pathways.72,73 On the other hand, it is a wide thought that Hesp and its derivatives can be considered as the substantial and effective traditional flavonoids on oxidative stress and inflammation along with proliferation, apoptosis, DNA damage, free radicals, carcinogenesis, hypertension, hyperglycemia, and hypolipidemia.74,75 The studies on anti-inflammatory mechanisms of Hesp indicate that Hesp can reduce the level of inflammatory factors such as VCAM-1, COX-2, MMP-2, MMP-9, PGE2 (prostaglandin E2), IL-4, IL-6, iNOS, and NO2.74,76 The anti-oxidative, chelating, and strong reducing properties along with the hydroxyl, hydrogen peroxide, superoxide, and free radical scavenging activities of Hesp occurred depending on the concentration and originated from the chemical structure by acting as a hydrogen donor to the radical molecules, and played a radical target role to form new complexes between the antioxidant radicals and the lipid radicals thanks to the presence of 300-hydroxy, 400-o-methoxy system in the B ring.2
Moreover, the radical scavenging activity is increased due to the entity of 300-OH and 5-OH groups, in combination with a 4-carbonyl function and C40–C80 double bond though the anti-oxidant activity of Hesp can be considered as moderate according to the other flavonoids due to the lack of OH at C4.2,9,68,77 On the other hand, the non-glycosylated form of Hesp named hesperetin displays a stronger anti-oxidant activity than hesp because it has an additional hydroxyl group in its molecular structure and O-glycosylation reduces the antioxidant activity along with the electronic delocalization capacity (Figure 1).12,78 The anti-oxidant activity of Hesp and hesperetin, which can be achieved by boosting cellular anti-oxidant defense and radical scavenging, has been verified by various in vivo and in vitro experimental models.2,74 For example, it was reported that Hesp could protect the cellular components such as DNA and proteins by radical scavenging and ROS neutralizing activity.12,76,79 Moreover, it has been proved in the experimental models that Hesp counteracts the harmful activities of hydrogen peroxide, peroxynitrite, carbon tetrachloride, cadmium, acetaminophen, nicotine, cyclophosphamide, acrylonitrile, dimethylbenz[a]anthracene, tert-butyl hydroperoxide, lipopolysaccharide, benzo[α]pyrene, technetium, gamma radiation and many others by increasing the level and activity of antioxidant enzymes and compounds such as GSH (reduced glutathione), GPx (glutathione peroxidase), GST (glutathione S-transferase), GR (glutathione reductase), SOD (superoxide dismutase), CAT (catalase), vitamin C and vitamin A, and decreasing the level of cellular damage markers such as LDH (lactate dehydrogenase), LPO (lipid peroxides), GGT (gamma glutamyl transpeptidase), AHH (aryl hydrocarbon hydroxylase), 5 ND (5 nucleotidase), ALP (alkaline phosphatase), AST (aspartate aminotransferase), ALT (alanine aminotransferase), and bilirubin.76,80,81
The Hesp and hesperetin induced increase in the level and activity of the anti-oxidant enzymes can be achieved via Keap1-Nrf2 (nuclear factor erythroid 2-related factor 2) pathway, which is known as the major regulator of oxidative and electrophilic stress.75 Briefly, Hesp and hesperetin increase the expression of Nrf2, separate Keap1-Nrf2 complex, and increase the nuclear translocation of Nrf2, and the production of anti-oxidant enzymes is increased by the activation of gene transcription thanks to the binding of Nrf2 to the antioxidant response element (ARE) within gene promoter region.75,82 Further, Hesperetin derivative-14 (HD-14) has also been reported to have anti-inflammatory potential and has been shown to inhibit p-JAK1/p-STAT2 via PPAR-γ upregulation in LPS-treated RAW264.7 cells.83 Similar results were reported using another Hesperetin derivative-12 (HDND-12) in RAW264.7 cells and were reported to down-regulate p-JAK2/p-STAT3 expression.84 Consequently, the derived perception from the published studies about hesperidin and its derivatives indicates that the anticancer property of Hesp seems to be largely originated from its anti-oxidant and anti-inflammatory activity (Figure 4).
Figure 4.
The anti-oxidant activity mechanisms of hesperidin. (A color version of this figure is available in the online journal.)
Synergistic effect of hesperidin with other anti-cancer agents
Hesp, a flavanone, is present in different citrus fruits and possesses a number of biological activities.85 Hesp is identified to possess potent anti-inflammatory, anti-carcinogenic, and anti-oxidant activities in different studies.5 The chemical structure of Hesp consists of hesperetin (methyl eriodictyol) bound to rutinose.12 The glycoside entity of Hesp is a disaccharide that comprises rhamnose and glucose.12 The research in present times on anticancer activities of natural compounds has been targeted on induction of cancer cell death. These natural compound induces cancer cell death initiated by apoptosis (type I programmed cell death), autophagic cell death (type II programmed cell death), and necroptosis (programmed necrosis).85–88 Hesp and Hesperetin cause cell proliferation delay in different cancer models. Different effects of these compounds have been reported depending on the factors including dose, type of compound, and cell line under study (Table 3). Cell cycle arrest related to cytostatic effects has been reported in cells that have elevated p53 and cyclin-dependent kinase inhibitor levels, along with lowered levels of cyclins and cyclin-dependent kinases.55
Table 3.
Synergistic relationship of hesperidin with other natural compounds against cancer.
