Open Access

The antitumor effects of geraniol: Modulation of cancer hallmark pathways (Review)

  • Authors:
    • Minsoo Cho
    • Insuk So
    • Jung Nyeo Chun
    • Ju-Hong Jeon
  • View Affiliations

  • Published online on: March 9, 2016     https://doi.org/10.3892/ijo.2016.3427
  • Pages: 1772-1782
  • Copyright: © Cho et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Geraniol is a dietary monoterpene alcohol that is found in the essential oils of aromatic plants. To date, experimental evidence supports the therapeutic or preventive effects of geraniol on different types of cancer, such as breast, lung, colon, prostate, pancreatic, and hepatic cancer, and has revealed the mechanistic basis for its pharmacological actions. In addition, geraniol sensitizes tumor cells to commonly used chemotherapy agents. Geraniol controls a variety of signaling molecules and pathways that represent tumor hallmarks; these actions of geraniol constrain the ability of tumor cells to acquire adaptive resistance against anticancer drugs. In the present review, we emphasize that geraniol is a promising compound or chemical moiety for the development of a safe and effective multi-targeted anticancer agent. We summarize the current knowledge of the effects of geraniol on target molecules and pathways in cancer cells. Our review provides novel insight into the challenges and perspectives with regard to geraniol research and to its application in future clinical investigation.

1. Introduction

Monoterpenes, specifically the C10 isoprenoids, are dietary compounds that are found in the essential oils of many fruits, vegetables and herbs. Monoterpenes have been considered to play a physiological role in the mediation of plant-environment interactions (1). Monoterpenes have been widely used as flavorings and fragrances in the food and cosmetic industries, and as active ingredients in many pharmaceutical products due to their analgesic, anti-inflammatory, anti-pain and wound healing properties (29). Several dietary monoterpenes, such as d-limonene and perillyl alcohol, have been known to exert chemotherapeutic or chemopreventive effects against cancer (4,1015).

Geraniol is an acyclic monoterpene alcohol that is derived from the essential oils of aromatic plants (see ref. 16 for the detailed physicochemical and toxicological properties of geraniol). In addition to its use in various commercial products, including cosmetics and fine fragrances, geraniol exerts a broad spectrum of pharmacological activities, such as anti-microbial, anti-inflammatory, anti-oxidant, anti-ulcer and neuroprotective activities (1720). Geraniol has garnered attention for its potential role in the treatment of a range of cancers, including breast, lung, colon, prostate, pancreatic and hepatic cancers (2126). In addition, geraniol has been shown to sensitize tumor cells to commonly used chemotherapy agents, such as 5-fluorouracil (5-FU) and docetaxel (25,27,28). Because geraniol is classified into the generally-recognized-as-safe (GRAS) category by the Flavor and Extract Manufacturers Association (FEMA) and the Food and Drug Administration (FDA) of the United States (16), its therapeutic effects against cancer have important clinical and translational implications.

Geraniol has been demonstrated to control multiple signaling molecules and pathways that are involved in various biological processes, such as the cell cycle, cell survival and proliferation, apoptosis, autophagy and metabolism (22,23,25,2931). Multi-target therapeutic agents are more efficacious for the treatment of complex diseases, such as cancer, and are less vulnerable to adaptive resistance (3236). Therefore, geraniol may be a promising resource that may be used as an active ingredient, a lead compound, or a chemical moiety for future drug development. In the present review, we highlight the antitumor activity of geraniol against various types of cancer. We also summarize the current knowledge with regards to the effects of geraniol on target molecules or pathways. Our review provides novel insight into the challenges and perspectives of geraniol research as it pertains to future clinical investigations.

2. The effects of geraniol on cancer hallmarks

Successive genetic lesions promote the transition of a cell from a normal to a neoplastic state because they confer relative fitness benefits, which ensure a high degree of adaptability to altered environmental cues. When tumor cells evolve progressively under selective pressures, they acquire certain cancer hallmarks that enable the tumor to survive and grow and ultimately become malignant (3742). Therefore, the molecules or pathways that are responsible for the acquisition of these hallmarks have been considered promising targets for the development of effective anticancer agents.

Geraniol has been demonstrated to affect many signaling molecules and pathways that represent cancer hallmarks, such as sustaining proliferative signaling (26,4347), evading growth suppressors (30), enabling replicative immortality (22,23,25,28,46,4850), tumor-promoting inflammation (68,45,46,5053), inducing angiogenesis (46,54), genome instability and mutation (49), and resisting cell death (25,26,30,45,46,49,50,54) and deregulating cellular energetics (23,24,26,2931,47,52). The pleiotropic effects of geraniol on cancer hallmarks are schematically illustrated in Fig. 1 and are summarized in Table I. However, the detailed molecular mechanisms through which geraniol functions will be described in the next section. These results suggest that geraniol effectively abolishes various properties of tumor cells that allow them to adapt and survive under selective pressure.

Table I

The effects of geraniol on cancer hallmarks.

Table I

The effects of geraniol on cancer hallmarks.

Cancer hallmarkThe affected molecule or pathway (ref.)
Sustaining proliferative signalingc-fos↓ (46), cholesterol↓ (26,44), ERK1/2↓ (43,45), PKC↓ (43), Ras↓ (26,45), Raf↓ (45), RhoA↓ (47)
Evading growth suppressorspAKT↓, pmTOR↓ (30)
Avoiding immune destructionN.D.a
Enabling replicative immortalityPCNA↓ (46,50), cyclin-A↓ (23,25,48), cyclin-B↓ (25,48), cyclin-D1↓ (25,46,48), cyclin-E↓ (23,25), CDK1↓, p27Kip1↑ (25,48), CDK2↓ (48), CDK4↓ (23,25), p21Cip1↑ (25,48), ODC↓ (22), TS↓, TK↓ (28)
Tumor-promoting inflammationCAT↑ (51,52), COX-2↓ (45,46,52), GPX↑ (45,50,52), reduced-GSH↑ (45,50,52), GST↑ (45,50), GR↑, MDA↓ (45,52), NF-κB↓ (50,52), SOD↑ (51,52), vitamin E↑ (51), IL-1β↓, IL-6↓, TNF-α↓, NO↓, pp38↓, QR↑ (52), n-Nrf-2↑ (53)
Activating invasion and metastasisN.D.
Inducing angiogenesisVEGF↓ (46), VEGFR-2↓ (54)
Genome instability and mutationDNA damage↓ (49)
Resisting cell deathATG5↑, LC3↑ (30), Bax↑ (25,45,46), Bcl-2↓ (25,30,45,46,49), Bax↑, Bcl-xL↓ (30), Bcl-w↓, BNIP3↑ (25), caspase-3↑ (25,26,30,46,50,54), caspase-8↑ (50), caspase-9↑ (46,50), Kim-1↓ (50), p53↓ (46,50)
Deregulating cellular energeticsG6PD↑ (52), HMG-CoA reductase↓ (23,26,29,31,47), pAMPK↑ (30)

a N.D., not determined.

