Abstract
In recent years, naturally occurring dietary compounds have received greater attention in the field of cancer prevention and treatment research. Among them, isoflavone genistein and curcumin are very promising anti-cancer agents because of their non-toxic and potent anti-cancer properties. However, it is important to note that the low water solubility, poor in vivo bioavailability and unacceptable pharmacokinetic profile of these natural compounds limit their efficacy as anti-cancer agents for solid tumors. Therefore, the development of synthetic analogs of isoflavone and curcumin based on the structure-activity assay, and the encapsulation of isoflavone and curcumin with liposome or nanoparticle for enhancing the anti-tumor activity of these natural agents, is an exciting area of research. Emerging in vitro and in vivo studies clearly suggest that these analogs and formulations of natural compounds could be much more potent for the prevention and/or treatment of various cancers. In this review article, we will summarize the current knowledge regarding the anti-cancer effect of natural compounds and their analogs, the regulation of cell signaling by these agents, and the structure-activity relationship for better design of novel anti-cancer agents, which could open newer avenues for the prevention of tumor progression and/or treatment of human malignancies.
Keywords: Isoflavone, curcumin, chemoprevention, cancer therapy
INTRODUCTION
It is well known that most human cancers are induced by environmental factors including chemical, radioactive and biological factors that exist in our environment. There are significant differences in the cancer incidence, mortality, and survival among ethnic groups, who have different lifestyles and have been exposed to different environmental factors [1,2]. Studies have also shown that 30 to 40 percent of cancers are directly linked to dietary choices [3] and these cases of cancer are preventable by appropriate diets. This means that appropriate diets may prevent 3 to 4 million cases of cancer every year globally. The consumption of fruits, soybean and vegetables has been associated with reduced risk of several types of cancers [4,5]. Diets containing substantial and varied amounts of vegetables and fruits may prevent 20% or more of all cases of cancer [3]. The in vitro and in vivo experimental studies have demonstrated that some dietary components including isoflavones, curcumin, indole-3-carbinol (I3C), 3,3’-diindolyl-methane (DIM), (−)-epigallocatechin-3-gallate (EGCG), etc, have inhibitory effects on human cancers [6–9], suggesting that they may serve as potent non-toxic agents for the prevention of tumor progression and/or could serve as therapeutic agents alone or in combination with conventional therapeutics.
In recent years, the dietary compounds and their synthetic analogs have received greater attention in the field of cancer research. Among them, isoflavone genistein, curcumin, and their analogs are promising anti-cancer agents because of their non-toxic and potent anti-cancer properties, which is the focus of this article although many other dietary agents are equally important. Soy isoflavones are mainly derived from soybean showing inhibitory effects on various cancers including breast and prostate cancers. Interestingly, the lower risks of breast and prostate cancers in Asians, who consume 20–50 times more soy than Americans, suggest that the components in the soybean may act as natural chemopreventive agents [10–13]. Soy isoflavones including genistein have been demonstrated to be responsible for reducing the incidence of hormone-related cancers. In laboratory in vitro various cancer cells [14,15]. Moreover, the evidences from in vivo studies have demonstrated that isoflavone genistein exerts its inhibitory effects on the development and progression of cancers [16]. Furthermore, we have shown that isoflavone genistein inhibits cancer cell invasion, metastasis, and angiogenesis through the regulation of genes critically involved in the control of cell proliferation, apoptotic cell death, migration, invasion, angiogenesis and metastasis [17,18], suggesting that isoflavone genistein could a promising agent for cancer prevention and/or treatment of human malignancies.
Curcumin is a polyphenolic compound derived from dietary spice turmeric and possesses diverse pharmacological effects including antioxidant, anti-inflammatory, anti-proliferative, and anti-angiogenic activities. Curcumin has been used for centuries in Asia, both in traditional medicine and in cooking where curcumin gives natural yellow color to the food. The first article regarding the use of curcumin to treat human biliary diseases was published in The Lancet in 1937 [19]. After several decades of research since then, it has been well known that curcumin possesses potent antiinflammatory activity because of its inhibitory effects on cyclooxygenases 1, 2 (COX-1, COX-2), lipoxygenase (LOX), TNF-α, interferon γ (IFN-γ), inducible nitric oxide synthase (iNOS), and NF-κB [20,21]. Importantly, experimental evidences suggest that curcumin could exert its inhibitory effects on cancer development and progression. The mechanisms implicated in the inhibition of tumorigenesis by curcumin are unclear but could involve a combination of anti-oxidant, anti-proliferation, pro-apoptotic, and anti-angiogenic properties through the regulation of genes and molecules that are involved in multiple signaling pathways. Moreover, preclinical animal experiments and phase I clinical trials have demonstrated minimal toxicity of curcumin even at relatively high doses (12 g/day) [22]. However, curcumin exhibits poor bioavailability because of poor absorption and rapid metabolism [22]. To improve the bioavailability of curcumin, liposomal curcumin, nanoparticle curcumin, and structural analogs of curcumin have been synthesized and investigated to determine the absorption and anti-cancer activity [23,24]. The results are promising, which further suggest that curcumin or its novel structural analogs could serve as potent agents for the prevention and/or treatment of human malignancies, and thus requires more phase II and III clinical trials.
ISOFLAVONE GENISTEIN AND ITS ANALOG
Molecular Structure and Biological Properties of Isoflavone Genistein
Isoflavones are a subclass of a family of ubiquitous flavonoids, and have been found in relatively high concentration in soybeans and most soy-protein products. The basic structure of isoflavone is the flavone nucleus, which is composed of 2 benzene rings linked through a heterocyclic pyrane ring (Fig. (1)). In contrast to flavonoids, isoflavones possess a 3-phenylchroman skeleton that is biogenetically derived from the 2-phenylchroman skeleton of the flavonoids. The structure of isoflavone genistein is very similar to that of estrogen (Fig. (1)), thus isoflavones have also been known as phytoestrogens. Because of the structural similarity to estrogen, isoflavone genistein have been shown to bind to estrogen receptors (ER). By interaction with ER, genistein blocks the binding of more potent estrogens, thereby exerting a potential role in the prevention of hormone related cancer. However, it is important to note that genistein could either induce cell proliferation by estrogenic agonistic properties (at concentrations ≤1 µM) or prevent hormone-dependent growth of cancer cells by potential estrogen-antagonistic activity (at concentrations ≥5 µM) dependent on its concentrations [25]. Moreover, we and others have found that genistein could also inhibit cell growth and induce apoptosis in ER negative breast cancer cells [14,26], suggesting that genistein may exert its effects through ER dependent or independent mechanisms.
Fig. 1.
Molecular structures of isoflavone, estrogen, and representative isoflavone analogs.
It has been well known that isoflavone genistein has many important biological effects that are beneficial for human health, especially the anti-cancer effect of isoflavone genistein. In addition to its function as an estrogen agonist or antagonist, genistein is a known inhibitor of protein-tyrosine kinase [27]. The inhibitory activity of genistein on protein-tyrosine kinase could contribute to anti-proliferative or pro-apoptotic effects of genistein. Moreover, it has been reported that soy isoflavones have antioxidant effect against TNF-α induced NF-κB activation in humans both in vitro and in vivo [15,28,29], which could be responsible for the inhibition of cancer cell growth. In animal studies, soy isoflavones have been found to suppress spontaneously developed and chemical induced prostate and breast cancers [30,31], suggesting their inhibitory effects on tumorigenesis. Growing body of evidences have shown that isoflavone genistein can inhibit cancer cell growth and induce apoptotic cell death in various cancer cells through regulation of cell signaling transduction pathways [32,33]. Moreover, genistein could potentiate anti-cancer activity of chemotherapeutic agents, and also inhibit angiogenesis and cancer metastasis [17,18,34], suggesting that isoflavone genistein could be a potent agent for the treatment of cancers in combination with conventional chemotherapeutics, which of course must be tested through innovative clinical trials in order to fully appreciate the value of isoflavones in human health and diseases.
The Effects of Isoflavone Genistein on Cellular Signaling
It has been known that isoflavone genistein exerts its effects on multiple signaling pathways (Fig. (2)). We have found that genistein significantly inhibited the NF-κB DNA-binding activity in vitro in prostate, breast, lung, and pancreatic cancer cells [35–37]. Other investigators have reported similar results showing that isoflavone genistein inhibited the activation of NF-κB in human lung epithelial cells and myeloid cells [38,39]. Moreover, we have investigated the in vivo effects of isoflavone genistein on NF-κB signaling. We found that when human volunteers received 50 mg of soy isoflavone supplements Novasoy™ (containing genistein, daidzein, and glycitein at a 1.3:1:0.3 ratio) twice daily for three weeks, TNF-α treatment ex-vivo failed to activate NF-κB activity in lymphocytes harvested from these volunteers, while lymphocytes from these volunteers collected prior to soy isoflavone intervention showed activation of NF-κB DNA binding activity upon TNF-α treatment ex-vivo [40]. These results demonstrate that soy isoflavone supplementation has a protective effect against TNF-α induced NF-κB activation in humans, suggesting that soy isoflavone could exert its cancer chemopreventive activity through the regulation of NF-κB signaling. More importantly, we also found that isoflavone genistein could enhance the anti-tumor activity of chemotherapeutic agents via the down-regulation of NF-κB signaling [36], suggesting the therapeutic value of isoflavone in cancer treatment.
Fig. 2.
Cellular signaling altered by isoflavone.
In addition to the role of NF-κB as a target of genistein, the activation of NF-κB is also known to be mediated by the activation of Akt. Akt signaling is another target of isoflavone genistein (Fig. (2)). We found that isoflavone genistein treatment significantly decreased the phosphorylation of Akt protein at Ser473 and inhibited the Akt kinase activity [15], which could be responsible for the inhibition of NF-κB. Genistein pre-treatment also abrogated the activation of Akt by EGF. To further explore the inhibitory mechanism(s) of genistein on Akt and NF-κB pathways, Akt expression construct was transiently co-transfected with NF-κB-Luc reporter construct into PC-3 prostate cancer cells. Luciferase assay showed an increased luciferase activity in PC-3 cells co-transfected with the constructs. However, genistein treatment decreased the luciferase activity in the co-transfected PC-3 cells. These results were further confirmed by examining the NF-κB DNA-binding activity in co-transfected cells using EMSA, and the results suggests that isoflavone genistein inhibits NF-κB signaling through Akt pathway. We also observed similar results in MDA-MB-231 breast cancer cells [37]. Therefore, the down-regulation of NF-κB and Akt signaling pathways by isoflavone genistein could contribute to the inhibition of cancer cell growth and the induction of apoptosis.