Molecules in synergism | Effects | Refs |
---|---|---|
Doxorubicin – hesperidin | Alterations in the expression levels of HK2 and LDHA | 89 |
Doxorubicin – hesperidin | PgP expression inhibition | 90 |
Cytarabine – hesperidin | Lowering in IC50 values of Cytarabine | 91 |
Tamoxifen – hesperidin | Apoptosis induction, cell cycle arrest and downregulation of EGFR (epidermal growth factor receptor) and ERα (estrogen receptor alpha). | 92 |
Quercetin – hesperidin | H2O2 scavenging | 99 |
Synergistic interaction of hesperidin with different compounds
Doxorubicin
Doxorubicin (DOX) is widely employed in anti-tumour therapies.94,95 In spite of the systematic chemotherapy with doxorubicin, it provides only peripheral improvements in a survival of the hepatocellular carcinoma patients.96,97 The main mode of action of doxorubicin includes intercalation within DNA base pairs, thus resulting in DNA strand breakage and an inhibition of DNA and RNA synthesis by inhibition of topoisomerase II, resulting in DNA damage and apoptosis induction.98 The doxorubicin also initiates the ROS generation thus causing cell death. The application of apigenin and Hesp alongside doxorubicin revealed the effect on doxorubicin-induced toxicity. These compounds altered the expression levels of glycolytic pathway genes – HK2 (hexokinase 2) and LDHA (lactate dehydrogenase A), which possess a major role in the Warburg effect. The simultaneous administration of doxorubicin and apigenin or Hesp eradicated the damage with the simultaneous increase in the doxorubicin toxicity. In another study, effect of Hesp has been investigated in combination with doxorubicin to check its effect on doxorubicin-resistant MCF7 breast cancer cell lines. The cytotoxic effects were studied using MTT assay. Hesp combined with doxorubicin was unable to increase the apoptotic initiation but inhibited the PgP (P-glycoprotein) expression90 which is mainly responsible for developing the multidrug resistance after administration of doxorubicin.
Cytarabine
In another study, the use of Silibinin and Hesp showed 50% cell inhibition at 16.2 μM and 50.12 μM, respectively,91 whereas the fixed doses of Hesp and Silibinin in conjunction with Cytarabine at different concentrations lowered the IC 50 value of Cytarabine by 5.9 and 4.5 folds, respectively. Drug interaction analysis exhibited that Silibinin and Hesp showed synergistic effect with Cytarabine in 1:50 to 1:250 ratios.91 Hence, these compounds can be employed as chemotherapeutic agents either alone or in combination.
Tamoxifen
Tamoxifen (Tam) is a commonly used anticancer drug for estrogen receptor (ER)-positive breast cancer treatment. In different studies, it was shown that the some natural compounds, like Hesp, piperine (Pip) and bee venom (BV) have inhibitory effect on the breast cancer cells growth when used individually. The combined effect of these natural compounds and Tam was investigated in a study with a hypothesis that these compounds can increase the potential efficacy of growth inhibitory activity of Tam. The cytotoxic activity of Hesp, BV, and Pip was examined on MCF7 and T47D breast cancer cell lines via MTT assay and achieved equitable IC50 comparable to Tam results.92 The effect of different combinations was investigated and enhanced anti-proliferative result was obtained on MCF7 and T47D cell lines due to the synergistic effect. These natural compounds can synergistically increase the potential anticancer property of Tam against MCF7 and T47D cells probably by inducing the apoptosis, cell cycle size and EGFR (epidermal growth factor receptor), and ERα (estrogen receptor alpha) downregulation.
Quercetin
Etoposide [40-demethylepipodo- phyllotoxin- 9–(4,6-O-ethylidene) -b-d glucopyranoside] is a derivative of natural product, podophyllotoxin. Etoposide acts as therapeutic agent in different types of cancer due to its ability to inhibit the topoisomerase II enzyme and induction of DNA breaks. In a study, the synergistic effect of Hesp and quercetin was studied to improve oxidative damage caused by Etoposide on reproductive system in male rats. In this study, it was observed that administration of quercetin at concentration levels of 20 mg/kg body weight and Hesp at 25 mg/kg body weight for two months significantly enhanced sperm motility and count in experimental groups in comparison to etoposide-treated group. This improvement in sperm motility and count can be attributed to the H2O2 scavenging by quercetin and Hesp, resulting in inhibition of cellular DNA damage93,99.
Role of nano-technology in hesperidin delivery
Natural anti-oxidants such as hesperidin has shown promising impact for the treatment of malignant growth and other alike diseases due to high efficacy and lower side effects as compared to synthetic drugs.100,101 But Hesp’s clinical use was extremely restricted due to lower aqueous solubility and poor bioavailability.102 So there is need to overcome these issues for optimal use of this compound.103 Nanotechnology is an interdisciplinary area of research having broad applications like molecular imaging, molecular diagnosis, and particularly targeted drug delivery.104 Further, this technology can also overcome the solubility and bioavailability issues of drugs105 as these parameters have tremendous impact on treatment of cancer.106 Therefore, several studies were initiated on developing Hesp-based nanoparticles to improve the bioavailability, absorption, and bio-distribution of this flavonoids.103,1062525–109 Gu et al.106 have developed two nano-based formulations, i.e. hesperetin-TPGS (D-α-tocopheryl polyethylene glycol 1000 succinate) micelles and hesperetin-phosphatidylcholine (PC) complexes to improve the water solubility, antioxidant activity, and oral absorption of hesperetin and these formulation led to the increase of 16.2- and 18.0-fold in in-vitro antioxidant potential and in vivo oral absorption of hesperetin.
Similarly, Duranolu et al.103 developed hesperetin-loaded nanoparticles with high encapsulation efficiency and low particle size and various process parameters were optimized for the optimal use of hesperetin. Further to develop new targeting strategies Ferrari et al.108 have developed Hesp-coated solid lipid nanoparticles and evaluated there various physiochemical properties for optimum-targeted delivery for the treatment of several disorders. Praveen Kumar et al.109 evaluated cerebro-protective potential of hesperidin nanoparticles for the effective treatment of cerebral ischemia in rats. To increase the drug delivery potential for topical applications, Menezes et al.110 reported to fabricate textile-based Hesp-loaded nanocapsules. These fabric-based nanocapsules were found to be suitable for sustained release of drug. Although efforts were initiated for the fabrication of Hesp-coated nanoparticles for the effective treatment of various disorders, but there is lacunae of reports where Hesp-coated nanoparticles were clinically tried for cancer treatment. Therefore, more progressive endeavors are required to integrate hesperidin-based nanoparticles for the effective treatment of cancer and other diseases.