{ label (or @symbol) needed for fn[@id='tfn2-ijo-48-05-1772'] } Arrows indicate upregulation or downregulation of the indicated molecules in terms of expression or activity. AMPK, AMP-activated protein kinase; ATG5, autophagy-related protein 5; Bax, Bcl2-associated X protein; Bcl-xL, B-cell lymphoma-extra large; Bcl-w, Bcl-2-like 2; Bcl-2, B-cell lymphoma 2; BNIP3, Bcl2/adenovirus E1B 19 kDa interacting protein 3; CAT, catalase; CDK, cyclin-dependent kinase; COX-2, cyclooxygenase-2; ERK, extracellular signal-regulated kinase; GPX, glutathione peroxidase; GR, glutathione reductase; GSK-3β, glycogen synthase kinase-3β; GSH, glutathione; G6PD, glucose-6-phosphate dehydrogenase; HMG-CoA, 3-hydroxy-3-methyl-glutaryl-CoA; IL, interleukin; iNOS, inducible nitric oxide synthase; Kim-1, kidney injury molecule-1; LC3, microtubule-associated protein light chain 3; MDA, malondialdehyde; MPO, myeloperoxidase; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor-kappa B; NO, nitric oxide; n-Nrf-2, nuclear Nrf-2; ODC, ornithine decarboxylase; PCNA, proliferating cell nuclear antigen; PGE2, prostaglandin E2; pIκB-α, phospho-I kappa B-alpha; PKC, protein kinas C; PPARγ, peroxisome proliferator-activated receptor gamma; RhoA, Ras homolog gene family member A; QR, quinone reductase; SOD, superoxide dismutase; TBARS, thiobarbituric acid reactive substances; TNF-α, tumor necrosis factor-α; TS, thymidylate synthase; TK, thymidine kinase, VEGF, vascular endothelia growth factor.

3. The effects of geraniol on various types of cancer

Geraniol has been found to be effective against a broad range of cancers, including breast, lung, colon, prostate, pancreatic, skin, liver, kidney and oral cancers (Fig. 2). The molecular mechanisms that underlie the therapeutic effects of geraniol on various types of cancer are summarized in Table II. Of cancer hallmarks, enabling replicative immortality and resisting cell death appear to be the most central targets of geraniol (Table II). However, it is emphasized that this can be research or publication bias: for example, it is still not reported whether geraniol affects avoidance of immune destruction or activating invasion and metastasis. The effective dose range of geraniol in vitro (cell culture), and in in vivo (xenograft or chemical carcinogenesis) models are presented in Table III.

Table II

The antitumor activity of geraniol against cancer.

Table II

The antitumor activity of geraniol against cancer.

Cancer hallmarks (ref.)

Sustaining proliferative signalingEvading growth suppressorsAvoiding immune destructionEnabling replicative immortalityTumor-promoting inflammationActivating invasion and metastasisInducing angiogenesisGenome instability and mutationResisting cell deathDeregulating cellular energetics
Breast cancerN.D.aN.D.N.D.CDK2↓, CDK4↓, cyclin-D1↓, cyclin-E↓, cyclin-A↓, p27Kip1↑ (23)N.D.N.D.N.D.N.D.N.D.HMG-CoA reductase↓ (23)
Lung cancerRas↓ (26)N.D.N.D.N.D.N.D.N.D.N.D.N.D.Caspase-3↑ (26)HMG-CoA reductase↓ (26)
Colon cancerERK1/2↓, PKC↓ (43)N.D.N.D.ODC↓ (22), TS↓, TK↓ (28)N.D.N.D.VEGFR-2↓ (54)DNA damage↓ (49Bcl-2↓ (49), caspase-3↑ (54)N.D.
Prostate cancerN.D.pAKT↓, pmTOR↓ (30)N.D.CDK1↓, CDK4↓, cyclin-A↓, cyclin-B↓, cyclin-D↓, cyclin-E↓, p21Cip1↑, p27Kip1↑ (25)N.D.N.D.N.D.N.D.ATG5↑, LC3↑, Bcl-xL↓ (30), Bax↑, Bcl-2↓, caspase-3↑ (25,30), Bcl-w↓, Bax↑, BNIP3↑ (25)pAMPK↑ (30)
Pancreatic cancerN.D.N.D.N.D.CDK2↓, cyclin-A↓, cyclin-B1↓, cyclin-D1↓, p21Cip1↑, p27Kip1↑ (48)N.D.N.D.N.D.N.D.N.D.N.D.
Skin cancerERK1/2↓, Raf↓, Ras↓ (45)N.D.N.D.N.D.CAT↑, COX-2↓, GPX↑, reduced-GSH↑, GR↑, MDA↓, (45,52), GST↑ (45), GR↓, IL-1β↓, IL-6↓, NF-κB↓, NO↓, pp38↓, QR↓, SOD↓, TNF-α↓ (52)N.D.N.D.N.D.Bax↑, Bcl-2↓ (45)G6PD↑ (52)
Liver cancerRhoA↓ (47)N.D.N.D.N.D.N.D.N.D.N.D.N.D.N.D.HMG-CoA reductase↓ (29,31,47)
Kidney cancerN.D.N.D.N.D.PCNA↓ (50)CAT↑, GPX↑, GST↑, reduced-GSH↑, NF-κB↓ (50)N.D.N.D.N.D.Caspase-3↑, caspase-8↑, caspase-9↑, Kim-1↓, p53↓ (50)N.D.
Oral cancerc-fos↓ (46)N.D.N.D.Cyclin-D1↓, PCNA↓ (46)CAT↑, GPX↑, GSH↑, SOD↑ vitamin E↑ (51), COX-2↓, NF-κB↓, p53↓ (46), n-Nrf-2↑ (53)N.D.VEGF↓ (46)N.D.Bax↑, Bcl-2↓, caspase-3↑, caspase-9↑ (46)N.D.

a N.D., not determined.

