Targeting multiple signal pathways by chemopreventive agents for cancer prevention and therapy
Invited review

Targeting multiple signal pathways by chemopreventive agents for cancer prevention and therapy1

Fazlul H Sarkar2, Yi-wei Li

Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA

1This work was funded by grants from the National Cancer Institute (No 5R01CA083695, 5R01CA101870, and 5R01CA108535 awarded to FHS), a subcontract award to FHS from the University of Texas MD Anderson Cancer Center through a Specialized Program of Research Excellence (SPORE) grant (No 5P20-CA101936) on pancreatic cancer awarded to James Abbruzzese, and a grant from the Department of Defense (DOD Prostate Cancer Research Program DAMD17-03-1-0042 awarded to FHS).

2Correspondence to Dr Fazlul H SARKAR.
Phn 1-313-576-8327. Fax 1-313-576-8389.
E-mail fsarkar@med.wayne.edu


Abstract: In recent years, growing interest has been focused on the field of cancer prevention. Cancer prevention by chemopreventive agents offers significant promise for reducing the incidence and mortality of cancer. Chemopreventive agents may exert their effects either by blocking or metabolizing carcinogens or by inhibiting tumor cell growth. Another important benefit of chemopreventive agents is their non-toxic nature. Therefore, chemopreventive agents have recently been used for cancer treatment in combination with chemotherapeutics or radiotherapy, uncovering a novel strategy for cancer therapy. This strategy opens a new avenue from cancer prevention to cancer treatment. In vitro and in vivo studies have demonstrated that chemopreventive agents could enhance the antitumor activity of chemotherapeutics, improving the treatment outcome. Growing evidence has shown that chemopreventive agents potentiate the efficacy of chemotherapy and radiotherapy through the regulation of multiple signaling pathways, including Akt, NF-κB, c-Myc, cyclooxygenase-2, apoptosis, and others, suggesting a multitargeted nature of chemopreventive agents. However, further in-depth mechanistic studies, in vivo animal experiments, and clinical trials are needed to investigate the effects of chemopreventive agents in combination treatment of cancer with conventional cancer therapies. More potent natural and synthetic chemo-preventive agents are also needed to improve the efficacy of mechanism-based and targeted therapeutic strategies against cancer, which are likely to make a significant impact on saving lives. Here, we have briefly reviewed the role of chemopreventive agents in cancer prevention, but most importantly, we have reviewed how they could be useful for cancer therapy in combination with conventional therapies.

Keywords: chemoprevention; chemotherapy; Akt; NF-κB; c-Myc; cyclooxygenase-2


Submitted Apr 26, 2007. Accepted for publication Jul 12, 2007.

doi: 10.1111/j.1745-7254.2007.00689.x


Introduction

Despite significant investments in capital, manpower, and intellectual innovations for the development of cancer therapies over the past several decades, cancer still remains the second leading cause of death in the United States[1]. There-fore, cancer prevention has become an important avenue through which the fight against cancer could be feasible. Cancer chemoprevention applies specific natural or synthetic chemical compounds to inhibit or reverse carcinogenesis and to suppress the development of cancer from premalignant lesions. The emerging field of cancer prevention by chemopreventive agents offers significant promise for reducing the incidence and mortality of cancer. Several studies have shown that the incidence of cancer could be decreased by chemoprevention[24]. Chemoprevention appears to be a promising avenue for reducing cancer incidence in both high-risk groups and the general population with a low risk of developing cancer. Because of its promising effects, chemoprevention has been increasingly recognized as a powerful tool by cancer investigators and the National Cancer Institute[5].

Chemopreventive agents may exert their effects either by blocking or metabolizing carcinogens or by inhibiting tumor cell growth. Another important benefit of chemopre-ventive agents is their non-toxic nature. Therefore, chemo-preventive agents have been recently used in cancer treatment in combination with conventional chemotherapeutics or radiotherapy. In vitro and in vivo preclinical studies have demonstrated that chemopreventive agents could enhance the antitumor activity of chemotherapeutics[611], improving treatment outcome. The combination treatment may also decrease the systemic toxicity caused by chemotherapies because lower doses of therapeutic agents could be used, and as such, no systemic toxicity has been found from chemopreventive agents. Growing in vitro and in vivo data have shown that chemopreventive agents enhance the efficacy of chemotherapy and radiotherapy in various cancers through the regulation of Akt, NF-κB, c-Myc, cyclooxygenase (COX)-2, apoptotic, and other pathways, suggesting a novel and multitargeted therapeutic strategy against cancer[6,7,1114]. This strategy opens a new avenue from cancer prevention to cancer treatment.