Conclusion and future perspectives
The importance of citrus bioflavonoids is attributed to the pharmacological activity of Hesp. Despite extensive treatment protocols and regimes for cancer patients ranging from surgery to chemotherapy to immunotherapy, cancer is not completely curable and the conventional treatment regime is associated with short-term and long-term adverse events. Recently, lot of research is ongoing to explore bioflavonoids, e.g. Hesp in cancer treatment in view of their potential antioxidant property. Recent literature from different research groups have emphasized on the potent antioxidant capacity of Hesp and its potential role as anticancer agent. This review demonstrates the anti-tumor effects of Hesp in different malignancies with special emphasis on its molecular mechanism of action. The importance of Hesp as anti-cancer agents is evident from the experimental evidences emphasizing on how Hesp modulates oxidative stress, inflammation, and cancer cell death (hallmarks of cancer). Further, Hesp has been reported to augment apoptosis in malignant cells via NF-κB B, mTOR, PI3K/AKT pathways.111 Hesp has also been documented to down-regulate pro-inflammatory mediators and enzymes (IL-1/6, TNF, COX-2) in tumorigenesis and improve anti-oxidant defense mechanism. In addition, Hesp significantly improves pharmacological symptoms of other diseases such as arthritis, myocardial, and infertility.112–116 Therefore, main benefit of using flavonoids in contrast to chemotherapeutic agents is attributed to their low toxicity and tolerability, and it is worthwhile to mention that even at highest dose, Hesp/hesperetin do not cause cytotoxic effects or acute oxidative damage. However, further studies are required to unravel the therapeutic effects of Hesp/hesperetin in cancer treatment. Though, Hesp is presently in pre-clinical trials, promising data from clinical trials are warranted to increase the translation applicability of Hesp in cancer treatment. From the future perspective, future in vitro and in vivo studies focusing on the following dimensions of Hesp need to be studied to translate the practical applicability of Hesp as anticancer agent:
To increase the bioavailability and absorption of Hesp/hesperetin (aglycone form).
Defining the precise molecular mechanisms of action of hesp through meticulously designed and executed experimental studies for anticancer effects Hesp.
Standardizing the optimum effective dose of Hesp for translation in clinical trials in combination with conventional chemotherapeutic agents and targeted therapies.
Evaluation of safety and efficacy findings in cancer patients undergoing treatment with Hesp.
ACKNOWLEDGMENTS
The authors acknowledge the Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh and Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala for providing an opportunity to complete the review.
Authors’ contributions
VA and HST: performed literature survey, data extraction and compiled the manuscript; KS: wrote the introduction section; Md. AK: Contributed in role of hesperidin in apoptosis induction and cell cycle arrest; VA composed the text on chemistry of hesperidin and its role in modulating angiogenesis and metastasis; SS and AP: prepared the section synergistic interaction of hesperidin with different compounds; MV; Composed the text of anti-oxidant and anti-inflammatory activity of hesperidin; DA wrote role of nanotechnology in hesperidin delivery; FT and PS: helped with conclusion sections, design and drew figures of the indicated sections and prepared the in vivo and in vitro study tables for the hesperidin manuscript; GS: critically reviewed and edited the manuscript; All authors read and approved the final manuscript.
DECLARATION OF CONFLICTING INTERESTS
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
FUNDING
The author(s) received no financial support for the research, authorship, and/or publication of this article.
ORCID iD
Gautam Sethi https://orcid.org/0000-0002-8677-8475
References
- 1.Barreca D, Gattuso G, Bellocco E, Calderaro A, Trombetta D, Smeriglio A, Lagana G, Daglia M. Meneghini S, Nabavi SM. Flavanones: citrus phytochemical with health-promoting properties. Biofactors 2017; 43:495–506 [DOI] [PubMed] [Google Scholar]
- 2.Ahmadi A, Shadboorestan A. Oxidative stress and cancer; the role of hesperidin, a citrus natural bioflavonoid, as a cancer chemoprotective agent. Nutr Cancer 2016; 68:29–39 [DOI] [PubMed] [Google Scholar]
- 3.Li C, Schluesener H. Health-promoting effects of the citrus flavanone hesperidin. Crit Rev Food Sci Nutr 2017; 57:613–31 [DOI] [PubMed] [Google Scholar]
- 4.Aishatwi AA, Ramesh E, Periasamy VS, Subash-Babu P. The apoptotic effect of hesperetin on human cervical cancer cells is mediated through cell cycle arrest, death receptor, and mitochondrial pathways. Fundam Clin Pharmacol 2013; 27:581–92 [DOI] [PubMed] [Google Scholar]
- 5.Roohbakhsh A, Parhiz H, Soltani F, Rezaee R, Iranshahi M. Molecular mechanisms behind the biological effects of hesperidin and hesperetin for the prevention of cancer and cardiovascular diseases. Life Sci 2015; 124:64–74 [DOI] [PubMed] [Google Scholar]