{ label (or @symbol) needed for fn[@id='tfn4-ijo-48-05-1772'] } Arrows indicate upregulation or downregulation of the indicated molecules in terms of expression or activity.

Table III

The effective doses of geraniol in cancer models.

Table III

The effective doses of geraniol in cancer models.

CancerModelIn vivo routeEffective dose (ref.)
BreastMCF-7 cell culture300, 500 and 700 μM (23)
LungA549 cell culture727.2 μM (26)
A549 xenograftOral25, 50 and 75 mmol/kg (26)
ColonCaco-2 cell culture150 and 400 μM (22,27,28,43)
SW620 cell culture150 μM (28)
TC118 xenograftI.P.a150 mg/kg (28)
Dimethylhydrazine-treated Wistar ratI.P.250 mg/kg (49)
ProstatePC-3 cell culture250, 500 and 1,000 μM (25,30)
PC-3 xenograftI.T.a60 mg/kg (25)
PancreaticMIA PaCa-2 cell culture100, 200, 400 and 500 μM (21,48)
BxPC-3 cell culture10, 20, 40, 80, 160, 400 and 800 μM (48,60)
PC-1 xenograftOral20 g/kg (21)
Skin 12-O-tetradecanoylphorbolDermal10, 50 and 100 mg/kg (45,52)
13-acetate-treated mice
LiverHepG2 cell culture100, 200 and 400 μM (29,31,61)
Morris hepatoma 7777 xenograftOral23 mmol/kg (62)
2-acetylaminofluorene-treated Wistar ratOral250 mg/kg (24,47)
KidneyFerric nitrilotriacetate-treated Wistar ratOral100 and 200 mg/kg (50)
Oral 7,12-dimethylbenz[α]anthracene-treated hamsterOral250 and 500 mg/kg (46,51)
4-niroquinoline-1-oxide-treated miceOral200 mg/kg (53)
Other cancersB16 melanoma cell culture150 and 160 μM (64,66)
B16 xenograftOral6.5 and 65 mmol/kg (62)
P388 leukemia cell culture900 μM (63)

a I.P., intraperitoneal injection; I.T., intratumoral injection.

Breast cancer

Geraniol suppressed the growth of MCF-7 breast cancer cells via the induction of cell cycle arrest in the G1 phase (23). The protein levels of cyclin D1, cyclin-dependent kinase 4 (CDK4), cyclin E, and cyclin A were reduced in geraniol-treated MCF-7 cells, whereas those of p27Kip1 were elevated by geraniol (23). In contrast, geraniol did not affect the growth of MCF-10F normal breast epithelial cells (23), which indicates that its activity is tumor-specific. Although geraniol inhibited 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase activity, geraniol-induced cell cycle arrest was not rescued by supplementation with mevalonate, which is a reaction product of HMG-CoA reductase (23). These results indicate that the anti-proliferative effect of geraniol on MCF-7 cells is not related to a reduction in HMG-CoA reductase activity or the limitation of cellular mevalonate levels, but rather, that it is linked to other molecular mechanisms.

Lung cancer

Experiments in xenograft models revealed that geraniol reduced tumor weight and volume in mice bearing tumors that formed from A549 cells (26), which showed an increased TUNEL-positive tumor cell population and elevated caspase-3 activity. Geraniol also inhibited cholesterol synthesis in A549 cells and reduced the serum levels of cholesterol in mice with A549 cell xenografts through the suppression of HMG-CoA reductase activity (26). In addition, geraniol reduced the expression levels of membrane-bound Ras in A549 cell-bearing mice without corresponding changes in the total protein levels of Ras (26). This was probably due to the actions of geraniol that are related to the inhibition of mevalonate pathways and the resultant reduction in Ras prenylation (55). However, it is unclear whether mevalonate supplementation can rescue geraniol-induced phenotypes.

Colon cancer

Geraniol exerted anti-proliferative activity against Caco-2 colorectal adenocarcinoma cells through the induction of S phase cell cycle arrest (22). In addition, geraniol elicited apoptosis in mouse xenograft models using CT26 colon carcinoma cells (54). Geraniol affected polyamine metabolism in Caco-2 cells through the reduction of ornithine decarboxylase (ODC) activity (22). This is clinically relevant because polyamine metabolism is a promising target of chemotherapeutic and chemopreventive drug development strategies (56). Geraniol also induced the depolarization of the plasma membrane potential with a decrease in membrane resistance in Caco-2 cells (43), which led to the reduction in the activities of protein kinase C (PKC) and extracellular signal-regulated protein kinases (ERK) (43). Under the experimental conditions, mevalonate supplementation did not reverse geraniol-mediated inhibition of cell growth (43), which indicates that HMG-CoA reductase is not linked to the antitumor actions of geraniol.

Geraniol sensitized cultured Caco-2 cells and SW620 colorectal adenocarcinoma cells to 5-FU and also sensitized mice with xenografts of TC118 colon cancer cells to 5-FU (27,28). In addition, geraniol elevated the uptake rate of 5-FU in colon cancer cells (27). Geraniol effectively decreased the expression levels of thymidine kinase and thymidylate synthase (28), which are important targets for the development of anticancer drugs and chemosensitizing agents (5759). These results suggest that geraniol might serve as a chemosensitizer.

Geraniol was found to elicit chemopreventive activities (49). It decreased the total number of aberrant crypt foci in the colons of Wistar rats that were treated with dimethylhydrazine (49). Under this condition, geraniol reduced DNA damage in the colonic mucosa. In addition, apoptosis in the distal colon was significantly higher in the geraniol-treated group compared with the control group. Moreover, the expression levels of Bcl-2 were significantly decreased in the geraniol-treated group (49).

Prostate cancer

Geraniol inhibited tumor cell growth via the induction of apoptosis in cultured and grafted PC-3 prostate cancer cells (25,30) due to the depolarization of mitochondrial membrane potential, the activation of caspase-3, the reduction in the expression levels of Bcl-2 and Bcl-w, and the increased expression levels of Bax and BNIP3 (25). Geraniol also potently induced autophagy, which is crucial for the effective stimulation of cell death (30). The combined inhibition of apoptosis and autophagy restored cell growth in geraniol-treated PC-3 cells. In addition, cell cycle arrest at G1 phase, and a slight arrest at G2 phase, were observed in geraniol-treated PC-3 cells (25,30), which was confirmed by the decreased expression levels of cyclins A, B, D and E and CDK1 and CDK4, as well as by the elevated levels of p21Cip1 and p27Kip1.