It has been known that Wnt signaling interacts with Akt signaling, promoting cancer cell proliferation. We have found that isoflavone up-regulated the expression of GSK-3β, enhanced GSK-3β binding to β-catenin, and increased the phosphorylation of β-catenin, suggesting that isoflavone could inactivate Wnt signaling, leading to the inhibition of prostate cancer cell growth [41]. The results from other laboratories also showed that isoflavone genistein decreased basal and Wnt-1-induced cell proliferation and attenuated the expression of Wnt-1 target c-Myc and Cyclin D1 [42] and that isoflavone also inhibited the expression of Wnt-5a [43]. All these results suggest that isoflavone genistein has the promising inhibitory effects on Wnt signaling, which could be useful for targeted treatment of certain human malignancies.
We have also investigated the effects of isoflavone genistein on Notch signaling. We found that isoflavone genistein inhibited Notch signaling, leading to the down-regulation of NF-κB activity, resulting in the inhibition of cell proliferation, and the induction of apoptosis in pancreatic cancer cells [44,45]. Other investigators also reported that genistein could inhibit the expression of Notch-2 [43], which is consistent with our findings. Therefore, isoflavone genistein could inhibit cancer cell growth and induce apoptosis through the inhibition of Notch signaling as well (Fig. (2)), implicating that isoflavone is a multi-targeted agents.
Isoflavone genistein could also target AR signaling (Fig. (2)). We have previously found that genistein transcriptionally down-regulated AR, decreased nuclear AR binding to androgen responsive element (ARE) and, thereby, inhibited the transcription and protein expression of PSA in androgen-sensitive LNCaP cells [46,47]. In an in vivo animal study, dietary genistein has also been found to down-regulate the expression of AR [48]. Furthermore, we have found that isoflavone-induced inhibition of cell proliferation and induction of apoptosis are partly mediated through the regulation of the Akt/FOXO3a/GSK-3β/AR signaling network [49]. These results suggest that the down-regulation of AR expression could be an important strategy for the prevention and/or treatment of prostate cancer, especially hormone refractory prostate cancer for which there is no curative treatment.
The above mentioned findings along with those that we were unable to cite in this brief review article clearly suggest that isoflavone genistein exerts pleiotropic effects on cell signal transduction pathways in cancer cells; however, pure genistein by itself may not be an attractive agent for the treatment of human cancers as documented earlier by our laboratory [32,33,50]. In order to further enhance the activity of isoflavone, synthetic analogs of isoflavone with robust biological activity could be very attractive for the treatment of human cancers, and thus it is further discussed below.
Novel Synthetic Analogs of Isoflavone as Anti-Cancer Agents
It is well accepted that the molecular structure of an agent is tightly related with its biological activity. The extensive changes in biological activity have been observed after the modifications of molecular structure. The structure and function analysis showed that the anti-tumor properties of isoflavonoids are in part due to some structural motifs that include a benzopyran motif with a double bond between C2-C3 positions and a side chain containing a phenyl ring having metal chelating ability [51]. These structures can be easily built into a compound 3-formylchromone by condensing it with various amines in alcoholic medium yielding corresponding Schiff bases. Moreover, these compounds could form metal conjugates with therapeutically important metal ions such as copper to become more effective.
We have designed and synthesized copper conjugated isoflavone derivatives [50] (Fig. (1)). We tested the effects of synthetic derivatives of isoflavone on the growth of BT-20 breast, PC-3 prostate, Colo357 and BxPC-3 pancreatic cancer cells. We found that the synthetic derivatives of isoflavone inhibited cell proliferation in all cancer cell lines tested at much lower IC50 value (10 µM) compared to genistein, suggesting that these derivatives of isoflavone are more potent in inhibiting the growth of cancer cells. Moreover, we observed that synthetic derivatives of isoflavone at lower doses could induce more apoptosis compared to parent genistein. Furthermore, we found that synthetic derivatives of isoflavone significantly inhibited the activation of Akt and NF-κB. These results are similar to those observed by genistein treatment, suggesting that the growth inhibitory and apoptosis inducing effects of the synthetic derivatives of isoflavone are partly mediated by the inactivation of Akt and NF-κB signaling pathways [50].
Studies from other laboratory have shown that Phenoxodiol, a synthetic analog of the plant isoflavone genistein resulted in improved broad-spectrum anticancer efficacy. The most important property of phenoxodiol is its ability to sensitize resistant tumor cells to chemotherapy. Therefore, it has been used for early- and late-stage prostate cancer including hormone-refractory prostate cancer, early stage cervical and vaginal cancer, chemo-resistant ovarian cancer, and renal cancer [52–54]. It has also been reported that phenoxodiol inhibited DMBA-induced mammary carcinogenesis in Sprague-Dawley rats [55]. Importantly, phenoxodiol could sensitize cancer cells to the anti-tumor effects of standard chemotherapeutics [53,56,57]. Moreover, it has been found that phenoxodiol suppresses the function of tNOX, leading to the induction of apoptosis through the inhibition of anti-apoptotic proteins XIAP and FLICE [53]. In animal studies, phenoxodiol has been found to stimulate both NK and tumor-specific cell lytic activity, causing significantly reduced tumor growth rates and prolonged survival [58]. In clinical trials, phenoxodiol has shown some beneficial effects on disease stabilization without severe toxicity [59]; however further in-depth research in this area is certainly needed.
Phytoestrogen biochanin A is another isoflavone with anti-carcinogenic properties and has been found to inhibit N-nitroso-N-methylurea-induced rat mammary carcinogenesis [60]. However, recent report showed that genistein was found to be more effective than biochanin A in providing protection against oxidative stress [61]. In order to further investigate the relationship between structure and anti-cancer activities, Vasselin et al. have reported a series of synthetic fluoro- and amino-substituted isoflavones as potential anti-tumor agents based on structural similarities to known isoflavones [62]. These isoflavone analogs have shown significant inhibitory effects on cancer cell proliferation at very low IC50 (<1µM) in breast cancer cells, suggesting their potent anti-tumor activity. Certain oxime- and methyloxime-containing isoflavone analogs were also synthesized and evaluated for their antiproliferative activity against cervical, hepatocellular, and oral epithelial cell carcinoma. These analogs of isoflavone arrested cancer cells at G2/M phase and showed strong anti-proliferative activities at GI50 values of less than 1 µM [63]. Whatmore, et al. also reported that several synthesized isoflavone analogs, SU1433, SU 5416 and SU6668, were potent inhibitors of VEGF-induced angiogenesis compared to naturally occurring isoflavones, suggesting the potent anti-tumor and anti-angiogenic effects of these compounds [64]. In order to enhance the intrinsic activity of genistein, its glycosidic derivatives were also synthesized and found to be more potent in their cytostatic and cytotoxic effect than genistein [65]. These reports all suggest that synthetic isoflavone analogs could have more potent activity than naturally occurring isoflavone and could be useful for combination therapy with conventional chemotherapeutics in order to achieve better treatment outcome, and thus such studies must be done in human patients not only for assessing its value as therapeutics but also for assessing pharmacokinetic, pharmacodynamic, and toxicity profile.
Clinical Trials Using Isoflavone for Cancer Treatment
Because isoflavone genistein transcriptionally down-regulates AR and its target PSA, an important biomarker for the diagnosis of prostate cancer and the prediction of prostate cancer progression, we have conducted a phase II clinical trial to investigate the modulation in serum PSA levels in patients diagnosed with prostate cancer by soy isoflavone supplementation [66]. This pilot clinical data demonstrated that soy isoflavone supplementation could decrease the rate of rise in serum PSA levels without any toxicity in prostate cancer patients [66]. Another phase II clinical trial has been conducted to evaluate the efficacy of isoflavone in patients with PSA recurrent prostate cancer after prior therapy [67]. It was found that dietary intervention with isoflavone supplementation decreased the slope of rising PSA, providing beneficial evidence in support of the use of isoflavone supplements against prostate cancer [67]. In another clinical trial focusing on determining the biological effects of soy protein isolate (SPI) consumption on circulating hormone profiles and AR expression patterns in men at high risk for developing advanced prostate cancer, the authors have found that consumption of SPI significantly suppressed AR expression but did not alter ER-β expression or circulating hormones, suggesting that isoflavone could be beneficial in preventing prostate cancer by inhibition of AR and PSA expression [68]. Several other clinical trials have also shown similar results documenting reduced PSA after receiving isoflavone supplements [69,70]. Based on the molecular evidence showing that isoflavone targets multiple cellular signaling pathways, more and more clinical trials are being conducted to investigate the value of isoflavone in human cancer (www.ClinicalTrials.gov). These clinical trials are focused on investigating the effects of isoflavone on the prevention of various cancer developments, and the combination treatment of various cancers with chemotherapeutic agents or other “natural products” are emerging. We believe that the conventional cancer therapeutics combined with supplement of isoflavone and its analog could be an important novel strategy for the treatment of human cancers and/or the prevention of cancer progression. Further rationally-designed clinical trials are needed in order to fully appreciate the health benefit of isoflavone and its analogs in humans.
CURCUMIN AND ITS ANALOGS
Molecular Structure and Biological Properties of Curcumin
Curcumin is a naturally occurring compound present in turmeric. It is the main curcuminoid from turmeric which is a member of the ginger family. The curcuminoid is responsible for the yellow color of turmeric. Curcumin exists in at least two tautomeric forms, keto and enol. The enol form is more energetically stable in the solid phase and in solution. The structure of curcumin was first identified in 1910. Curcumin is a homodimer of feruloylmethane and consists of several functional groups (Fig. (3)). It contains a methoxy group and a hydroxyl group, a heptadiene with two Michael acceptors, and an α,β-diketone.
Fig. 3.
Molecular structures of curcumin and its representative analogs.
Curcumin has been known to possess anti-inflammatory, antioxidant, and anti-cancer activities. Therefore, it has been used for the treatment of inflammatory diseases, diabetes, depression, arthritis, neurological diseases, Crohn's disease, and the other disorders of the cardiovascular, pulmonary, and neurological systems [22,71–74]. Accumulating experimental evidence suggests that curcumin interferes with a variety of molecules which are involved in cancer development and progression, leading to the inhibition of cancer cell growth [20]. Importantly, curcumin could be useful as a chemo-sensitizer in cancer chemotherapy [75], which may result in better treatment outcome in patients although such clinical trials have not be done yet which indeed could be due to poor systemic and target tissue bioavailability of curcumin.