- 6.Ahmadi A, Shadboorestan A, Nabavi SF, Setzer WN, Nabavi SM. The role of hesperidin in cell signal transduction pathway for the prevention or treatment of cancer. Curr Med Chem 2015; 22:3462–71 [DOI] [PubMed] [Google Scholar]
- 7.Nagai H, Kim YH. Cancer prevention from the perspective of global cancer burden patterns. J Thorac Dis 2017; 9:448–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68:394–424 [DOI] [PubMed] [Google Scholar]
- 9.Wang CZ, Zhang Z, Anderson S, Yuan CS. Natural products and chemotherapeutic agents on cancer: prevention vs. treatment. Am J Chin Med 2014; 42:1555–8 [DOI] [PubMed] [Google Scholar]
- 10.Bishayee G, Sethi G. Bioactive natural products in cancer prevention and therapy: progress and promise. Semin Cancer Biol 2016; 40-41:1–3 [DOI] [PubMed] [Google Scholar]
- 11.Dai X, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp Biol Med 2015; 240:760–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Garg A, Garg S, Zaneveld LJ, Singla AK. Chemistry and pharmacology of the citrus bioflavonoid hesperidin. Phytother Res 2001; 15:655–69 [DOI] [PubMed] [Google Scholar]
- 13.Mesquita E, Monteiro M. Simulatneous HPLC determination of flavonoids and phenolic acids profile in Pera-Rio orange juice. Food Res Int 2018; 106:54–63 [DOI] [PubMed] [Google Scholar]
- 14.Shimoda K, Hamada H, Hamada H. Glycosylation of hesperetin by plant cell cultures. Phytochemistry 2008; 69:1135–40 [DOI] [PubMed] [Google Scholar]
- 15.Perfetti GA, Joe FL, Jr., Fazio T, Page SW. Liquid chromatographic methodology for the characterization of orange juice. J Assoc off Anal Chem 1988; 71:469–73 [PubMed] [Google Scholar]
- 16.Ranganna S, Govindarajan VS, Ramana KV. Citrus fruits – varieties, chemistry, technology, and quality evaluation. Part II. Chemistry, technology, and quality evaluation. A. chemistry. Crit Rev Food Sci Nutr 1983; 18:313–86 [DOI] [PubMed] [Google Scholar]
- 17.Mitsunaga Y, Takanaga H, Matsuo H, Naito M, Tsuruo T, Ohtani H, Sawada Y. Effect of bioflavonoids on vincristine transport across blood-brain barrier. Eur J Pharmacol 2000; 395:193–201 [DOI] [PubMed] [Google Scholar]
- 18.Shahbazi R, Cheraghpour M, Homayounfar R, Nazari M, Nasrollahzadeh J, Davoodi SH. Hesperidin inhibits insulin-induced phosphoinositide 3-kinase/akt activation in human pre-B cell line NALM-6. J Cancer Res Ther 2018; 14:503–8 [DOI] [PubMed] [Google Scholar]
- 19.Palit S, Kar S, Sharma G, Das PK. Hesperetin induces apoptosis in breast carcinoma by triggering accumulation of ROS and activation of ASK1/JNK pathway. J Cell Physiol 2015; 230:1729–39 [DOI] [PubMed] [Google Scholar]
- 20.Zhang J, Song J, Wu D, Wang J, Dong W.. Hesperetin Induces the Apoptosis of Hepatocellular Carcinoma Cells via Mitochondrial Pathway Mediated by the Increased Intracellular Reactive Oxygen Species, ATP AND Calcium. Med Oncol 2015;32:101. [DOI] [PubMed] [Google Scholar]
- 21.Wu D, Zhang J, Wang J, Li J, Liao F, Dong W. Hesperetin induces apoptosis of esophageal cancer cells via mitochondrial pathway mediated by the increased intracellular reactive oxygen species. Tumour Biol 2016; 37:3451–9 [DOI] [PubMed] [Google Scholar]
- 22.Zhang J, Wu D, Vikash Song J, Wang J, Yi J, Dong W. Hesperetin induces the apoptosis of gastric cancer cells via activating mitochondrial pathway by increasing reactive oxygen species. Dig Dis Sci 2015; 60:2985–95 [DOI] [PubMed] [Google Scholar]
- 23.Birsu Cincin Z, Unlu M, Kiran B, Sinem Bireller E, Baran Y, Cakmakoglu B. Anti-proliferative, apoptotic and signal transduction effects of hesperidin in non-small cell lung cancer cells. Cell Oncol 2015; 38:195–204 [DOI] [PubMed] [Google Scholar]
- 24.Xia R, Sheng X, Xu X, Yu C, Lu H. Hesperidin induces apoptosis and G0/G1 arrest in human non-small cell lung cancer A549 cells. Int J Mol Med 2018; 41:464–72 [DOI] [PubMed] [Google Scholar]
- 25.Wang Y, Yu H, Zhang J, Gao J, Ge X, Lou G.. Hesperidin Inhibits Hela Cell Proliferation through Apoptosis Mediated by Endoplasmic Reticulum Stress Pathways and Cell Cycle Arrest. BMC Cancer 2015;15:682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pandey P, Sayyed U, Tiwari RK, Siddiqui MH, Pathak N, Bajpai P. Hesperidin induces ROS-mediated apoptosis along with cell cycle arrest at G2/M phase in human gall bladder carcinoma. Nutr Cancer 2019; 71:676–87 [DOI] [PubMed] [Google Scholar]
- 27.Naz H, Tarique M, Ahamad S, Alajmi MF, Hussain A, Rehman MT, Luqman S, Hassan MI. Hesperidin-CAMKIV interaction and its impact on cell proliferation and apoptosis in the human hepatic carcinoma and neuroblastoma cells. J Cell Biochem 2019; 120:15119–30 [DOI] [PubMed] [Google Scholar]
- 28.Saiprasad G, Chitra P, Manikandan R, Sudhandiran G. Hesperidin induces apoptosis and triggers autophagic markers through inhibition of Aurora-a mediated phosphoinositide-3-kinase/akt/mammalian target of rapamycin and glycogen synthase kinase-3 beta signalling Cascades in experimental Colon carcinogenesis. Eur J Cancer 2014; 50:2489–507 [DOI] [PubMed] [Google Scholar]