At the molecular level, geraniol has been found to inhibit AKT-mTOR signaling without affecting mitogen-activated protein kinase (MAPK) activity (30). For example, the PP2A inhibitor okadaic acid, but not the PP1 inhibitor tautomycin, reversed geraniol-mediated inhibition of AKT. Concomitantly, geraniol activated AMP-activated protein kinase (AMPK). The combined treatment with the AKT inhibitor perifosine and the AMPK activator AICAR synergistically inhibited tumor cell growth (30). However, under the condition, geraniol did not affect the activity of transient receptor potential melastatin type 8 (TRPM8) channel or calcium mobilization (25,30). In addition, mevalonate supplementation did not reverse the geraniol-mediated inhibition of cell growth (30).

Geraniol increased the therapeutic response of PC-3 cells to chemotherapy agents, such as docetaxel, doxorubicin, paclitaxel, etoposide, 5-FU and cisplatin (25). Particularly, the combination of the sub-optimal doses of geraniol and docetaxel noticeably suppressed the growth of prostate cancer cells in culture and those in mouse xenografts; the percentage of active caspase-3-positive cells was markedly increased in tumor tissues that were treated with geraniol and docetaxel compared with the treatment of cells with either drug alone.

Pancreatic cancer

Geraniol inhibited the proliferation of MIA PaCa-2 pancreatic carcinoma cells and BxPC-3 pancreatic adenocarcinoma cells and reduced tumor volume in hamsters injected with PC-1 pancreatic ductal adenocarcinoma cells (21,48,60). In addition, geraniol induced G1 phase cell cycle arrest in MIA PaCa-2 cells (48) and elevated the expression levels of p21Cip1 and p27Kip1 but reduced those of cyclin A, cyclin B1 and CDK2. siRNAs against p21Cip1 and p27Kip1 subsequently alleviated geraniol-induced cell cycle arrest (48). Finally, the protein levels of cyclin D1 were reduced by geraniol in MIA PaCa-2 cells, but not in BxPC-3 cells (48).

When the tumor cells were incubated with geraniol before they were treated with gemcitabine, which is one of the first-line treatments for advanced pancreatic cancer, proliferation of BxPC-3 cells was significantly inhibited compared with when the cells were treated with either drug alone (60). In addition, geraniol markedly enhanced the apoptosis-inducing effect of gemcitabine on BxPC-3 cells (60).

Skin cancer

Geraniol inhibited skin edema and epidermal hyperplasia in mice that were stimulated with 12-O-tetradecanolyphorbal-13-acetate (TPA)/7, 12-dimethyl-benzanthracene (DMBA) (45,52). The chemopreventive effects of geraniol on skin tumorigenesis are ascribed to the activation of apoptotic pathways and the attenuation of Ras-mediated cell proliferation pathways. Particularly, geraniol reduced the levels of membrane-bound Ras in DMBA/TPA-stimulated mice (45) and also decreased Raf expression and ERK activity. However, it is unclear whether geraniol inhibits HMG-CoA reductase activity in this setting and whether mevalonate rescues the geraniol-induced phenotypic changes. In addition, geraniol suppressed the TPA-induced increase in cutaneous ODC activity and the thymidine incorporation rate in mouse skin (45).

Geraniol suppressed skin inflammation through the inhibition of cyclooxygenase (COX)-2 induction in DMBA/TPA-treated mice (45). Geraniol also inhibited the altered expression of nuclear factor (NF)-κB; the production of cutaneous proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β and pp38; and the formation of superoxide dismutase (SOD) and nitric oxide (NO) (52). In addition, geraniol exerted antioxidant activity to reduce TPA-induced malondialdehyde (MDA) production through modulation of the following: glutathione (GSH), glutathione reductase (GR), glutathione peroxidase (GPX), glutathione-S-transferase (GST), catalase (CAT), lipid peroxidation, glucose 6-phosphate dehydrogenase (G6PD) and quinone reductase (QR) (45,52).

Liver cancer

In one study, geraniol exerted anti-proliferative activity in HepG2 hepatocarcinoma cells (29,31,61), and G1 phase cell cycle arrest and apoptosis were observed in geraniol-treated cells (31). In addition, geraniol suppressed tumor growth in mice that received transplanted Morris 7777 hepatoma cells (62). Geraniol inhibited HMG-CoA reductase activity in HepG2 cells and 2-acetylaminofluorene-treated Wistar rat (29,47) and reduced the level of RhoA in hepatic membranes (47). Although geraniol affected various lipid metabolic pathways, including HMG-CoA reductase or Δ5-desaturase (29,31), it is uncertain whether its effects are linked to tumor suppression.

Geraniol exhibited chemopreventive activity against hepatocarcinogenesis in rats that were stimulated with diethylnitrosamine and 2-acetylaminofluorene. It also inhibited cell proliferation and DNA damage in preneoplastic lesions (PNLs) and caused an elevation in apoptosis in PNLs (24,47).

Kidney cancer

Geraniol demonstrated chemopreventive activity in Wistar rats treated with ferric nitrilotriacetate (Fe-NTA) (50) in that it effectively reduced renal oxidative stress and tumor incidence via the modulation of multiple molecular targets that are involved in proliferation, apoptosis, or inflammation. Geraniol restored the activities of GSH-dependent antioxidant enzymes, such as glutathione S-transferase, glutathione peroxidase, and catalase in Fe-NTA-injected rats, which suppressed Fe-NTA-induced renal toxicity (50). In the rats that were treated with Fe-NTA and N-nitrosodiethylamine, geraniol downregulated kidney injury molecule-1 (Kim-1), NF-κB, proliferating cell nuclear antigen (PCNA), and p53 and upregulated caspase-3, -8 and -9 (50).

Oral squamous cell carcinoma

Geraniol prevented DMBA-induced buccal pouch carcinogenesis in the hamster (46,51). Its anti-inflammatory, anti-angiogenic, anti-proliferative, and apoptosis-inducing properties contributed to the suppression of histological changes, such as keratosis, dysplasia and hyperplasia, in the buccal pouch of DMBA-treated hamsters. Geraniol also exerted its chemopreventive potential against 4-nitroquinoline-1-oxide (4NQO)-induced oral carcinogenesis in Wistar rats (53).