The Effects of Curcumin on Cellular Signaling
Curcumin has been used for centuries as a therapeutic agent for the treatment of inflammatory diseases in Asian medicine. Since NF-κB is significantly activated in the processes of inflammation, it has been well known that curcumin is a strong inhibitor of NF-κB (Fig. (4)). Curcumin could inhibit IKK, suppress both constitutive and inducible NF-κB activation, regulate Bax-mediated apoptosis, and potentiate TNF-induced apoptosis [76,77]. Curcumin also sensitizes human colorectal cancer xenografts in nude mice to γ-radiation by targeting NF-κB-regulated gene products [78]. It has also been reported that treatment with a liposomal formulation of curcumin resulted in a dose-dependent growth suppression of cancer cells and a decreased activation of NF-κB [79]. Moreover, expression of NF-κB target genes including cyclin D1, cyclooxygenase-2 (COX-2), matrix metalloproteinase-9 (MMP-9), Bcl-2, Bcl-xL, Mcl-1L, and Mcl-1S were reduced [78], indicating the effect of curcumin on NF-κB signaling pathway. Furthermore, clinical trial showed that curcumin down-regulated NF-κB and COX-2 in peripheral blood mononuclear cells from patients with pancreatic cancer [80] although the anti-cancer activity of curcumin in this trial was found to be minimal. However, it is clear that curcumin could inhibit NF-κB signaling in vitro and in vivo but the clinical benefit of curcumin has not yet been proven.
Fig. 4.
Cellular signaling altered by curcumin.
In addition to NF-κB signaling, Akt signaling could also be inhibited by curcumin (Fig. (4)). It has been shown that curcumin inhibited the phosphorylation of Akt, mTOR, and their downstream targets in prostate cancer cells [81]. Curcumin has also been found to inhibit the proliferation of cisplatin-resistant ovarian cancer cells through the inhibition of Akt activation [82]. The analog of curcumin, 4-hydroxy-3-methoxybenzoic acid methyl ester (HM-BME), also inhibited the proliferation of cancer cells and induced apoptosis through the down-regulation of phosphorylated Akt, the inhibition of Akt kinase activity, and the reduction of NF-κB DNA-binding activity [83]. These findings together with other reports [84,85] all suggest that curcumin could inhibit Akt signaling and facilitate inhibition of proliferation and induction of apoptosis in various cancers.
It has been also shown that curcumin could exert its inhibitory effects on Wnt signaling (Fig. (4)). Experimental studies have shown that curcumin suppressed β-catenin mediated transcription activated by Wnt3a, and inhibited the growth of colon cancer cells [86]. Curcumin also induced caspase-3-mediated degradation of β-catenin [87], resulting in the decreased binding of β-catenin to TCF, and thereby causing inactivation of Wnt signaling. In addition, curcumin has shown to down-regulate p300 which is a positive regulator of the Wnt/β-catenin pathway [87], further suggesting its inhibitory effects on Wnt signaling. Moreover, gene expression profile analysis showed that the expression of Frizzled-1 (Wnt receptor) was most strongly attenuated by curcumin [88]; therefore it is quiet clear that curcumin could inhibit proliferation of cancer cells through the inhibition of Wnt signaling.
Curcumin also showed its inhibitory effects in both androgen-dependent and independent prostate cancers. Curcumin has been found to down-regulate the transactivation and expression of AR and AR-related molecules (AP-1 and NF-κB), and inhibited colony formation in prostate cancer cells [89]. Moreover, some curcumin analogs have been shown to possess potent anti-androgenic activities [90], and were found to be superior than hydroxyflutamide which is the currently available anti-androgenic agent for the treatment of prostate cancer; however, clinical studies using curcumin analogs has not been done yet. Structure-activity relationship studies demonstrated that some moieties seem to be important factors related to the anti-androgenic activity [90]. Therefore, these analogs could serve as newer anti-androgenic agents for controlling the growth of AR-mediated prostate cancers.
Curcumin has been used for the prevention and treatment of a number of inflammatory diseases because of its anti-inflammatory activity. The role of LOX and COX isoforms, particularly COX-2, in the inflammation has been well established. Therefore, the effects of curcumin on COX and LOX were investigated. Experimental evidence has shown that curcumin could regulate LOX and COX-2 predominately at the transcriptional level, and to a certain extent, at the posttranslational level [91]. Thus, the dual COX/LOX inhibitory potential of curcumin provides distinctive advantages over synthetic COX-2-specific inhibitors. Because the up-regulation of COX-2 is commonly observed in various cancers, the inhibition of COX-2 expression by curcumin could contribute in suppressing the growth of cancer cells; however clinical trials have not been completed to-date.
Recent Advances in Novel Synthetic Analogs of Curcumin and their Biological Activity
Despite the biological activity of curcumin in vitro and in selected models in vivo, curcumin has shown poor systemic and tissue bioavailability [22,92]. Preclinical and clinical studies have shown that the concentrations of curcumin achieved in plasma and target tissues are very low. The plasma and colorectal tissue concentrations of curcumin in patients receiving 3,600 mg curcumin orally were 11.1 nM [93] and 12.7 nM [94], respectively. Moreover, the level of curcumin concentration in the liver was found to be below the limit of detection [94]. These data suggests that novel synthetic analog of curcumin must be developed or the bioavailability of curcumin must be improved by novel methods such nano-formulation or other methods so that the value of curcumin or its synthetic analog could be appreciated toward launching novel clinical trials.
Recently, we have designed and synthesized new difluoro Knoevenagel condensates of curcumin and Schiff bases along with their copper (II) complexes (Fig. (3)), and evaluated their biological activities with respect to the suppression of 26S proteasome, the inhibition of proliferation, and the induction of apoptosis in colon and pancreatic cancer cells [95]. We found that all copper complexes possessed distorted square planar geometries with 1:1 metal to ligand stoichiometry with reversible copper redox couple. The difluoro compound CDF inhibited the activities of rabbit 20S proteasome and cellular 26S proteasome, leading to the inhibition of cell proliferation and the induction of apoptosis, which was consistent with our previous findings [96]. These results suggest that our newly synthesized classes of curcumin analogs especially CDF could be useful as chemopreventive and/or therapeutic agents against cancers. These preliminary results were encouraging, and thus further studies were conducted and are also being done to test whether curcumin analog could show superior bioavailability compared to curcumin and, in turn, could show better anti-tumor activity. Indeed, animal experiments and pharmacokinetic analysis revealed that CDF had better retention and bioavailability, and that the concentration of CDF in the pancreas tissue of mice was 10-fold higher compared to curcumin [97], suggesting better tissue bioavailability of CDF. We have also tested the effects of CDF on NF-κB and PGE2, an enzymatic product of COX-2, and we found that CDF significantly down-regulated the NF-κB DNA binding activity and decreased PGE2 expression level [97]. The molecular docking studies have shown that the fluorocurcumin analogs did not introduce any major steric changes compared to the parent curcumin molecule, which was consistent with the down-regulation of NF-κB and the reduced level of PGE2 in cells treated with CDF. These results demonstrate that CDF could inhibit cancer cell growth and induce apoptotic cell death through the inactivation of proteasome, NF-κB and COX-2 signaling. More importantly, our results have shown that the bioavailability of CDF is much superior compared to curcumin, suggesting that CDF could be clinically useful as an anti-cancer agent, which awaits further investigations.
Another synthetic curcumin analog GO-Y030 has been tested for its inhibitory effects on human breast and pancreatic cancer cells [98]. It has been found that both curcumin and GO-Y030 reduced cell viability and induced apoptosis, but GO-Y030 was substantially more potent compared to curcumin. Further investigation revealed that GO-Y030 inhibited STAT3 phosphorylation and transcriptional activity whereas comparable dosages of curcumin had little or no effect on STAT3. These results suggest that GO-Y030 inhibits cell growth through the down-regulation of STAT3 activation and that GO-Y030 could be potentially useful as a therapeutic agent for the treatment of cancers expressing high levels of activated STAT3 [98]. Other curcumin analogs, FLLL11 and FLLL12, also inhibited phosphorylation of STAT3 in breast and prostate cancer cells [99]. In addition, FLLL11 and FLLL12 exhibited more potent activities than curcumin on the down-regulation of STAT3, Akt, and HER-2/neu, as well as the inhibition of cancer cell growth and migration [99]. GO-Y030, FLLL11, and FLLL12 also induced apoptosis through the increased cleavage of PARP and caspase-3 in pancreatic and colorectal cancer cells, suggesting their potential use as chemopreventive or therapeutic agents for various cancers [100,101]. However, further preclinical animal experiments, pharmacokinetics, and clinical studies are needed in order to appreciate whether these analogs could have any value for the treatment of human malignancies.
Although curcumin has long been used for the treatment of many diseases, it remains unknown whether the activity of curcumin is based on its scaffold or whether it results from the Michael acceptor properties of the α,β-unsaturated diketone moiety central to its structure. By modifying diketone system of curcumin, two curcumin analogs (benzyloxime and isoxazole) were synthesized and tested. Results showed that the analogs remarkably increased anti-tumor potency in MCF-7 and MDR transfected MCF-7 breast cancer cells. Furthermore, these curcumin analogs potently reduced the expression levels of Bcl-2, Bcl-XL, and COX-2 in MCF-7 and MDR transfected MCF-7 breast cancer cells. These results suggest that these analogs could be effective anticancer agents for the treatment of hormone-independent MDR breast cancer [102]. In another report, electron-rich pyrazole and isoxazole analogs were synthesized and evaluated against two breast cancer cell lines [103]. All these analogs showed much lower IC50 values in sub-micromolar range, and showed ten to fifty times more potent than curcumin, suggesting that they could serve as potential antitumor agents toward cancer therapy [103,104] although in vivo studies and pharmacokinetics are yet to be done using these analogs.