- 29.Adan A, Baran Y. Fistein and hesperetin induced apoptosis and cell cycle in chronic myeloid leukemia cells accompanied by modulation of cellular signaling. Tumour Biol 2015; 37:5781–95 [DOI] [PubMed] [Google Scholar]
- 30.Siddiqi A, Hasan SK, Nafees S, Rashid S, Saidullah B, Sultana S. Chemopreventive efficacy of hesperidin against chemically induced nephrotoxicity and renal carcinogenesis via amelioration of oxidative stress and modulation of multiple molecular pathways. Exp Mol Pathol 2015; 99:641–53 [DOI] [PubMed] [Google Scholar]
- 31.Du GY, He SW, Zhang L, Sun CX, Mi LD, Sun ZG. Hesperidin exhibits in vitro and in vivo antitumor effects in human osteosarcoma MG-63 cells and xenograft mice models via inhibition of cell migration and invasion, cell cycle arrest and induction of mitochondrial-mediated apoptosis. Oncol Lett 2018; 16:6299–306 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 32.Mahmoud AM, Mohammed HM, Khadrawy SM, Galaly SR. Hesperidin protects against chemically induced hepatocarcinogenesis via modulation of Nrf2/ARE/HO-1, PPARgamma and TGF-beta1/Smad3 signaling, and amelioration of oxidative stress and inflammation. Chem Biol Interact 2017; 277:146–58 [DOI] [PubMed] [Google Scholar]
- 33.Fernandez-Bedmar Z, Anter J, Alonso-Moraga A, Martin de Las Mulas J, Millan-Ruiz Y, Guil-Luna S. Demethylating and anti-hepatocarcinogenic potential of hesperidin, a natural polyphenol of citrus juices. Mol Carcinog 2017; 56:1653–62 [DOI] [PubMed] [Google Scholar]
- 34.Zaghloul RA, Elsherbiny NM, Kenawy HI, El-Karef A, Eissa LA, El-Shishtawy MM. Hepatoprotective effect of hesperidin in hepatocellular carcinoma: involvement of Wnt signaling pathways. Life Sci 2017; 185:114–25 [DOI] [PubMed] [Google Scholar]
- 35.Nandakumar N, Jayaprakash R, Balasubramanian MP. Influence of hesperidin on renal cell surface glycoprotein content, nucleic acids, lysosomal enzymes and macromolecules against 7, 12-dimethylbenz [a] anthracene induced experimental breast carcinoma. J Exp Ther Oncol 2012; 9:265–80 [PubMed] [Google Scholar]
- 36.Huang SM, Tsai SY, Lin JA, Wu CH, Yen GC. Cytoprotective effects of hesperetin and hesperedin against amyloid β-induced impairment of glucose transport through downregulation of neuronal autophagy. Mol Nutr Food Res 2012; 56:601–9 [DOI] [PubMed] [Google Scholar]
- 37.Siddiqi A, Saidullah B, Sultana S. Anti-carcinogenic effect of hesperidin against renal cell carcinoma by targeting COX-2/PGE2 pathway in Wistar rats. Environ Toxicol 2018; 33:1069–77 [DOI] [PubMed] [Google Scholar]
- 38.He S, Wang X, Zhong Y, Tang L, Zhang Y, Ling Y, Tan Z, Yang P, Chen A. Hesperetin post-treatment prevents rat cardiomyoctes from hypoxia/reoxygenation injury in vitro via activating PI3K/akt signaling pathway. Biomed Pharmacother 2017; 91:1106–12 [DOI] [PubMed] [Google Scholar]
- 39.Kamaraj S, Anandakumar P, Jagan S, Ramakrishnan G, Devaki T. Modulatory effect of hesperidin on benzo(a)pyrene induced experimental lung carcinogenesis with reference to COX-2, MMP-2 and MMP-9. Eur J Pharmacol 2010; 649:320–7 [DOI] [PubMed] [Google Scholar]
- 40.Hosseinimehr SJ, Jalayer Z, Naghshvar F, Mahmoudzadeh A. Hesperidin inhibits cyclophosphamide-induced tumor growth delay in mice. Integr Cancer Ther 2012; 11:251–6 [DOI] [PubMed] [Google Scholar]
- 41.Nandakumar N, Balasubramanian MP. Hesperidin a citrus bioflavonoid modulates hepatic biotransformation enzymes and enhances intrinsic antioxidants in experimental breast cancer rats challenged with 7, 12-dimethylbenz (a) anthracene. J Exp Ther Oncol 2012; 9:321–35 [PubMed] [Google Scholar]
- 42.Kamaraj S, Ramakrishnan G, Anandakumar P, Jagan S, Devaki T. Antioxidant and anticancer efficacy of hesperidin in benzo(a)pyrene induced lung carcinogenesis in mice. Invest New Drugs 2009; 27:214–22 [DOI] [PubMed] [Google Scholar]
- 43.Shanmugam MK, Warrier S, Kumar AP, Sethi G, Arfuso F. Potential role of natural compounds as anti-angiogenic agents in cancer. Curr Vasc Pharmacol 2017; 15:503–19 [DOI] [PubMed] [Google Scholar]
- 44.Siveen KS, Ahn KS, Ong TH, Shanmugam MK, Li F, Yap WN, Kumar AP, Fong CW, Terganokar V, Hui KM, Sethi G. Y-tocotrienol inhibits angiogenesis-dependent growth of human hepatocellular carcinoma through abrogation of AKT/mTOR pathway in an orthotopic mouse model. Oncotarget 2014; 5:1897–911 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lentini A, Forni C, Provenzano B, Beninati S. Enhancement of transglutaminase activity and polyamine depletion in B16-F10 melanoma cells by flavonoids naringenin and hesperitin correlate to reduction of the in vivo metastatic potential. Amino Acids 2007; 32:95–100 [DOI] [PubMed] [Google Scholar]
- 46.Yeh MH, Kao ST, Hung CM, Liu CJ, Lee KH, Yeh CC. Hesperidin inhibited acetaldehyde-induced matrix metalloproteinase-9 gene expression in human hepatocellular carcinoma cells. Toxicol Lett 2009; 184:204–10 [DOI] [PubMed] [Google Scholar]
- 47.Lee KH, Yeh MH, Kao ST, Hung CM, Liu CJ, Huang YY, Yeh CC. The inhibitory effect of hesperidin on tumor cell invasiveness occurs via suppression of activator protein 1 and nuclear factor-kappaB in human hepatocellular carcinoma cells. Toxicol Lett 2010; 194:42–9 [DOI] [PubMed] [Google Scholar]