Geraniol reduced the plasma concentrations of lipid peroxidation by-products in DMBA-stimulated hamsters (51). Geraniol also restored the DMBA-induced disturbance in the status of antioxidants in terms of glutathione peroxidase, reduced glutathione, vitamin E, superoxide dismutase, and catalase in the plasma and in the buccal mucosa (51). In addition, the chemopreventive potential of geraniol relies on modulatory effects on phase I (cytochrome P450 and cytochrome b5) and II (glutathione-S-transferase, glutathione reductase and DT-diaphorase) detoxification enzymes to excrete carcinogenic metabolites during DMBA-induced buccal pouch carcinogenesis in the hamster (51). Similar effects of geraniol were observed in 4NQO-stimulated models of oral squamous cell carcinoma (53).

Geraniol reduced the expression levels of Bcl-2, PCNA, mutant p53, VEGF, c-fos, COX-2, NF-κB, and cyclin D1, whereas it elevated Bax, caspase-3 and -9 activity in DMBA-treated hamsters (46). In addition, geraniol increased the nuclear levels of nuclear factor erythroid 2-related factor 2 (Nrf-2), a redox-sensitive transcription factor, in 4NQO-stimulated rats (53).

Other cancers

Geraniol inhibited cell growth in P388 leukemia cells (63). Geraniol exerted anti-proliferative activity against B16 melanoma cells in culture and in xenograft models (6366). Mevalonate supplementation rescued geraniol-induced phenotypes of P388 leukemia cells, but not those of B16 melanoma cells (63). These results suggest that limitations in mevalonate levels suppress tumor growth in a cancer type-specific manner.

4. Discussion and future perspective

Geraniol has been approved as GRAS by FDA and FEMA. The acute oral LD50 was calculated as 3.6 to 4.8 g/kg in rats and the intramuscular LD50 was reported to be 4 g/kg. In addition, dose toxicity was only slightly observed in repeated administration of geraniol at 1,000 mg/kg/day in the diet for 16 weeks (see the ref. 16 for the detailed toxicological data of geraniol). The recent experimental observations obtained from in vitro and in vivo models have supported the idea that geraniol exerts therapeutic, chemosensitizing, or preventive effects against different cancer types by modulating multiple cancer hallmark pathways (Tables I and II). Particularly, the antitumor effects of geraniol were achieved by oral delivery in different cancer types (Table III), which is unlike poor oral bioavailability of many phytochemicals (67). These results suggest that geraniol might be beneficial for achieving a balance between therapeutic outcome and adverse drug responses and useful for improvement in quality of life of patients and palliative treatment. Therefore, geraniol might be promising for further development as a lead compound or a chemical moiety.

The non-toxic properties of geraniol (16) and the experimental results summarized in this review have demonstrated that geraniol specifically suppresses tumor growth, but does not affect normal physiology at an individual level. Geraniol commonly affects the signaling molecules that are involved in the acquisition of cancer hallmarks, particularly those that involve the cell cycle and apoptosis, across various types of cancer, as summarized in Tables I and II. These findings suggest that geraniol elicits tumor-specific effects through common molecular mechanisms. However, several geraniol-reactive signaling pathways are also important physiological cell functions. For example, geraniol has been shown to inhibit angiogenesis, which does not only occur during tumor growth but also during embryo development, wound healing and other physiological conditions (68). Therefore, the cancer type-specific actions and the adverse effects of geraniol need to be carefully evaluated.

HMG-CoA reductase catalyzes the synthesis of mevalonate, which is the committed step in the synthesis of prenyl groups (55) and is the limiting step in cholesterol biosynthesis (69). The expression levels of HMG-CoA reductase are frequently upregulated in various types of cancer in humans (23,26,29,31,47). It has been shown that geraniol inhibits HMG-CoA reductase in most types of tumor cells, which raises the possibility that the reduced prenylation of small G-proteins, such as Ras or RhoA, accounts for the antitumor effects of geraniol. However, mevalonate supplementation did not reverse geraniol-induced growth suppression in any type of cancer tested (23,26) except leukemia (63). These results suggest that geraniol-induced molecular changes do not necessarily induce phenotypic outcomes and that the inhibition of HMG-CoA reductase might be effective against specific types of cancer, including leukemia.

The molecular mechanisms by which geraniol exerts its anticancer effects are still largely unknown. Therefore, much effort is required to obtain additional information on the effectiveness and efficacy of geraniol. In this regard, computational approaches using omics technology-driven data should be emphasized to broaden our understanding of the pharmacological mechanisms of action of geraniol and for the translation of this biological knowledge into clinical practice (7076). Data-driven approaches allow us novel insights into geraniol-induced changes in terms of the molecular makeup of biological systems, which contributes to the efficient discovery of the latent target molecules of geraniol.

There seem to be no doubt that geraniol inhibits cancer growth in vitro and in vivo models. However, in vitro models, submillimolar concentrations of geraniol have been used to define the therapeutic effects and the mechanism of action (Table III). This may be a weakness of geraniol studies because these concentrations are not clinically relevant. Nonetheless, geraniol effectively and efficiently suppresses tumor growth in in vivo models. These findings are similar to those of metformin, an anti-diabetic drug. Although milli-molar concentrations of metformin usually exerts anticancer activity in in vitro models, there is consensus that metformin is a clinically promising cancer therapeutic agent (77,78). Therefore, it is crucial that the mechanisms of action of geraniol are elucidated in in vivo studies.

The present review has highlighted the pleiotropic mechanisms of action of geraniol in various types of cancer. Geraniol is therefore useful in the development of multi-target therapies against cancer, and our review provides a scientific basis for future basic and translational research.

Acknowledgements

We are very grateful to Ms. Da-Kyung Lee, Ms. Young Hyun Yun, and Prof. Jeong Han Kim (Bio-Medical Arts and Design Education Center, Department of Contemporary Art and Ceramics and Fiber Arts, College of Fine Art, Seoul Women's University) for preparation of the figures. This research was supported by the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT and Future Planning (2014R1A2A1A11050616) and the Education and Research Encouragement Fund of Seoul National University Hospital.