In addition to the above mentioned studies, several new curcumin analogs were also studied regarding their structure-activity relationships and the mechanism of action. It has been reported that analogs with furan moiety have excellent inhibitory effect on thioredoxin reductase (TrxR) in an irreversible manner, suggesting that the furan moiety could serve as a possible pharmacophore during the interaction of curcumin analogs with TrxR [105]. A synthetic monoketone compound, EF24, was developed from curcumin and showed potent anticancer activity through the down-regulation of NF-κB signaling [106]. EF24 induced apoptotic cell death of lung, breast, ovarian, and cervical cancer cells with ten times higher potency than that of curcumin has been reported. EF24 inhibited the phosphorylation and degradation of IκB and blocked the nuclear translocation of NF-κB, leading to the inhibition of NF-κB activation [106]. EF24 also induced cell cycle arrest and apoptosis by means of a redox-dependent mechanism in MDA-MB-231 human breast cancer cells and DU-145 human prostate cancer cells [107]; however, it is really surprising why these analogs has not been tested for their systemic and tissue bioavailability yet. Enone analogs of curcumin with a 3-carbon, 5-carbon, or 7-carbon spacer were compared with curcumin for their abilities to inhibit the TNF-α-induced activation of NF-κB and it was found that enone analogs with the 5-carbon spacer were especially active for the inhibition of NF-κB [108]. Four novel dienone cyclopropoxy curcumin analogs were further synthesized and tested in mice bearing Ehrlich ascites tumor (EAT) in vivo [109]. It has been found that the analogs increased the life span of mice bearing EAT with significant reduction in the microvessel density in the peritoneum walls of mice with concomitant reduction in the ascites volume in this animal model [109].
Moreover, curcumin analogs, symmetrical 1,5-diarylpenta-dienone whose aromatic rings possess an alkoxy substitution at each of the positions 3 and 5, were designed and synthesized. It has been found that these analogs down-regulated β-catenin, Ki-ras, cyclin D1, c-Myc, and ErbB-2 at as low as one eighth the concentration at which curcumin normally showed its effects [110]. Furthermore, in a study regarding structure and activity, new curcumin analogs were classified into four series: monophenyl analogs, heterocycle-containing analogs, analogs bearing various substituent on the phenyl rings, and analogs with various linkers. These new analogs were tested for cytotoxicity against androgen-dependent LNCaP and androgen-independent PC-3 cells. It was found that ten analogs possessed potent cytotoxicity against both LNCaP and PC-3 cells, seven only against LNCaP, and one solely against PC-3, demonstrating their structure-activity relationship [111]. A number of curcumin analogs were further synthesized and evaluated as potential AR antagonists against PC-3 and DU-145 prostate cancer cells in the presence of AR and AR co-activator, ARA70. Structure-activity relationship studies indicated that the bis(3,4-dimethoxyphenyl) moieties, the conjugated β-diketone moiety, and the intramolecular symmetry of the molecules was important factors related to anti-androgenic activity. Further analysis demonstrated that the coplanarity of the β-diketone moiety and the presence of a strong hydrogen bond donor group were also crucial for the anti-androgenic activity [90]. In addition, it has been found that ASC-J9 and its analogs have inhibitory effects on prostate cancer cell proliferation through a novel mechanism of enhancing AR degradation [112]. These together with other information will likely guide further design of new curcumin analogs with better anti-prostate cancer activity in the near future.
Other synthetic curcumin analogs or copper complexes also showed their effects on COX signaling, angiogenesis, cell proliferation, and apoptotic cell death.
In order to find more selective COX-1 inhibitors, a series of novel curcumin analogs were synthesized and evaluated for their ability to inhibit COX-1 by measuring COX-1, COX-2, and PGE2. It was found that the most potent curcumin analogs were (1E,6E)-1,7-di-(2,3,4-trimethoxyphenyl)-1,6-heptadien-3,5-dione and (1E,6E)-methyl 4-[7-(4-methoxycarbonyl)phenyl]-3,5-dioxo-1,6-heptadienyl]benzoate [113]. Another synthetic curcumin analog, dimethoxycurcumin was compared with curcumin for the ability to inhibit cell proliferation and induction of apoptosis in human HCT116 colon cancer cells, and the results showed much more potent activity than curcumin in inhibiting cell proliferation and inducing apoptosis [114]. Curcumin analogs also showed their anti-angiogenic activity in vivo. Intraperitoneal administration of the analogs tetrahydrocurcumin (THC), salicyl curcumin (SC) and curcuminIII (C-III) reduced the number of tumor directed capillaries induced by injecting B16F-10 melanoma cells on the ventral side of C57BL/6 mice [115]. Curcumin analogs also down-regulated the expression of angiogenesis-associated genes, VEGF and MMP-9 [116]. Four synthetic curcuminoids, 1,7-bis(4-hydroxy-3-methoxyphenyl)-1, 6-heptadiene-3, 5-dione (curcumin1), 1,7-bis(piperonyl)-1,6-heptadiene-3, 5-dione (piperonyl curcumin), 1, 7-bis(2-hydroxy naphthyl)-1, 6-heptadiene-2, 5-dione (2-hydroxy naphthyl curcumin), 1,1-bis(phenyl)-1, 3, 8, 10- undecatetraene-5, 7-dione (cinnamyl curcumin) and their copper(II) complexes were investigated for their in vivo anti-tumor activities. Copper chelates of synthetic curcuminoids showed enhanced anti-tumor activity with a significant reduction of solid tumor volume in mice [117]. Copper complexes of curcuminoids with a hydroxy group on the ring such as 2-hydroxy naphthyl curcumin were found to be most active [117], suggesting the importance of structure-activity relationship.
In order to further increasing the curcumin absorption by oral administration, several studies have focused on the preparation of liposome-encapsulated curcumin, and tested the biochemical properties of liposome-encapsulated curcumin. In an animal experiment using Sprague-Dawley (SD) rats, pharmacokinetic parameters showed that liposome-encapsulated curcumin possessed high bioavailability of curcumin, demonstrating a faster rate and better absorption of curcumin after administration of liposome-encapsulated curcumin [118]. Oral liposome-encapsulated curcumin gave higher Cmax and shorter Tmax values, indicating the enhanced gastrointestinal absorption by liposome encapsulation. Moreover, the plasma antioxidant activity after oral ministration of liposome-encapsulated curcumin was significantly higher than that of controls and the plasma curcumin concentration was significantly correlated with plasma antioxidant activities [118]. The minimum effective dose and the optimal dosing schedule of liposomal curcumin in a xenograft mouse model of human pancreatic cancer were investigated [119]. It was found that 20 mg/kg dose three times per week had the greatest decrease (52%) in tumor growth [119]. The effects of liposome-encapsulated curcumin were also examined in different cancer system. The liposomal curcumin has been found to dose-dependently inhibit growth of head and neck squamous cell carcinoma through the down-regulation of NF-κB and its targets, cyclin D1, COX-2, MMP-9, Bcl-2, Bcl-xL, Mcl-1L [24]. In vivo, liposomal curcumin also suppressed pancreatic cancer growth in murine xenograft models and inhibited tumor angiogenesis with down-regulation of NF-κB signaling [120]. Moreover, combination of liposomal forms of curcumin and resveratrol significantly inhibited the growth of prostate cancer in vivo [121]. In vitro studies showed that curcumin plus resveratrol significantly inhibited cell proliferation and induced apoptosis with down-regulation of p-Akt, cyclin D1, mTOR, and AR [121]. All these results demonstrate that liposomal curcumin could be more potent for the treatment of cancer because of its improved bioavailability although such studies have not been done for assessing the target tissue bioavailability or toxicity, which must be done prior to any clinical investigation.
Other than liposomal formulation, nanoparticulate formulation of curcumin is another approach for improving the bioavailability of curcumin for enhancing treatment efficacy. A polymer-based nanoparticle approach to improve bioavailability of curcumin has been designed and tested where the authors have found that nanoparticle curcumin exhibited very rapid and more efficient cellular uptake than curcumin [23]. Nanoparticle curcumin was more potent than curcumin in inducing apoptosis and suppressing proliferation of various cancer cells which was concomitant with down-regulation of NF-κB, cyclin D1, MMP-9, and VEGF [23]. Another recent report also showed that nanoparticle encapsulation improved oral bioavailability of curcumin by at least 9 fold when compared to curcumin [122]. Several other forms of nanoparticle curcumin including lipid-based and polymer-based nanoparticle-encapsulated curcumin also showed enhanced bioavailability and improved anticancer activities [123–125], suggesting that nanoparticle curcumin could serve as potent agents for the treatment of cancer; however their toxicity profile must be investigated prior to assess their anticancer efficacy against human malignancies.
Clinical Trials Using Curcumin for Cancer Therapy
Several clinical trials have been conducted to evaluate the value of curcumin in cancer treatment. In a phase I clinical trial using combination treatment with curcumin and quercetin to regress adenomas in patients with familial adenomatous polyposis (FAP), all 5 patients had a decreased polyp number and size from baseline after 6 months of curcumin and quercetin treatment without appreciable toxicity [126], suggesting that curcumin could inhibit adenoma growth in vivo. Moreover, another phase I trial has been conducted to evaluate the dose and the biomarkers in fifteen patients with advanced colorectal cancer refractory to standard chemotherapies. It was found that a daily dose of 3.6 g curcumin caused 62% decreases in inducible PGE2 production in blood samples, indicating the biological activity in humans [127]. In addition, the effects of curcumin has been tested in a phase I clinical trial in patients with resected urinary bladder cancer, arsenic Bowen's disease of the skin, uterine cervical intraepithelial neoplasm, oral leucoplakia, and intestinal metaplasia of the stomach [128]. The results showed that histologic improvement of precancerous lesions was seen in 1 out of 2 patients with resected bladder cancer, 2 out of 7 patients of oral leucoplakia, 1 out of 6 patients of intestinal metaplasia of the stomach, 1 out of 4 patients with cervical intraepithelial neoplasm and 2 out of 6 patients with Bowen's disease after curcumin treatment. This study also demonstrated that curcumin was non-toxic to humans. Recently, a phase II trial of curcumin in patients with advanced pancreatic cancer has been reported [129]. In this study, out of 21 cases, two patients showed clinical evidence for biological activity after curcumin treatment. Among them, one had ongoing stable disease for more than 18 months and another patient had a significant tumor regression (73%) [129]. Theses results suggest that oral curcumin is well tolerated and has biological activity in some patients with cancers. More new clinical trials are being conducted to investigate the value of curcumin in cancer patients (www.ClinicalTrials.gov). These clinical trials have investigated the efficacy of curcumin in the prevention setting for the development of various cancers, and the combination treatment of cancer with conventional chemotherapeutic agents or other dietary agents. We also believe that cancer chemotherapy combined with curcumin or its analog supplement could be an important novel strategy for the treatment of cancers and/or for the prevention of cancer progression of most human malignancies.