- 48.Yang Y, Wolfram J, Shen H, Fang X, Ferrari M. Hesperetin: an inhibitor of the transforming growth factor-beta (TGF-beta) signaling pathway. Eur J Med Chem 2012; 58:390–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kim GD. Hesperidin inhibits vascular formation by blocking the AKT/mTOR signaling pathways. Prev Nutr Food Sci 2015; 20:221–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhao X, Liu J, Feng L, Ge S, Yang S, Chen C, Li X, Peng L, Mu Y, Wang Y, Gu D, Guo Y, Lin G, Deng B, Cheng Z, Cai D. Anti-angiogenic effects of qingdu granule on breast cancer through inhibiting NFAT signaling pathway. J Ethnopharmacol 2018; 222:261–9 [DOI] [PubMed] [Google Scholar]
- 51.Lee J, Lee J, Kim M, Kim JH. Fermented extraction of citrus unshiu peel inhibits viability and migration of human pancreatic cancers. J Med Food 2018; 21:5–12 [DOI] [PubMed] [Google Scholar]
- 52.Xia R, Xu G, Huang Y, Sheng X, Xu X, Lu H. Hesperidin suppresses the migration and invasion of non-small cell lung cancer cells by inhibiting the SDF-1/CXCR-4 pathway. Life Sci 2018; 201:111–20 [DOI] [PubMed] [Google Scholar]
- 53.Wang Y, Liu S, Dong W, Qu X, Huang C, Yan T, Du J.. Combination of Hesperetin and Platinum Enhances Anticancer Effect on Lung Adenocarcinoma. Biomed Pharmacother 2019;113:108779. [DOI] [PubMed] [Google Scholar]
- 54.Lee J, Kim D-H, Kim J H.. Combined Administration of Naringenin and Hesperetin with Optimal Ratio Maximizes the anti-Cancer Effect in Human Pancreatic Cancer via down Regulation of FAK AND p38 Signaling Pathway. Phytomedicine 2019;58:152762. [DOI] [PubMed] [Google Scholar]
- 55.Banjerdpongchai R, Wudtiwai B, Khaw-On P, Rachakhom W, Duangnil N, Kongtawelert P. Hesperidin from citrus seed induces human hepatocellular carcinoma HepG2 cell apoptosis via both mitochondrial and death receptor pathways. Tumour Biol 2016; 37:227–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lee KA, Lee SH, Lee YJ, Baeg SM, Shim JH. Hesperidin induces apoptosis by inhibiting Sp1 and its regulatory protein in MSTO-211H cells. Biomol Ther 2012; 20:273–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhao J, Li Y, Gao J, De Y. Hesperidin inhibits ovarian cancer cell viability through endoplasmic reticulum stress signaling pathways. Oncol Lett 2017; 14:5569–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ghorbani A, Nazari M, Jeddi-Tehrani M, Zand H. The citrus flavonoid hesperidin induces p53 and inhibits NF-kappaB activation in order to trigger apoptosis in NALM-6 cells: involvement of PPARgamma-dependent mechanism. Eur J Nutr 2012; 51:39–46 [DOI] [PubMed] [Google Scholar]
- 59.Al-Ashaal HA, El-Sheltawy ST. Antioxidant capacity of hesperidin from citrus peel using electron spin resonance and cytotoxic activity against human carcinoma cell lines. Pharm Biol 2011; 49:276–82 [DOI] [PubMed] [Google Scholar]
- 60.Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol 2015; 15:760–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Goldberg JE, Schwertfeger KL. Proinflammatory cytokines in breast cancer: mechanisms of action and potential targets for therapeutics. Curr Drug Targets 2010; 11:1133–46 [DOI] [PubMed] [Google Scholar]
- 62.Mantovani A. Molecular pathways linking inflammation and cancer. Curr Mol Med 2010; 10:369–73 [DOI] [PubMed] [Google Scholar]
- 63.Chai EZ, Siveen KS, Shanmugam MK, Arfuso F, Sethi G. Analysis of the intricate relationship between chronic inflammation and cancer. Biochem J 2015; 468:1–15 [DOI] [PubMed] [Google Scholar]
- 64.Shanmugam MK, Sethi G. Role of epigenetics in inflammation-associated diseases. Subcell Biochem 2013; 61:627–57 [DOI] [PubMed] [Google Scholar]
- 65.Sethi G, Shanmugam MK, Ramachandran L, Kumar AP, Tergaonkar V. Multifaceted link between cancer and inflammation. Biosci Rep 2012; 32:1–15 [DOI] [PubMed] [Google Scholar]
- 66.Blaser H, Dostert C, Mak TW, Brenner D. TNF and ROS crosstalk in inflammation. Trends Cell Biol 2016; 26:249–61 [DOI] [PubMed] [Google Scholar]
- 67.Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Possible new role for NF-kappaB in the resolution of inflammation. Nat Med 2001; 7:1291–7 [DOI] [PubMed] [Google Scholar]
- 68.Minami T, Aird WC. Endothelial cell gene regulation. Trends Cardiovasc Med 2005; 15:174–84 [DOI] [PubMed] [Google Scholar]
- 69.Nizamutdinova IT, Jeong JJ, Xu GH, Lee SH, Kang SS, Kim YS, Chang KC, Kim HJ. Hesperidin, hesperidin methyl chalone and phellopterin from poncirus trifoliata (rutaceae) differentially regulate the expression of adhesion molecules in tumor necrosis factor-alpha-stimulated human umbilical vein endothelial cells. Int Immunopharmacol 2008; 8:670–8 [DOI] [PubMed] [Google Scholar]
- 70.Zarubin T, Han J. Activation and signaling of the p38 MAP kinase pathway. Cell Res 2005; 15:11–8 [DOI] [PubMed] [Google Scholar]
- 71.Zhu Y, Zhu M, Lance P. iNOS signaling interacts with COX-2 pathway in colonic fibroblasts. Exp Cell Res 2012; 318:2116–27 [DOI] [PubMed] [Google Scholar]
- 72.Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009; 30:1073–81 [DOI] [PubMed] [Google Scholar]