References

1 

Mahmoud SS and Croteau RB: Strategies for transgenic manipulation of monoterpene biosynthesis in plants. Trends Plant Sci. 7:366–373. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Guimarães AG, Quintans JS and Quintans LJ Jr: Monoterpenes with analgesic activity: a systematic review. Phytother Res. 27:1–15. 2013. View Article : Google Scholar

3 

de Cássia da Silveira e Sá R, Andrade LN and de Sousa DP: A review on anti-inflammatory activity of monoterpenes. Molecules. 18:1227–1254. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Bhalla Y, Gupta VK and Jaitak V: Anticancer activity of essential oils: A review. J Sci Food Agric. 93:3643–3653. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Barreto RS, Albuquerque-Júnior RL, Araújo AA, Almeida JR, Santos MR, Barreto AS, DeSantana JM, Siqueira-Lima PS, Quintans JS and Quintans-Júnior LJ: A systematic review of the wound-healing effects of monoterpenes and iridoid derivatives. Molecules. 19:846–862. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Hasan SK and Sultana S: Geraniol attenuates 2-acetylamino-fluorene induced oxidative stress, inflammation and apoptosis in the liver of wistar rats. Toxicol Mech Methods. 25:559–573. 2015.

7 

Medicherla K, Sahu BD, Kuncha M, Kumar JM, Sudhakar G and Sistla R: Oral administration of geraniol ameliorates acute experimental murine colitis by inhibiting pro-inflammatory cytokines and NF-κB signaling. Food Funct. 6:2984–2995. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Soubh AA, Abdallah DM and El-Abhar HS: Geraniol ameliorates TNBS-induced colitis: Involvement of Wnt/β-catenin, p38MAPK, NF-κB, and PPARγ signaling pathways. Life Sci. 136:142–150. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Kozioł A, Stryjewska A, Librowski T, Sałat K, Gaweł M, Moniczewski A and Lochyński S: An overview of the pharmacological properties and potential applications of natural monoterpenes. Mini Rev Med Chem. 14:1156–1168. 2014. View Article : Google Scholar

10 

Gould MN: Cancer chemoprevention and therapy by monoterpenes. Environ Health Perspect. 105(Suppl 4): 977–979. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Crowell PL: Prevention and therapy of cancer by dietary monoterpenes. J Nutr. 129:775S–778S. 1999.PubMed/NCBI

12 

Chen TC, Fonseca CO and Schönthal AH: Preclinical development and clinical use of perillyl alcohol for chemoprevention and cancer therapy. Am J Cancer Res. 5:1580–1593. 2015.PubMed/NCBI

13 

Samaila D, Toy BJ, Wang RC and Elegbede JA: Monoterpenes enhanced the sensitivity of head and neck cancer cells to radiation treatment in vitro. Anticancer Res. 24:3089–3095. 2004.PubMed/NCBI

14 

Bardon S, Foussard V, Fournel S and Loubat A: Monoterpenes inhibit proliferation of human colon cancer cells by modulating cell cycle-related protein expression. Cancer Lett. 181:187–194. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Sobral MV, Xavier AL, Lima TC and de Sousa DP: Antitumor activity of monoterpenes found in essential oils. Sci World J. 2014:9534512014. View Article : Google Scholar

16 

Lapczynski A, Bhatia SP, Foxenberg RJ, Letizia CS and Api AM: Fragrance material review on geraniol. Food Chem Toxicol. 46(Suppl 11): S160–S170. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Solórzano-Santos F and Miranda-Novales MG: Essential oils from aromatic herbs as antimicrobial agents. Curr Opin Biotechnol. 23:136–141. 2012. View Article : Google Scholar

18 

Tiwari M and Kakkar P: Plant derived antioxidants - Geraniol and camphene protect rat alveolar macrophages against t-BHP induced oxidative stress. Toxicol In Vitro. 23:295–301. 2009. View Article : Google Scholar : PubMed/NCBI

19 

de Carvalho KI, Bonamin F, Dos Santos RC, Périco LL, Beserra FP, de Sousa DP, Filho JM, da Rocha LR and Hiruma-Lima CA: Geraniol - a flavoring agent with multifunctional effects in protecting the gastric and duodenal mucosa. Naunyn Schmiedebergs Arch Pharmacol. 387:355–365. 2014. View Article : Google Scholar

20 

Rekha KR, Selvakumar GP, Sethupathy S, Santha K and Sivakamasundari RI: Geraniol ameliorates the motor behavior and neurotrophic factors inadequacy in MPTP-induced mice model of Parkinson's disease. J Mol Neurosci. 51:851–862. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Burke YD, Stark MJ, Roach SL, Sen SE and Crowell PL: Inhibition of pancreatic cancer growth by the dietary isoprenoids farnesol and geraniol. Lipids. 32:151–156. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Carnesecchi S, Schneider Y, Ceraline J, Duranton B, Gosse F, Seiler N and Raul F: Geraniol, a component of plant essential oils, inhibits growth and polyamine biosynthesis in human colon cancer cells. J Pharmacol Exp Ther. 298:197–200. 2001.PubMed/NCBI

23 

Duncan RE, Lau D, El-Sohemy A and Archer MC: Geraniol and beta-ionone inhibit proliferation, cell cycle progression, and cyclin-dependent kinase 2 activity in MCF-7 breast cancer cells independent of effects on HMG-CoA reductase activity. Biochem Pharmacol. 68:1739–1747. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Ong TP, Heidor R, de Conti A, Dagli ML and Moreno FS: Farnesol and geraniol chemopreventive activities during the initial phases of hepatocarcinogenesis involve similar actions on cell proliferation and DNA damage, but distinct actions on apoptosis, plasma cholesterol and HMGCoA reductase. Carcinogenesis. 27:1194–1203. 2006. View Article : Google Scholar