CONCLUSIONS AND PERSPECTIVES
The data from in vitro experiments and in vivo animal and limited human studies clearly suggest that naturally occurring compounds including isoflavones and curcumin could exert their inhibitory effects on carcinogenesis, cancer cell growth and tumor progression, which could be mediated through the regulation of multiple signaling pathways. Both isoflavone and curcumin inhibit NF-κB and Akt pathways by targeting similar molecules in these two pathways. However, isoflavone has stronger inhibitory effect on AR and Notch signaling while curcumin show its potent effects on the inhibition of COX-2 and Wnt3/β-catenin, suggesting their differential effects on cellular signaling. It is important to note that the water solubility and bioavailability of these natural agents could significantly influence the efficacy of these natural compounds, suggesting that the synthetic analogs of isoflavone and curcumin based on the structure-activity assays, and the encapsulation of isoflavone, curcumin and perhaps their analogs with liposome or nanoparticle could significantly enhance anti-tumor activity of these natural agents. The in vitro and in vivo preclinical studies also suggest that these analogs and formulations of natural compound could serve as potent agents for the prevention of tumor progression and/or treatment of human malignancies either alone or in combination with conventional chemotherapeutics, which will likely open newer avenues for the successful treatment of cancer. However, further in-depth experimental investigations along with animal studies, and finally clinical trials are needed in order to fully evaluate the value of these naturally occurring compounds and their synthetic analogs together with novel formulations for the prevention of tumor progression and/or treatment of human malignancies guided by superior systemic as well as target tissue bioavailability and pharmacokinetic profiles.
ACKNOWLEDGMENTS
The authors’ work cited in this review was funded by grants from the National Cancer Institute, NIH (5R01CA083695, 5R01CA131151, and 1R01CA132794 awarded to FHS), and a sub-contract award to FHS from the University of Texas MD Anderson Cancer Center through a SPORE grant (5P20-CA101936) on pancreatic cancer awarded to James Abbruzzese. We also thank Guido and Puschelberg Foundation for their generous contribution for the completion of these studies cited in this review article.
REFERENCES
- 1.Johnson-Thompson MC, Guthrie J. Ongoing research to identify environmental risk factors in breast carcinoma. Cancer. 2000;88:1224–1229. doi: 10.1002/(sici)1097-0142(20000301)88:5+<1224::aid-cncr8>3.3.co;2-8. [DOI] [PubMed] [Google Scholar]
- 2.Walker B, Figgs LW, Zahm SH. Differences in cancer incidence, mortality, and survival between African Americans and whites. Environ Health Perspect. 1995;103 Suppl 8:275–281. doi: 10.1289/ehp.95103s8275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wiseman M. The second World Cancer Research Fund/American Institute for Cancer Research expert report. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Proc Nutr Soc. 2008;67:253–256. doi: 10.1017/S002966510800712X. [DOI] [PubMed] [Google Scholar]
- 4.Lee MM, Gomez SL, Chang JS, Wey M, Wang RT, Hsing AW. Soy and isoflavone consumption in relation to prostate cancer risk in China. Cancer Epidemiol Biomarkers Prev. 2003;12:665–668. [PubMed] [Google Scholar]
- 5.Smith-Warner SA, Spiegelman D, Yaun SS, Albanes D, Beeson WL, van den Brandt PA, et al. Fruits, vegetables and lung cancer: a pooled analysis of cohort studies. Int J Cancer. 2003;107:1001–1011. doi: 10.1002/ijc.11490. [DOI] [PubMed] [Google Scholar]
- 6.Lamartiniere CA, Cotroneo MS, Fritz WA, Wang J, Mentor-Marcel R, Elgavish A. Genistein chemoprevention: timing and mechanisms of action in murine mammary and prostate. J Nutr. 2002;132 doi: 10.1093/jn/132.3.552S. 552S-8. [DOI] [PubMed] [Google Scholar]
- 7.Mukhopadhyay A, Bueso-Ramos C, Chatterjee D, Pantazis P, Aggarwal BB. Curcumin downregulates cell survival mechanisms in human prostate cancer cell lines. Oncogene. 2001;20:7597–7609. doi: 10.1038/sj.onc.1204997. [DOI] [PubMed] [Google Scholar]
- 8.Li Y, Li X, Sarkar FH. Gene expression profiles of I3C- and DIM- treated PC3 human prostate cancer cells determined by cDNA microarray analysis. J Nutr. 2003;133:1011–1019. doi: 10.1093/jn/133.4.1011. [DOI] [PubMed] [Google Scholar]
- 9.Hastak K, Gupta S, Ahmad N, Agarwal MK, Agarwal ML, Mukhtar H. Role of p53 and NF-kappaB in epigallocatechin-3-gallate-induced apoptosis of LNCaP cells. Oncogene. 2003;22:4851–4859. doi: 10.1038/sj.onc.1206708. [DOI] [PubMed] [Google Scholar]
- 10.Lee SA, Shu XO, Li H, Yang G, Cai H, Wen W, et al. Adolescent and adult soy food intake and breast cancer risk: results from the Shanghai Women's Health Study. Am J Clin Nutr. 2009;89:1920–1926. doi: 10.3945/ajcn.2008.27361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kim MK, Kim JH, Nam SJ, Ryu S, Kong G. Dietary intake of soy protein and tofu in association with breast cancer risk based on a case-control study. Nutr Cancer. 2008;60:568–576. doi: 10.1080/01635580801966203. [DOI] [PubMed] [Google Scholar]
- 12.Hirose K, Imaeda N, Tokudome Y, Goto C, Wakai K, Matsuo K, et al. Soybean products and reduction of breast cancer risk: a case-control study in Japan. Br J Cancer. 2005;93:15–22. doi: 10.1038/sj.bjc.6602659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sonoda T, Nagata Y, Mori M, Miyanaga N, Takashima N, Okumura K, et al. A case-control study of diet and prostate cancer in Japan: possible protective effect of traditional Japanese diet. Cancer Sci. 2004;95:238–242. doi: 10.1111/j.1349-7006.2004.tb02209.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Li Y, Upadhyay S, Bhuiyan M, Sarkar FH. Induction of apoptosis in breast cancer cells MDA-MB-231 by genistein. Oncogene. 1999;18:3166–3172. doi: 10.1038/sj.onc.1202650. [DOI] [PubMed] [Google Scholar]
- 15.Li Y, Sarkar FH. Inhibition of nuclear factor kappaB activation in PC3 cells by genistein is mediated via Akt signaling pathway. Clin Cancer Res. 2002;8:2369–2377. [PubMed] [Google Scholar]
- 16.Barnes S. Effect of genistein on in vitro and in vivo models of cancer. J Nutr. 1995;125:777S–783S. doi: 10.1093/jn/125.3_Suppl.777S. [DOI] [PubMed] [Google Scholar]
- 17.Li Y, Sarkar FH. Gene expression profiles of genistein-treated PC3 prostate cancer cells. J Nutr. 2002;132:3623–3631. doi: 10.1093/jn/132.12.3623. [DOI] [PubMed] [Google Scholar]
- 18.Li Y, Sarkar FH. Down-regulation of invasion and angiogenesis-related genes identified by cDNA microarray analysis of PC3 prostate cancer cells treated with genistein. Cancer Lett. 2002;186:157–164. doi: 10.1016/s0304-3835(02)00349-x. [DOI] [PubMed] [Google Scholar]
- 19.Oppenheimer A. TURMERIC (CURCUMIN) IN BILIARY DISEASES. The Lancet. 1937;229:619–621. [Google Scholar]
- 20.Kunnumakkara AB, Anand P, Aggarwal BB. Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett. 2008;269:199–225. doi: 10.1016/j.canlet.2008.03.009. [DOI] [PubMed] [Google Scholar]
- 21.Surh YJ, Chun KS, Cha HH, Han SS, Keum YS, Park KK, et al. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res. 2001;480–481:243–268. doi: 10.1016/s0027-5107(01)00183-x. [DOI] [PubMed] [Google Scholar]
- 22.Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm. 2007;4:807–818. doi: 10.1021/mp700113r. [DOI] [PubMed] [Google Scholar]
- 23.Anand P, Nair HB, Sung B, Kunnumakkara AB, Yadav VR, Tekmal RR, et al. Design of curcumin-loaded PLGA nanoparticles formulation with enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo. Biochem Pharmacol. 2009 doi: 10.1016/j.bcp.2009.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 24.Wang D, Veena MS, Stevenson K, Tang C, Ho B, Suh JD, et al. Liposome-encapsulated curcumin suppresses growth of head and neck squamous cell carcinoma in vitro and in xenografts through the inhibition of nuclear factor kappaB by an AKT-independent pathway. Clin Cancer Res. 2008;14:6228–6236. doi: 10.1158/1078-0432.CCR-07-5177. [DOI] [PubMed] [Google Scholar]
- 25.Martin PM, Horwitz KB, Ryan DS, McGuire WL. Phytoestrogen interaction with estrogen receptors in human breast cancer cells. Endocrinology. 1978;103:1860–1867. doi: 10.1210/endo-103-5-1860. [DOI] [PubMed] [Google Scholar]
- 26.Fioravanti L, Cappelletti V, Miodini P, Ronchi E, Brivio M, Di FG. Genistein in the control of breast cancer cell growth: insights into the mechanism of action in vitro. Cancer Lett. 1998;130:143–152. doi: 10.1016/s0304-3835(98)00130-x. [DOI] [PubMed] [Google Scholar]
- 27.Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, et al. Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem. 1987;262:5592–5595. [PubMed] [Google Scholar]
- 28.Zhou Y, Lee AS. Mechanism for the suppression of the mammalian stress response by genistein, an anticancer phytoestrogen from soy. J Natl Cancer Inst. 1998;90:381–388. doi: 10.1093/jnci/90.5.381. [DOI] [PubMed] [Google Scholar]