- 73.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010; 140:883–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lorzadeh E, Ramezani-Jolfaie N, Mohammadi M, Khoshbakht Y, Salehi-Abargouei A. The effect of hesperidin supplementation on inflammatory markers in human adults: a systematic review and meta-analysis of randomized controlled clinical trials. Chem Biol Interact 2019; 307:8–15 [DOI] [PubMed] [Google Scholar]
- 75.Li X, Xie X, Zhang L, Meng Y, Li N, Wang M, Zhai C, Liu Z, Di T, Zhang L, Li P. Hesperidin inhibits keratinocyte proliferation and imiquimod-induced psoriasis-like dermatitis via the IRS-1/ERK1/2 pathway. Life Sci 2019; 219:311–21 [DOI] [PubMed] [Google Scholar]
- 76.Parhiz H, Roohbakhsh A, Soltani F, Rezaee R, Iranshahi M. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: an updated review of their molecular mechanisms and experimental models. Phytother Res 2015; 29:323–31 [DOI] [PubMed] [Google Scholar]
- 77.Rice-Evans CA, Miller NJ, Paganga G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med 1996; 20:933–56 [DOI] [PubMed] [Google Scholar]
- 78.Heim KE, Tagliaferro AR, Bobilya DJ. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem 2002; 13:572–84 [DOI] [PubMed] [Google Scholar]
- 79.Wilmsen PK, Spada DS, Salvador M. Antioxidant activity of the flavonoid hesperidin in chemical and biological systems. J Agric Food Chem 2005; 53:4757–61 [DOI] [PubMed] [Google Scholar]
- 80.Kamaraj S, Anandakumar P, Jagan S, Ramakrishnan G, Devaki T. Hesperidin attenuates mitochondrial dysfunction during benzo(a)pyrene-induced lung carcinogenesis in mice. Fundam Clin Pharmacol 2011; 25:91–8 [DOI] [PubMed] [Google Scholar]
- 81.Pradeep K, Ko KC, Choi MH, Kang JA, Chung YJ, Park SH. Protective effect of hesperidin, a citrus flavanoglycone, against gamma-radiation-induced tissue damage in Sprague-Dawley rats. J Med Food 2012; 15:419–27 [DOI] [PubMed] [Google Scholar]
- 82.Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem 2009; 284:13291–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Chen X, Ding HW, Li HD, Huang HM, Li XF, Yang Y, Zhang YL, Pan XY, Huang C, Meng XM, Li J. Hesperetin derivative-14 alleviates inflammation by activating PPAR-γ in mice with CCl4-induced acute liver injury and LPS-treated RAW264.7 cells. Toxicol Lett 2017; 274:51–63 [DOI] [PubMed] [Google Scholar]
- 84.Kong LN, Lin X, Huang C, Ma TT, Meng XM, Hu CJ, Wang QQ, Liu YH, Shi QP, Li J. Hesperetin derivatiove-12 (HDND-12) regulates macrophage polarization by modulating JAK2/STAT3 signaling pathway. Chin J Nat Med 2019; 17:122–30 [DOI] [PubMed] [Google Scholar]
- 85.Roohbakhsh A, Parhiz H, Soltani F, Rezaee R, Iranshahi M. Neuropharmacological properties and pharmacokinetics of the citrus flavonoids hesperidin and hesperetin – a mini-review. Life Sci 2014; 113:1–6 [DOI] [PubMed] [Google Scholar]
- 86.Banjerdpongchai R, Khaw-On P. Terpinen-4-ol induces autophagic and apoptotic cell death in human leukemic HL-60 cells. Asian Pac J Cancer Prev 2013; 14:7537–42 [DOI] [PubMed] [Google Scholar]
- 87.Banjerdpongchai R, Punyati P, Nakrob A, Pompimon W, Kongtawelert P. 4'-Hydroxycinnamaldehyde from alpinia galanga (linn.) induces human leukemic cell apoptosis via mitochondrial and endoplasmic reticulum stress pathways. Asian Pac J Cancer Prev 2011; 12:593–8 [PubMed] [Google Scholar]
- 88.Lin W, Tongyi S. Role of bax/bcl-2 family members in green tea polyphenol induced necroptosis of p53-deficient Hep3B cells. Tumour Biol 2014; 35:8065–75 [DOI] [PubMed] [Google Scholar]
- 89.Korga A, Ostrowska M, Jozefczyk A, Iwan M, Wojcik R, Zgorka G, Herbet M, Vilarrubla G G, Dudka J.. Apigenin and Hesperidin Augment the Toxic Effect of Doxorubicin against HepG2 Cells. BMC Pharmacol Toxicol 2019;20:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Febriansah R, Putri DD, Sarmoko Nurulita NA, Meiyanto E, Nugroho AE. Hesperidin as a preventive resistance agent in MCF-7 breast cancer cells line resistance to doxorubicin. Asian Pac J Trop Biomed 2014; 4:228–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Desai UN, Shah KP, Mirza SH, Panchal DK, Parikh SK, Rawal RM. Enhancement of the cytotoxic effects of cytarabine in synergism with hesperidine and silibinin in acute myeloid leukemia: an in-vitro approach. J Cancer Res Ther 2015; 11:352–7 [DOI] [PubMed] [Google Scholar]
- 92.Khamis AAA, Ali EMM, El-Moneim MAA, Abd-Alhaseeb MM, El-Magd MA, Salim EI. Hesperidin, piperine and bee venom synergistically potentiate the anticancer effect of tamoxifen against breast cancer cells. Biomed Pharmacother 2018; 105:1335–43 [DOI] [PubMed] [Google Scholar]
- 93.Dokumacioglu E, Iskender H, Sen T M, Ince I, Dokumacioglu A, Kanbay Y, Erbas E, Saral S.. The Effects of Hesperidin and Quercetin on Serum Tumor Necrosis Factor-Alpha and Interleukin-6 Levels in Streptozotocin-Induced Diabetes Model. Pharmacogn Mag 2018;14:167–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gabizon AA, Patil Y, La-Beck NM. New insights and evolving role of pegylated liposomal doxorubicin in cancer therapy. Drug Resist Updat 2016; 29:90–106 [DOI] [PubMed] [Google Scholar]
- 95.Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 2004; 56:185–229 [DOI] [PubMed] [Google Scholar]