25 

Kim SH, Bae HC, Park EJ, Lee CR, Kim BJ, Lee S, Park HH, Kim SJ, So I, Kim TW, et al: Geraniol inhibits prostate cancer growth by targeting cell cycle and apoptosis pathways. Biochem Biophys Res Commun. 407:129–134. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Galle M, Crespo R, Kladniew BR, Villegas SM, Polo M and de Bravo MG: Suppression by geraniol of the growth of A549 human lung adenocarcinoma cells and inhibition of the mevalonate pathway in culture and in vivo: Potential use in cancer chemotherapy. Nutr Cancer. 66:888–895. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Carnesecchi S, Langley K, Exinger F, Gosse F and Raul F: Geraniol, a component of plant essential oils, sensitizes human colonic cancer cells to 5-Fluorouracil treatment. J Pharmacol Exp Ther. 301:625–630. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Carnesecchi S, Bras-Gonçalves R, Bradaia A, Zeisel M, Gossé F, Poupon MF and Raul F: Geraniol, a component of plant essential oils, modulates DNA synthesis and potentiates 5-fluorouracil efficacy on human colon tumor xenografts. Cancer Lett. 215:53–59. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Polo MP and de Bravo MG: Effect of geraniol on fatty-acid and mevalonate metabolism in the human hepatoma cell line Hep G2. Biochem Cell Biol. 84:102–111. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Kim SH, Park EJ, Lee CR, Chun JN, Cho NH, Kim IG, Lee S, Kim TW, Park HH, So I, et al: Geraniol induces cooperative interaction of apoptosis and autophagy to elicit cell death in PC-3 prostate cancer cells. Int J Oncol. 40:1683–1690. 2012.

31 

Crespo R, Montero Villegas S, Abba MC, de Bravo MG and Polo MP: Transcriptional and posttranscriptional inhibition of HMGCR and PC biosynthesis by geraniol in 2 Hep-G2 cell proliferation linked pathways. Biochem Cell Biol. 91:131–139. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zimmermann GR, Lehár J and Keith CT: Multi-target therapeutics: When the whole is greater than the sum of the parts. Drug Discov Today. 12:34–42. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Efferth T and Koch E: Complex interactions between phytochemicals. The multi-target therapeutic concept of phytotherapy. Curr Drug Targets. 12:122–132. 2011. View Article : Google Scholar

34 

Rather MA, Bhat BA and Qurishi MA: Multicomponent phyto-therapeutic approach gaining momentum: Is the ‘one drug to fit all’ model breaking down? Phytomedicine. 21:1–14. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Tao L, Zhu F, Xu F, Chen Z, Jiang YY and Chen YZ: Co-targeting cancer drug escape pathways confers clinical advantage for multi-target anticancer drugs. Pharmacol Res. 102:123–131. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Huang S and Kauffman S: How to escape the cancer attractor: Rationale and limitations of multi-target drugs. Semin Cancer Biol. 23:270–278. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Floor SL, Dumont JE, Maenhaut C and Raspe E: Hallmarks of cancer: Of all cancer cells, all the time? Trends Mol Med. 18:509–515. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Ellenbroek SI and van Rheenen J: Imaging hallmarks of cancer in living mice. Nat Rev Cancer. 14:406–418. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Horne SD, Pollick SA and Heng HH: Evolutionary mechanism unifies the hallmarks of cancer. Int J Cancer. 136:2012–2021. 2015. View Article : Google Scholar

42 

Sonnenschein C and Soto AM: The aging of the 2000 and 2011 Hallmarks of Cancer reviews: A critique. J Biosci. 38:651–663. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Carnesecchi S, Bradaia A, Fischer B, Coelho D, Schöller-Guinard M, Gosse F and Raul F: Perturbation by geraniol of cell membrane permeability and signal transduction pathways in human colon cancer cells. J Pharmacol Exp Ther. 303:711–715. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Hoshi D, Nakajima A, Inoue E, Shidara K, Sato E, Kitahama M, Seto Y, Tanaka E, Urano W, Ichikawa N, et al: Incidence of serious respiratory infections in patients with rheumatoid arthritis treated with tocilizumab. Mod Rheumatol. 22:122–127. 2012. View Article : Google Scholar

45 

Chaudhary SC, Siddiqui MS, Athar M and Alam MS: Geraniol inhibits murine skin tumorigenesis by modulating COX-2 expression, Ras-ERK1/2 signaling pathway and apoptosis. J Appl Toxicol. 33:828–837. 2013. View Article : Google Scholar

46 

Vinothkumar V, Manoharan S, Sindhu G, Nirmal MR and Vetrichelvi V: Geraniol modulates cell proliferation, apoptosis, inflammation, and angiogenesis during 7,12-dimethylbenz[a] anthracene-induced hamster buccal pouch carcinogenesis. Mol Cell Biochem. 369:17–25. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Cardozo MT, de Conti A, Ong TP, Scolastici C, Purgatto E, Horst MA, Bassoli BK and Moreno FS: Chemopreventive effects of β-ionone and geraniol during rat hepatocarcinogenesis promotion: Distinct actions on cell proliferation, apoptosis, HMGCoA reductase, and RhoA. J Nutr Biochem. 22:130–135. 2011. View Article : Google Scholar

48 

Wiseman DA, Werner SR and Crowell PL: Cell cycle arrest by the isoprenoids perillyl alcohol, geraniol, and farnesol is mediated by p21Cip1 and p27Kip1 in human pancreatic adenocarcinoma cells. J Pharmacol Exp Ther. 320:1163–1170. 2007. View Article : Google Scholar

49 

Vieira A, Heidor R, Cardozo MT, Scolastici C, Purgatto E, Shiga TM, Barbisan LF, Ong TP and Moreno FS: Efficacy of geraniol but not of β-ionone or their combination for the chemoprevention of rat colon carcinogenesis. Braz J Med Biol Res. 44:538–545. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Ahmad ST, Arjumand W, Seth A, Nafees S, Rashid S, Ali N and Sultana S: Preclinical renal cancer chemopreventive efficacy of geraniol by modulation of multiple molecular pathways. Toxicology. 290:69–81. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Vinothkumar V and Manoharan S: Chemopreventive efficacy of geraniol against 7,12-dimethylbenz[a]anthracene-induced hamster buccal pouch carcinogenesis. Redox Rep. 16:91–100. 2011. View Article : Google Scholar

52 

Khan AQ, Khan R, Qamar W, Lateef A, Rehman MU, Tahir M, Ali F, Hamiza OO, Hasan SK and Sultana S: Geraniol attenuates 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced oxidative stress and inflammation in mouse skin: Possible role of p38 MAP Kinase and NF-κB. Exp Mol Pathol. 94:419–429. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Madankumar A, Jayakumar S, Gokuladhas K, Rajan B, Raghunandhakumar S, Asokkumar S and Devaki T: Geraniol modulates tongue and hepatic phase I and phase II conjugation activities and may contribute directly to the chemopreventive activity against experimental oral carcinogenesis. Eur J Pharmacol. 705:148–155. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Wittig C, Scheuer C, Parakenings J, Menger MD and Laschke MW: Geraniol suppresses angiogenesis by down-regulating vascular endothelial growth factor (VEGF)/VEGFR-2 signaling. PLoS One. 10:e01319462015. View Article : Google Scholar