- 29.Davis JN, Kucuk O, Djuric Z, Sarkar FH. Soy isoflavone supplementation in healthy men prevents NF-kappa B activation by TNF-alpha in blood lymphocytes. Free Radic Biol Med. 2001;30:1293–1302. doi: 10.1016/s0891-5849(01)00535-4. [DOI] [PubMed] [Google Scholar]
- 30.Wang J, Eltoum IE, Lamartiniere CA. Dietary genistein suppresses chemically induced prostate cancer in Lobund-Wistar rats. Cancer Lett. 2002;186:11–18. doi: 10.1016/s0304-3835(01)00811-4. [DOI] [PubMed] [Google Scholar]
- 31.Barnes S. The chemopreventive properties of soy isoflavonoids in animal models of breast cancer. Breast Cancer Res Treat. 1997;46:169–179. doi: 10.1023/a:1005956326155. [DOI] [PubMed] [Google Scholar]
- 32.Sarkar FH, Li Y, Wang Z, Kong D. Cellular signaling perturbation by natural products. Cell Signal. 2009;21:1541–1547. doi: 10.1016/j.cellsig.2009.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sarkar FH, Li YW. Targeting multiple signal pathways by chemopreventive agents for cancer prevention and therapy. Acta Pharmacol Sin. 2007;28:1305–1315. doi: 10.1111/j.1745-7254.2007.00689.x. [DOI] [PubMed] [Google Scholar]
- 34.Li Y, Kucuk O, Hussain M, Abrams J, Cher ML, Sarkar FH. Antitumor and antimetastatic activities of docetaxel are enhanced by genistein through regulation of osteoprotegerin/receptor activator of nuclear factor-kappaB (RANK)/RANK ligand/MMP-9 signaling in prostate cancer. Cancer Res. 2006;66:4816–4825. doi: 10.1158/0008-5472.CAN-05-3752. [DOI] [PubMed] [Google Scholar]
- 35.Davis JN, Kucuk O, Sarkar FH. Genistein inhibits NF-kappa B activation in prostate cancer cells. Nutr Cancer. 1999;35:167–174. doi: 10.1207/S15327914NC352_11. [DOI] [PubMed] [Google Scholar]
- 36.Li Y, Ahmed F, Ali S, Philip PA, Kucuk O, Sarkar FH. Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells. Cancer Res. 2005;65:6934–6942. doi: 10.1158/0008-5472.CAN-04-4604. [DOI] [PubMed] [Google Scholar]
- 37.Gong L, Li Y, Nedeljkovic-Kurepa A, Sarkar FH. Inactivation of NF-kappaB by genistein is mediated via Akt signaling pathway in breast cancer cells. Oncogene. 2003;22:4702–4709. doi: 10.1038/sj.onc.1206583. [DOI] [PubMed] [Google Scholar]
- 38.Chen CC, Sun YT, Chen JJ, Chiu KT. TNF-alpha-induced cyclooxygenase-2 expression in human lung epithelial cells: involvement of the phospholipase C-gamma 2, protein kinase C-alpha, tyrosine kinase, NF-kappa B-inducing kinase, and I-kappa B kinase 1/2 pathway. J Immunol. 2000;165:2719–2728. doi: 10.4049/jimmunol.165.5.2719. [DOI] [PubMed] [Google Scholar]
- 39.Natarajan K, Manna SK, Chaturvedi MM, Aggarwal BB. Protein tyrosine kinase inhibitors block tumor necrosis factor-induced activation of nuclear factor-kappaB, degradation of IkappaBalpha, nuclear translocation of p65, and subsequent gene expression. Arch Biochem Biophys. 1998;352:59–70. doi: 10.1006/abbi.1998.0576. [DOI] [PubMed] [Google Scholar]
- 40.Davis JN, Kucuk O, Djuric Z, Sarkar FH. Soy isoflavone supplementation in healthy men prevents NF-kappa B activation by TNF-alpha in blood lymphocytes. Free Radic Biol Med. 2001;30:1293–1302. doi: 10.1016/s0891-5849(01)00535-4. [DOI] [PubMed] [Google Scholar]
- 41.Li Y, Wang Z, Kong D, Li R, Sarkar SH, Sarkar FH. Regulation of Akt/FOXO3a/GSK-3beta/AR signaling network by isoflavone in prostate cancer cells. J Biol Chem. 2008;283:27707–27716. doi: 10.1074/jbc.M802759200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Su Y, Simmen RC. Soy isoflavone genistein upregulates epithelial adhesion molecule E-cadherin expression and attenuates beta-catenin signaling in mammary epithelial cells. Carcinogenesis. 2009;30:331–339. doi: 10.1093/carcin/bgn279. [DOI] [PubMed] [Google Scholar]
- 43.Su Y, Simmen FA, Xiao R, Simmen RC. Expression profiling of rat mammary epithelial cells reveals candidate signaling pathways in dietary protection from mammary tumors. Physiol Genomics. 2007;30:8–16. doi: 10.1152/physiolgenomics.00023.2007. [DOI] [PubMed] [Google Scholar]
- 44.Wang Z, Zhang Y, Banerjee S, Li Y, Sarkar FH. Notch-1 down-regulation by curcumin is associated with the inhibition of cell growth and the induction of apoptosis in pancreatic cancer cells. Cancer. 2006;106:2503–2513. doi: 10.1002/cncr.21904. [DOI] [PubMed] [Google Scholar]
- 45.Wang Z, Zhang Y, Li Y, Banerjee S, Liao J, Sarkar FH. Down-regulation of Notch-1 contributes to cell growth inhibition and apoptosis in pancreatic cancer cells. Mol Cancer Ther. 2006;5:483–493. doi: 10.1158/1535-7163.MCT-05-0299. [DOI] [PubMed] [Google Scholar]
- 46.Davis JN, Muqim N, Bhuiyan M, Kucuk O, Pienta KJ, Sarkar FH. Inhibition of prostate specific antigen expression by genistein in prostate cancer cells. Int J Oncol. 2000;16:1091–1097. doi: 10.3892/ijo.16.6.1091. [DOI] [PubMed] [Google Scholar]
- 47.Davis JN, Kucuk O, Sarkar FH. Expression of prostate-specific antigen is transcriptionally regulated by genistein in prostate cancer cells. Mol Carcinog. 2002;34:91–101. doi: 10.1002/mc.10053. [DOI] [PubMed] [Google Scholar]
- 48.Fritz WA, Wang J, Eltoum IE, Lamartiniere CA. Dietary genistein down-regulates androgen and estrogen receptor expression in the rat prostate. Mol Cell Endocrinol. 2002;186:89–99. doi: 10.1016/s0303-7207(01)00663-3. [DOI] [PubMed] [Google Scholar]
- 49.Li Y, Wang Z, Kong D, Li R, Sarkar SH, Sarkar FH. Regulation of Akt/FOXO3a/GSK-3beta/AR signaling network by isoflavone in prostate cancer cells. J Biol Chem. 2008;283:27707–27716. doi: 10.1074/jbc.M802759200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sarkar FH, Adsule S, Padhye S, Kulkarni S, Li Y. The role of genistein and synthetic derivatives of isoflavone in cancer prevention and therapy. Mini Rev Med Chem. 2006;6:401–407. doi: 10.2174/138955706776361439. [DOI] [PubMed] [Google Scholar]
- 51.Rice-Evans CA, Miller NJ, Paganga G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996;20:933–956. doi: 10.1016/0891-5849(95)02227-9. [DOI] [PubMed] [Google Scholar]
- 52.Choueiri TK, Wesolowski R, Mekhail TM. Phenoxodiol: isoflavone analog with antineoplastic activity. Curr Oncol Rep. 2006;8:104–107. doi: 10.1007/s11912-006-0044-2. [DOI] [PubMed] [Google Scholar]
- 53.Kamsteeg M, Rutherford T, Sapi E, Hanczaruk B, Shahabi S, Flick M, et al. Phenoxodiol--an isoflavone analog--induces apoptosis in chemoresistant ovarian cancer cells. Oncogene. 2003;22:2611–2620. doi: 10.1038/sj.onc.1206422. [DOI] [PubMed] [Google Scholar]
- 54.Mor G, Fu HH, Alvero AB. Phenoxodiol, a novel approach for the treatment of ovarian cancer. Curr Opin Investig Drugs. 2006;7:542–548. [PubMed] [Google Scholar]
- 55.Constantinou AI, Mehta R, Husband A. Phenoxodiol, a novel isoflavone derivative, inhibits dimethylbenz[a]anthracene (DMBA)-induced mammary carcinogenesis in female Sprague-Dawley rats. Eur J Cancer. 2003;39:1012–1018. doi: 10.1016/s0959-8049(03)00124-2. [DOI] [PubMed] [Google Scholar]
- 56.Alvero AB, O'Malley D, Brown D, Kelly G, Garg M, Chen W, et al. Molecular mechanism of phenoxodiol-induced apoptosis in ovarian carcinoma cells. Cancer. 2006;106:599–608. doi: 10.1002/cncr.21633. [DOI] [PubMed] [Google Scholar]
- 57.Sapi E, Alvero AB, Chen W, O'Malley D, Hao XY, Dwipoyono B, et al. Resistance of ovarian carcinoma cells to docetaxel is XIAP dependent and reversible by phenoxodiol. Oncol Res. 2004;14:567–578. doi: 10.3727/0965040042707943. [DOI] [PubMed] [Google Scholar]
- 58.Georgaki S, Skopeliti M, Tsiatas M, Nicolaou KA, Ioannou K, Husband A, et al. Phenoxodiol, an anticancer isoflavene, induces immunomodulatory effects in vitro and in vivo. J Cell Mol Med. 2009 doi: 10.1111/j.1582-4934.2009.00695.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Choueiri TK, Wesolowski R, Mekhail TM. Phenoxodiol: isoflavone analog with antineoplastic activity. Curr Oncol Rep. 2006;8:104–107. doi: 10.1007/s11912-006-0044-2. [DOI] [PubMed] [Google Scholar]
- 60.Gotoh T, Yamada K, Yin H, Ito A, Kataoka T, Dohi K. Chemoprevention of N-nitroso-N-methylurea-induced rat mammary carcinogenesis by soy foods or biochanin A. Jpn J Cancer Res. 1998;89:137–142. doi: 10.1111/j.1349-7006.1998.tb00541.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ullah MF, Shamim U, Hanif S, Azmi AS, Hadi SM. Cellular DNA breakage by soy isoflavone genistein and its methylated structural analogue biochanin A. Mol Nutr Food Res. 2009 doi: 10.1002/mnfr.200800547. [DOI] [PubMed] [Google Scholar]
- 62.Vasselin DA, Westwell AD, Matthews CS, Bradshaw TD, Stevens MF. Structural studies on bioactive compounds. 40.(1) Synthesis and biological properties of fluoro-, methoxyl-, and amino-substituted 3-phenyl-4H-1-benzopyran-4-ones and a comparison of their antitumor activities with the activities of related 2-phenylbenzothiazoles. J Med Chem. 2006;49:3973–3981. doi: 10.1021/jm060359j. [DOI] [PubMed] [Google Scholar]