- 96.Agudelo D, Bourassa P, Berube G, Tajmir-Riahi HA. Review on the binding of anticancer drug doxorubicin with DNA and tRNA: structural models and antitumor activity. J Photochem Photobiol B Biol 2016; 158:274–9 [DOI] [PubMed] [Google Scholar]
- 97.Lin S, Hoffmann K, Schemmer P. Treatment of hepatocellular carcinoma: a systematic review. Liver Cancer 2012; 1:144–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Leung TW, Johnson PJ. Systemic therapy for hepatocellular carcinoma. Semin Oncol 2001; 28:514–20 [DOI] [PubMed] [Google Scholar]
- 99.Iskender H, Dokumacioglu E, Sen T M, Ince I, Kanbay Y, Saral S.. The Effect of Hesperidin and Quercetin on Oxidative Stress, NF-Κb and SIRT1 Levels in a STZ-Induced Experimental Diabetes Model. Biomed Pharmacother 2017;90:500–8 [DOI] [PubMed] [Google Scholar]
- 100.Siddiqui IA, Sanna V. Impact of nanotechnology on the delivery of natural products for cancer prevention and therapy. Mol Nutr Food Res 2016; 60:1330–41 [DOI] [PubMed] [Google Scholar]
- 101.Wang S, Zhang J, Chen M, Wang Y. Delivering flavonoids into solid tumors using nanotechnologies. Expert Opin Drug Deliv 2013; 10:1411–28 [DOI] [PubMed] [Google Scholar]
- 102.Majumdar S, Srirangam R. Solubility, stability, physicochemical characteristics and in vitro ocular tissue permeability of hesperidin: a natural bioflavonoid. Pharm Res 2009; 26:1217–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Duranoglu D, Uzunoglu D, Mansuroglu B, Arasoglu T, Derman S. Synthesis of hesperetin-loaded PLGA nanoparticles by two different experimental design methods and biological evaluation of optimized nanoparticles. Nanotechnology 2018;29:395603 [DOI] [PubMed] [Google Scholar]
- 104.Nie S, Xing Y, Kim GJ, Simons JW. Nanotechnology applications in cancer. Annu Rev Biomed Eng 2007; 9:257–88 [DOI] [PubMed] [Google Scholar]
- 105.Merisko-Liversidge E, Liversidge GG, Cooper ER. Nanosizing: a formulation approach for poorly-water-soluble compounds. Eur J Pharm Sci 2003; 18:113–20 [DOI] [PubMed] [Google Scholar]
- 106.Gu SF, Wang LY, Tian YJ, Zhou ZX, Tang JB, Liu XR, Jiang HP, Shen YQ. Enhanced water solubility, antioxidant activity, and oral absorption of hesperetin by D-alpha-tocopheryl polyethylene glycol 1000 succinate and phosphatidylcholine. J Zhejiang Univ Sci B 2019; 20:273–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ansar S, Abudawood M, Alaraj ASA, Hamed SS. Hesperidin alleviates zinc oxide nanoparticle induced hepatotoxicity and oxidative stress. BMC Pharmacol Toxicol 2018; 19:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ferrari PC, Correia MK, Somer A, Ribeiro MA, Astrath NGC, Sato F, Novatski A. Hesperidin-loaded solid lipid nanoparticles: development and physicochemical properties evaluation. J Nanosci Nanotechnol 2019; 19:4747–57 [DOI] [PubMed] [Google Scholar]
- 109.Praveen Kumar P, Sunil Kumar KT, Kavya Nainita M, Sai Tarun A, Raghu Ramdu BG, Deepika K, Pramoda A, Yasmeen C. Cerebroprotective potential of hesperidin nanoparticles against bilateral common carotid artery occlusion reperfusion injury in rats and in silico approaches. Neurotox Res 2019; doi: 10.1007/s12640-019-00098-8. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 110.Menezes PD, Frank LA, Lima BD, de Carvalho YM, Serafini MR, Quintans-Junior LJ, Pohlmann AR, Guterres SS, Araujo AA. Hesperetin-loaded lipid-core nanocapsules in polyamide: a new textile formulation for topical drug delivery. Int J Nanomedicine 2017; 12:2069–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Amani H, Ajami M, Maleki SN, Pazoki-Toroudi H, Daglia M, Sokeng AJ, Di Lorenzo A, Nabavi SF, Devi KP, Nabavi SM. Targeting signal transducers and activators of transcription (STAT) in human cancer by dietary polyphenolic antioxidants. Biochimie 2017; 142:63–79 [DOI] [PubMed] [Google Scholar]
- 112.Ren DY, Xu T, Li R, Huang C, Huang Y, Li RQ, Li HY, Li J. 5,7,3'-Triacetyl hesperetin suppresses adjuvant-induced arthritis in rats through modulating JAK2/STAT3 pathway. Am J Chin Med 2013; 41:601–14 [DOI] [PubMed] [Google Scholar]
- 113.Li R, Cai L, Ren DY, Xie XF, Hu CM. Li Therapeutic effect of 7, 3'-dimethoxy hesperetin on adjuvant arthritis in rats through inhibiting JAK2-STAT3 signal pathway. J Int Immunopharmacol 2012; 14:157–63 [DOI] [PubMed] [Google Scholar]
- 114.Li X, Hu X, Wang J, Xu W, Yi C, Ma R, Jiang H. Inhibition of autophagy via activation of PI3K/akt/mTOR pathway contributes to the protection of hesperidin against myocardial ischemia/reperfusion injury. Int J Mol Med 2018; 42:1917–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Shokoohi M, Shoorei H, Khaki A, Khaki AA, Moghimian M, Abtahi‐Eivary SH. Hesperidin attenuated apoptotic‐related genes in testicle of a male rat model of varicocoele. Andrologia 2020; 8:249–58 [DOI] [PubMed] [Google Scholar]
- 116.Shoorei H, Banimohammad M, Kebria MM, Afshar M, Taheri MM, Shokoohi M, Farashah MS, Eftekharzadeh M, Akhiani O, Gaspar R, Pazoki-Toroudi H. Hesperidin improves the follicular development in 3D culture of isolated preantral ovarian follicles of mice. Exp Biol Med 2019; 244:352–61 [DOI] [PMC free article] [PubMed] [Google Scholar]