55 

Gysin S, Salt M, Young A and McCormick F: Therapeutic strategies for targeting ras proteins. Genes Cancer. 2:359–372. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Wallace HM: Targeting polyamine metabolism: A viable therapeutic/preventative solution for cancer? Expert Opin Pharmacother. 8:2109–2116. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Peters GJ, Backus HH, Freemantle S, van Triest B, Codacci-Pisanelli G, van der Wilt CL, Smid K, Lunec J, Calvert AH, Marsh S, et al: Induction of thymidylate synthase as a 5-fluorouracil resistance mechanism. Biochim Biophys Acta. 1587:194–205. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Popat S, Matakidou A and Houlston RS: Thymidylate synthase expression and prognosis in colorectal cancer: A systematic review and meta-analysis. J Clin Oncol. 22:529–536. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Topolcan O and Holubec L Jr: The role of thymidine kinase in cancer diseases. Expert Opin Med Diagn. 2:129–141. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Jin X, Sun J, Miao X, Liu G and Zhong D: Inhibitory effect of geraniol in combination with gemcitabine on proliferation of BXPC-3 human pancreatic cancer cells. J Int Med Res. 41:993–1001. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Polo MP, Crespo R and de Bravo MG: Geraniol and simvastatin show a synergistic effect on a human hepatocarcinoma cell line. Cell Biochem Funct. 29:452–458. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Yu SG, Hildebrandt LA and Elson CE: Geraniol, an inhibitor of mevalonate biosynthesis, suppresses the growth of hepatomas and melanomas transplanted to rats and mice. J Nutr. 125:2763–2767. 1995.PubMed/NCBI

63 

Shoff SM, Grummer M, Yatvin MB and Elson CE: Concentration-dependent increase of murine P388 and B16 population doubling time by the acyclic monoterpene geraniol. Cancer Res. 51:37–42. 1991.PubMed/NCBI

64 

He L, Mo H, Hadisusilo S, Qureshi AA and Elson CE: Isoprenoids suppress the growth of murine B16 melanomas in vitro and in vivo. J Nutr. 127:668–674. 1997.PubMed/NCBI

65 

Mo H, Tatman D, Jung M and Elson CE: Farnesyl anthranilate suppresses the growth, in vitro and in vivo, of murine B16 melanomas. Cancer Lett. 157:145–153. 2000. View Article : Google Scholar : PubMed/NCBI

66 

McAnally JA, Jung M and Mo H: Farnesyl-O-acetylhydroquinone and geranyl-O-acetylhydroquinone suppress the proliferation of murine B16 melanoma cells, human prostate and colon adenocarcinoma cells, human lung carcinoma cells, and human leukemia cells. Cancer Lett. 202:181–192. 2003. View Article : Google Scholar : PubMed/NCBI

67 

Aqil F, Munagala R, Jeyabalan J and Vadhanam MV: Bioavailability of phytochemicals and its enhancement by drug delivery systems. Cancer Lett. 334:133–141. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Chung AS and Ferrara N: Developmental and pathological angiogenesis. Annu Rev Cell Dev Biol. 27:563–584. 2011. View Article : Google Scholar : PubMed/NCBI

69 

DeBose-Boyd RA: Feedback regulation of cholesterol synthesis: Sterol-accelerated ubiquitination and degradation of HMG CoA reductase. Cell Res. 18:609–621. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Chun JN, Lim JM, Kang Y, Kim EH, Shin YC, Kim HG, Jang D, Kwon D, Shin SY, So I, et al: A network perspective on unraveling the role of TRP channels in biology and disease. Pflugers Arch. 466:173–182. 2014. View Article : Google Scholar

71 

Barabási AL, Gulbahce N and Loscalzo J: Network medicine: A network-based approach to human disease. Nat Rev Genet. 12:56–68. 2011. View Article : Google Scholar :

72 

Furlong LI: Human diseases through the lens of network biology. Trends Genet. 29:150–159. 2013. View Article : Google Scholar

73 

Ideker T and Krogan NJ: Differential network biology. Mol Syst Biol. 8:5652012. View Article : Google Scholar : PubMed/NCBI

74 

Grossglauser M and Saner H: Data-driven healthcare: From patterns to actions. Eur J Prev Cardiol. 21(Suppl): 14–17. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Lusher SJ, McGuire R, van Schaik RC, Nicholson CD and de Vlieg J: Data-driven medicinal chemistry in the era of big data. Drug Discov Today. 19:859–868. 2014. View Article : Google Scholar

76 

Quan Y, Wang ZY, Xiong M, Xiao ZT and Zhang HY: Dissecting traditional Chinese medicines by omics and bioinformatics. Nat Prod Commun. 9:1391–1396. 2014.

77 

Quinn BJ, Kitagawa H, Memmott RM, Gills JJ and Dennis PA: Repositioning metformin for cancer prevention and treatment. Trends Endocrinol Metab. 24:469–480. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Anisimov VN: Do metformin a real anticarcinogen? A critical reappraisal of experimental data. Ann Transl Med. 2:602014.PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 48 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cho M, So I, Chun JN and Jeon J: The antitumor effects of geraniol: Modulation of cancer hallmark pathways (Review). Int J Oncol 48: 1772-1782, 2016
APA
Cho, M., So, I., Chun, J.N., & Jeon, J. (2016). The antitumor effects of geraniol: Modulation of cancer hallmark pathways (Review). International Journal of Oncology, 48, 1772-1782. https://doi.org/10.3892/ijo.2016.3427
MLA
Cho, M., So, I., Chun, J. N., Jeon, J."The antitumor effects of geraniol: Modulation of cancer hallmark pathways (Review)". International Journal of Oncology 48.5 (2016): 1772-1782.
Chicago
Cho, M., So, I., Chun, J. N., Jeon, J."The antitumor effects of geraniol: Modulation of cancer hallmark pathways (Review)". International Journal of Oncology 48, no. 5 (2016): 1772-1782. https://doi.org/10.3892/ijo.2016.3427