- 63.Wang TC, Chen IL, Lu PJ, Wong CH, Liao CH, Tsiao KC, et al. Synthesis, antiproliferative, and antiplatelet activities of oxime- and methyloxime-containing flavone and isoflavone derivatives. Bioorg Med Chem. 2005;13:6045–6053. doi: 10.1016/j.bmc.2005.06.004. [DOI] [PubMed] [Google Scholar]
- 64.Whatmore JL, Swann E, Barraja P, Newsome JJ, Bunderson M, Beall HD, et al. Comparative study of isoflavone, quinoxaline and oxindole families of anti-angiogenic agents. Angiogenesis. 2002;5:45–51. doi: 10.1023/a:1021528628524. [DOI] [PubMed] [Google Scholar]
- 65.Polkowski K, Popiolkiewicz J, Krzeczynski P, Ramza J, Pucko W, Zegrocka-Stendel O, et al. Cytostatic and cytotoxic activity of synthetic genistein glycosides against human cancer cell lines. Cancer Lett. 2004;203:59–69. doi: 10.1016/j.canlet.2003.08.023. [DOI] [PubMed] [Google Scholar]
- 66.Hussain M, Banerjee M, Sarkar FH, Djuric Z, Pollak MN, Doerge D, et al. Soy isoflavones in the treatment of prostate cancer. Nutr Cancer. 2003;47:111–117. doi: 10.1207/s15327914nc4702_1. [DOI] [PubMed] [Google Scholar]
- 67.Pendleton JM, Tan WW, Anai S, Chang M, Hou W, Shiverick KT, et al. Phase II trial of isoflavone in prostate-specific antigen recurrent prostate cancer after previous local therapy. BMC Cancer. 2008;8:132. doi: 10.1186/1471-2407-8-132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hamilton-Reeves JM, Rebello SA, Thomas W, Slaton JW, Kurzer MS. Isoflavone-rich soy protein isolate suppresses androgen receptor expression without altering estrogen receptor-beta expression or serum hormonal profiles in men at high risk of prostate cancer. J Nutr. 2007;137:1769–1775. doi: 10.1093/jn/137.7.1769. [DOI] [PubMed] [Google Scholar]
- 69.Kumar NB, Cantor A, Allen K, Riccardi D, Besterman-Dahan K, Seigne J, et al. The specific role of isoflavones in reducing prostate cancer risk. Prostate. 2004;59:141–147. doi: 10.1002/pros.10362. [DOI] [PubMed] [Google Scholar]
- 70.Vere White RW, Hackman RM, Soares SE, Beckett LA, Li Y, Sun B. Effects of a genistein-rich extract on PSA levels in men with a history of prostate cancer. Urology. 2004;63:259–263. doi: 10.1016/j.urology.2003.09.061. [DOI] [PubMed] [Google Scholar]
- 71.Holt PR, Katz S, Kirshoff R. Curcumin therapy in inflammatory bowel disease: a pilot study. Dig Dis Sci. 2005;50:2191–2193. doi: 10.1007/s10620-005-3032-8. [DOI] [PubMed] [Google Scholar]
- 72.Wongcharoen W, Phrommintikul A. The protective role of curcumin in cardiovascular diseases. Int J Cardiol. 2009;133:145–151. doi: 10.1016/j.ijcard.2009.01.073. [DOI] [PubMed] [Google Scholar]
- 73.Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol. 2009;41:40–59. doi: 10.1016/j.biocel.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Pari L, Tewas D, Eckel J. Role of curcumin in health and disease. Arch Physiol Biochem. 2008;114:127–149. doi: 10.1080/13813450802033958. [DOI] [PubMed] [Google Scholar]
- 75.Limtrakul P. Curcumin as chemosensitizer. Adv Exp Med Biol. 2007;595:269–300. doi: 10.1007/978-0-387-46401-5_12. [DOI] [PubMed] [Google Scholar]
- 76.Bharti AC, Donato N, Singh S, Aggarwal BB. Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor-kappa B and IkappaBalpha kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis. Blood. 2003;101:1053–1062. doi: 10.1182/blood-2002-05-1320. [DOI] [PubMed] [Google Scholar]
- 77.Hussain AR, Ahmed M, Al-Jomah NA, Khan AS, Manogaran P, Sultana M, et al. Curcumin suppresses constitutive activation of nuclear factor-kappa B and requires functional Bax to induce apoptosis in Burkitt's lymphoma cell lines. Mol Cancer Ther. 2008;7:3318–3329. doi: 10.1158/1535-7163.MCT-08-0541. [DOI] [PubMed] [Google Scholar]
- 78.Kunnumakkara AB, Diagaradjane P, Guha S, Deorukhkar A, Shentu S, Aggarwal BB, et al. Curcumin sensitizes human colorectal cancer xenografts in nude mice to gamma-radiation by targeting nuclear factor-kappaB-regulated gene products. Clin Cancer Res. 2008;14:2128–2136. doi: 10.1158/1078-0432.CCR-07-4722. [DOI] [PubMed] [Google Scholar]
- 79.Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, et al. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res. 2008;14:4491–4499. doi: 10.1158/1078-0432.CCR-08-0024. [DOI] [PubMed] [Google Scholar]
- 80.Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, et al. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res. 2008;14:4491–4499. doi: 10.1158/1078-0432.CCR-08-0024. [DOI] [PubMed] [Google Scholar]
- 81.Yu S, Shen G, Khor TO, Kim JH, Kong AN. Curcumin inhibits Akt/mammalian target of rapamycin signaling through protein phosphatase-dependent mechanism. Mol Cancer Ther. 2008;7:2609–2620. doi: 10.1158/1535-7163.MCT-07-2400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Weir NM, Selvendiran K, Kutala VK, Tong L, Vishwanath S, Rajaram M, et al. Curcumin induces G2/M arrest and apoptosis in cisplatin-resistant human ovarian cancer cells by modulating Akt and p38 MAPK. Cancer Biol Ther. 2007;6:178–184. doi: 10.4161/cbt.6.2.3577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kumar AP, Garcia GE, Ghosh R, Rajnarayanan RV, Alworth WL, Slaga TJ. 4-Hydroxy-3-methoxybenzoic acid methyl ester: a curcumin derivative targets Akt/NF kappa B cell survival signaling pathway: potential for prostate cancer management. Neoplasia. 2003;5:255–266. doi: 10.1016/S1476-5586(03)80057-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Squires MS, Hudson EA, Howells L, Sale S, Houghton CE, Jones JL, et al. Relevance of mitogen activated protein kinase (MAPK) and phosphotidylinositol-3-kinase/protein kinase B (PI3K/PKB) pathways to induction of apoptosis by curcumin in breast cells. Biochem Pharmacol. 2003;65:361–376. doi: 10.1016/s0006-2952(02)01517-4. [DOI] [PubMed] [Google Scholar]
- 85.Woo JH, Kim YH, Choi YJ, Kim DG, Lee KS, Bae JH, et al. Molecular mechanisms of curcumin-induced cytotoxicity: induction of apoptosis through generation of reactive oxygen species, down-regulation of Bcl-XL and IAP, the release of cytochrome c and inhibition of Akt. Carcinogenesis. 2003;24:1199–1208. doi: 10.1093/carcin/bgg082. [DOI] [PubMed] [Google Scholar]
- 86.Ryu MJ, Cho M, Song JY, Yun YS, Choi IW, Kim DE, et al. Natural derivatives of curcumin attenuate the Wnt/beta-catenin pathway through down-regulation of the transcriptional coactivator p300. Biochem Biophys Res Commun. 2008;377:1304–1308. doi: 10.1016/j.bbrc.2008.10.171. [DOI] [PubMed] [Google Scholar]
- 87.Jaiswal AS, Marlow BP, Gupta N, Narayan S. Beta-catenin-mediated transactivation and cell-cell adhesion pathways are important in curcumin (diferuylmethane)-induced growth arrest and apoptosis in colon cancer cells. Oncogene. 2002;21:8414–8427. doi: 10.1038/sj.onc.1205947. [DOI] [PubMed] [Google Scholar]
- 88.Yan C, Jamaluddin MS, Aggarwal B, Myers J, Boyd DD. Gene expression profiling identifies activating transcription factor 3 as a novel contributor to the proapoptotic effect of curcumin. Mol Cancer Ther. 2005;4:233–241. [PubMed] [Google Scholar]
- 89.Nakamura K, Yasunaga Y, Segawa T, Ko D, Moul JW, Srivastava S, et al. Curcumin down-regulates AR gene expression and activation in prostate cancer cell lines. Int J Oncol. 2002;21:825–830. [PubMed] [Google Scholar]
- 90.Ohtsu H, Xiao Z, Ishida J, Nagai M, Wang HK, Itokawa H, et al. Antitumor agents. 217. Curcumin analogues as novel androgen receptor antagonists with potential as anti-prostate cancer agents. J Med Chem. 2002;45:5037–5042. doi: 10.1021/jm020200g. [DOI] [PubMed] [Google Scholar]
- 91.Rao CV. Regulation of COX and LOX by curcumin. Adv Exp Med Biol. 2007;595:213–226. doi: 10.1007/978-0-387-46401-5_9. [DOI] [PubMed] [Google Scholar]
- 92.Anand P, Thomas SG, Kunnumakkara AB, Sundaram C, Harikumar KB, Sung B, et al. Biological activities of curcumin and its analogues (Congeners) made by man and Mother Nature. Biochem Pharmacol. 2008;76:1590–1611. doi: 10.1016/j.bcp.2008.08.008. [DOI] [PubMed] [Google Scholar]
- 93.Sharma RA, Euden SA, Platton SL, Cooke DN, Shafayat A, Hewitt HR, et al. Phase I clinical trial of oral curcumin: biomarkers of systemic activity and compliance. Clin Cancer Res. 2004;10:6847–6854. doi: 10.1158/1078-0432.CCR-04-0744. [DOI] [PubMed] [Google Scholar]
- 94.Garcea G, Berry DP, Jones DJ, Singh R, Dennison AR, Farmer PB, et al. Consumption of the putative chemopreventive agent curcumin by cancer patients: assessment of curcumin levels in the colorectum and their pharmacodynamic consequences. Cancer Epidemiol Biomarkers Prev. 2005;14:120–125. [PubMed] [Google Scholar]
- 95.Padhye S, Yang H, Jamadar A, Cui QC, Chavan D, Dominiak K, et al. New difluoro Knoevenagel condensates of curcumin, their Schiff bases and copper complexes as proteasome inhibitors and apoptosis inducers in cancer cells. Pharm Res. 2009;26:1874–1880. doi: 10.1007/s11095-009-9900-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Milacic V, Banerjee S, Landis-Piwowar KR, Sarkar FH, Majumdar AP, Dou QP. Curcumin inhibits the proteasome activity in human colon cancer cells in vitro and in vivo. Cancer Res. 2008;68:7283–7292. doi: 10.1158/0008-5472.CAN-07-6246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Padhye S, Banerjee S, Chavan D, Pandye S, Swamy KV, Ali S, et al. Fluorocurcumins as Cyclooxygenase-2 Inhibitor: Molecular Docking, Pharmacokinetics and Tissue Distribution in Mice. Pharm Res. 2009;26:2438–2445. doi: 10.1007/s11095-009-9955-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hutzen B, Friedman L, Sobo M, Lin L, Cen L, De AS, et al. Curcumin analogue GO-Y030 inhibits STAT3 activity and cell growth in breast and pancreatic carcinomas. Int J Oncol. 2009;35:867–872. doi: 10.3892/ijo_00000401. [DOI] [PubMed] [Google Scholar]
- 99.Lin L, Hutzen B, Ball S, Foust E, Sobo M, Deangelis S, et al. New curcumin analogues exhibit enhanced growth-suppressive activity and inhibit AKT and signal transducer and activator of transcription 3 phosphorylation in breast and prostate cancer cells. Cancer Sci. 2009;100:1719–1727. doi: 10.1111/j.1349-7006.2009.01220.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Friedman L, Lin L, Ball S, Bekaii-Saab T, Fuchs J, Li PK, et al. Curcumin analogues exhibit enhanced growth suppressive activity in human pancreatic cancer cells. Anticancer Drugs. 2009;20:444–449. doi: 10.1097/CAD.0b013e32832afc04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cen L, Hutzen B, Ball S, Deangelis S, Chen CL, Fuchs JR, et al. New structural analogues of curcumin exhibit potent growth suppressive activity in human colorectal carcinoma cells. BMC Cancer. 2009;9:99. doi: 10.1186/1471-2407-9-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Labbozzetta M, Notarbartolo M, Poma P, Maurici A, Inguglia L, Marchetti P, et al. Curcumin as a possible lead compound against hormone-independent, multidrug-resistant breast cancer. Ann N Y Acad Sci. 2009;1155:278–283. doi: 10.1111/j.1749-6632.2009.03699.x. [DOI] [PubMed] [Google Scholar]
- 103.Amolins MW, Peterson LB, Blagg BS. Synthesis and evaluation of electron-rich curcumin analogues. Bioorg Med Chem. 2009;17:360–367. doi: 10.1016/j.bmc.2008.10.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Fuchs JR, Pandit B, Bhasin D, Etter JP, Regan N, Abdelhamid D, et al. Structure-activity relationship studies of curcumin analogues. Bioorg Med Chem Lett. 2009;19:2065–2069. doi: 10.1016/j.bmcl.2009.01.104. [DOI] [PubMed] [Google Scholar]
- 105.Qiu X, Liu Z, Shao WY, Liu X, Jing DP, Yu YJ, et al. Synthesis and evaluation of curcumin analogues as potential thioredoxin reductase inhibitors. Bioorg Med Chem. 2008;16:8035–8041. doi: 10.1016/j.bmc.2008.07.054. [DOI] [PubMed] [Google Scholar]
- 106.Kasinski AL, Du Y, Thomas SL, Zhao J, Sun SY, Khuri FR, et al. Inhibition of IkappaB kinase-nuclear factor-kappaB signaling pathway by 3,5-bis(2-flurobenzylidene)piperidin-4-one (EF24), a novel monoketone analog of curcumin. Mol Pharmacol. 2008;74:654–661. doi: 10.1124/mol.108.046201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Adams BK, Cai J, Armstrong J, Herold M, Lu YJ, Sun A, et al. EF24, a novel synthetic curcumin analog, induces apoptosis in cancer cells via a redox-dependent mechanism. Anticancer Drugs. 2005;16:263–275. doi: 10.1097/00001813-200503000-00005. [DOI] [PubMed] [Google Scholar]
- 108.Weber WM, Hunsaker LA, Roybal CN, Bobrovnikova-Marjon EV, Abcouwer SF, Royer RE, et al. Activation of NFkappaB is inhibited by curcumin and related enones. Bioorg Med Chem. 2006;14:2450–2461. doi: 10.1016/j.bmc.2005.11.035. [DOI] [PubMed] [Google Scholar]
- 109.Chandru H, Sharada AC, Bettadaiah BK, Kumar CS, Rangappa KS, Sunila, et al. In vivo growth inhibitory and anti-angiogenic effects of synthetic novel dienone cyclopropoxy curcumin analogs on mouse Ehrlich ascites tumor. Bioorg Med Chem. 2007;15:7696–7703. doi: 10.1016/j.bmc.2007.08.051. [DOI] [PubMed] [Google Scholar]
- 110.Ohori H, Yamakoshi H, Tomizawa M, Shibuya M, Kakudo Y, Takahashi A, et al. Synthesis and biological analysis of new curcumin analogues bearing an enhanced potential for the medicinal treatment of cancer. Mol Cancer Ther. 2006;5:2563–2571. doi: 10.1158/1535-7163.MCT-06-0174. [DOI] [PubMed] [Google Scholar]
- 111.Jankun J, Aleem AM, Malgorzewicz S, Szkudlarek M, Zavodszky MI, Dewitt DL, et al. Synthetic curcuminoids modulate the arachidonic acid metabolism of human platelet 12-lipoxygenase and reduce sprout formation of human endothelial cells. Mol Cancer Ther. 2006;5:1371–1382. doi: 10.1158/1535-7163.MCT-06-0021. [DOI] [PubMed] [Google Scholar]
- 112.Shi Q, Shih CC, Lee KH. Novel Anti-Prostate Cancer Curcumin Analogues that Enhance Androgen Receptor Degradation Activity. Anticancer Agents Med Chem. 2009 doi: 10.2174/187152009789124655. [DOI] [PubMed] [Google Scholar]
- 113.Handler N, Jaeger W, Puschacher H, Leisser K, Erker T. Synthesis of novel curcumin analogues and their evaluation as selective cyclooxygenase-1 (COX-1) inhibitors. Chem Pharm Bull (Tokyo) 2007;55:64–71. doi: 10.1248/cpb.55.64. [DOI] [PubMed] [Google Scholar]
- 114.Tamvakopoulos C, Dimas K, Sofianos ZD, Hatziantoniou S, Han Z, Liu ZL, et al. Metabolism and anticancer activity of the curcumin analogue, dimethoxycurcumin. Clin Cancer Res. 2007;13:1269–1277. doi: 10.1158/1078-0432.CCR-06-1839. [DOI] [PubMed] [Google Scholar]
- 115.Leyon PV, Kuttan G. Studies on the role of some synthetic curcuminoid derivatives in the inhibition of tumour specific angiogenesis. J Exp Clin Cancer Res. 2003;22:77–83. [PubMed] [Google Scholar]
- 116.Hahm ER, Gho YS, Park S, Park C, Kim KW, Yang CH. Synthetic curcumin analogs inhibit activator protein-1 transcription and tumor-induced angiogenesis. Biochem Biophys Res Commun. 2004;321:337–344. doi: 10.1016/j.bbrc.2004.06.119. [DOI] [PubMed] [Google Scholar]
- 117.John VD, Kuttan G, Krishnankutty K. Anti-tumour studies of metal chelates of synthetic curcuminoids. J Exp Clin Cancer Res. 2002;21:219–224. [PubMed] [Google Scholar]
- 118.Takahashi M, Uechi S, Takara K, Asikin Y, Wada K. Evaluation of an oral carrier system in rats: bioavailability and antioxidant properties of liposome-encapsulated curcumin. J Agric Food Chem. 2009;57:9141–9146. doi: 10.1021/jf9013923. [DOI] [PubMed] [Google Scholar]
- 119.Mach CM, Mathew L, Mosley SA, Kurzrock R, Smith JA. Determination of minimum effective dose and optimal dosing schedule for liposomal curcumin in a xenograft human pancreatic cancer model. Anticancer Res. 2009;29:1895–1899. [PubMed] [Google Scholar]
- 120.Li L, Braiteh FS, Kurzrock R. Liposome-encapsulated curcumin: in vitro and in vivo effects on proliferation, apoptosis, signaling, and angiogenesis. Cancer. 2005;104:1322–1331. doi: 10.1002/cncr.21300. [DOI] [PubMed] [Google Scholar]
- 121.Narayanan NK, Nargi D, Randolph C, Narayanan BA. Liposome encapsulation of curcumin and resveratrol in combination reduces prostate cancer incidence in PTEN knockout mice. Int J Cancer. 2009;125:1–8. doi: 10.1002/ijc.24336. [DOI] [PubMed] [Google Scholar]
- 122.Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MN. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur J Pharm Sci. 2009;37:223–230. doi: 10.1016/j.ejps.2009.02.019. [DOI] [PubMed] [Google Scholar]
- 123.Sou K, Inenaga S, Takeoka S, Tsuchida E. Loading of curcumin into macrophages using lipid-based nanoparticles. Int J Pharm. 2008;352:287–293. doi: 10.1016/j.ijpharm.2007.10.033. [DOI] [PubMed] [Google Scholar]
- 124.Bisht S, Feldmann G, Soni S, Ravi R, Karikar C, Maitra A, et al. Polymeric nanoparticle-encapsulated curcumin ("nanocurcumin"): a novel strategy for human cancer therapy. J Nanobiotechnology. 2007;5:3. doi: 10.1186/1477-3155-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Tiyaboonchai W, Tungpradit W, Plianbangchang P. Formulation and characterization of curcuminoids loaded solid lipid nanoparticles. Int J Pharm. 2007;337:299–306. doi: 10.1016/j.ijpharm.2006.12.043. [DOI] [PubMed] [Google Scholar]
- 126.Cruz-Correa M, Shoskes DA, Sanchez P, Zhao R, Hylind LM, Wexner SD, et al. Combination treatment with curcumin and quercetin of adenomas in familial adenomatous polyposis. Clin Gastroenterol Hepatol. 2006;4:1035–1038. doi: 10.1016/j.cgh.2006.03.020. [DOI] [PubMed] [Google Scholar]
- 127.Sharma RA, Euden SA, Platton SL, Cooke DN, Shafayat A, Hewitt HR, et al. Phase I clinical trial of oral curcumin: biomarkers of systemic activity and compliance. Clin Cancer Res. 2004;10:6847–6854. doi: 10.1158/1078-0432.CCR-04-0744. [DOI] [PubMed] [Google Scholar]
- 128.Cheng AL, Hsu CH, Lin JK, Hsu MM, Ho YF, Shen TS, et al. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 2001;21:2895–2900. [PubMed] [Google Scholar]
- 129.Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, et al. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res. 2008;14:4491–4499. doi: 10.1158/1078-0432.CCR-08-0024. [DOI] [PubMed] [Google Scholar]