Chemopreventive agents enhance antitumor activity of conventional cancer therapies

Most of the chemopreventive agents currently being studied are natural products or their derivatives. Many natural compounds, particularly plant products and dietary constituents, have been found to exhibit cancer chemopre-ventive activities both in vitro and in vivo[15,16]. Drug development from natural products is a rapidly emerging and highly promising strategy to identify novel anticancer agents. In recent years, novel combination treatments with conventional cancer therapies and chemopreventive agents have received much attention in cancer research. Experimental studies and clinical trials have demonstrated the beneficial effects of chemopreventive agents, including soy isoflavone, curcumin, epigallocatechin-3-gallate (EGCG), non-steroidal anti-inflammatory drugs (NSAIDs), resveratrol, indole-3-carbinol (I3C), and 3,3'-diindolyl-methane (DIM) in cancer prevention and treatment.

Evidence from epidemiologic and in vivo studies show a decreased risk of cancer associated with soy consumption[17]. Soy isoflavone genistein is believed to be responsible for the decreased risk of cancer. Isoflavone genistein has been found to inhibit the growth of various cancer cells in vitro and in vivo[17]. More importantly, the published studies have shown that isoflavone genistein could potentiate the antitumor effects of chemotherapeutic agents in various cancers in vitro and in vivo in preclinical studies. We have reported that in vitro genistein potentiated growth inhibition and apoptotic cell death caused by cisplatin, docetaxel, doxorubicin, gemcitabine, and CHOP (cyclophosphamidine, doxorubicin, vincristine, prednisone) in lymphoma and cancers of prostate, breast, pancreas, and lung[6,7,12,18]. We have also found that dietary genistein in vivo could enhance the antitumor activities of gemcitabine and docetaxel in a tumor model, resulting in apoptotic cell death and the inhibition of tumor growth[6,7]. Similar observations by other investigators have also showed that the antitumor effects of chemotherapeutics, including 5-fluorouracil (5-FU), adriamycin, and tamoxifen could be potentiated by genistein[13,19,20]. Genistein also enhanced the antitumor effect of bleomycin in HL-60 cells, but not in normal lymphocytes in an in vitro study[21]. The synergistic action of genistein and cisplatin or carmustine (BCNU) on the growth inhibition of glioblastoma and medulloblastoma cells has also been observ-ed[22,23]. These reports suggest that isoflavone genistein is not just a chemopreventive agent, but could also be used as a potential therapeutic agent in combination with other chemotherapeutics for cancer treatment. In radiotherapy, experimental studies have demonstrated that the combination of genistein and radiation exerted enhanced inhibitory effects on DNA synthesis, cell growth, colony formation, and metastasis[24,25]. Genistein also enhanced radiosensitivity in human esophageal and cervical cancer cells[26,27], suggesting the beneficial effects of genistein in cancer radio-therapy.

To enhance the antitumor activity of isoflavone, several isoflavone derivatives have been synthesized and used in in vitro and in vivo experiments and in clinical trials. These compounds have shown a low IC50 in the inhibition of cancer cell growth in vitro. Moreover, at low concentrations, these compounds were able to enhance the antitumor activity of clinically available chemotherapeutic agents, suggesting their potent effects as therapeutic agents for combination treatment. Phenoxodiol is one such analog of isoflav-one genistein and has shown a broad-spectrum, anticancer effect. In an animal study, phenoxodiol inhibited dimethyl-benz(a)anthracene (DMBA)-induced mammary carcinogenesis in female Sprague-Dawley rats, suggesting that phenoxodiol is an effective chemopreventive agent against DMBA-induced oncogenesis[28]. In experimental studies and clinical trials, phenoxodiol has been used both as a mono-therapy and in combination with standard chemothera-peutics. These studies have shown that in some cancers phenoxodiol appears to be strong enough to work on its own as a monotherapy. However, one of the major benefits of phenoxodiol is its ability to sensitize cancer cells to the antitumor effects of conventional chemotherapeutics[29]. It has been found in cancer cells that are susceptible to the effects of standard chemotherapeutics that phenoxodiol increases their sensitivity to those agents. In cancer cells that have become resistant to the effects of conventional chemo-therapeutics, phenoxodiol restores chemosensitivity[14,30]. By exposing chemoresistant cancer cells to phenoxodiol first, long-standing drug resistance is removed, making cancer cells susceptible once again to standard chemotherapeutics, such as cisplatin, carboplatin, taxanes, and gemcitabine. Phenoxodiol is currently undergoing clinical studies in the USA and Australia. So far, phase I/II clinical trials have shown some disease stabilization without severe toxicity[31].

In addition to isoflavone and its derivatives, other chemopreventive agents have shown their effects on the enhancement of the antitumor activities of chemotherapeutic agents. Curcumin has been found to inhibit the carcinogenic activity of azoxymethane or DMBA in the colon or orally in rats[32,33]. Curcumin also inhibited cancer cell growth and induced apoptotic cell death in various cancers[34]. Moreover, curcumin and celecoxib synergistically inhibited the growth of colorectal cancer cells[35]. Curcumin also potentiated the antitumor activities of cisplatin, doxorubicin, and Taxol in HA22T/VGH hepatic cancer cells, HeLa cells, or CAOV3 and SKOV3 ovarian cancer cells[810]. In addition, the combination treatment with curcumin and TRAIL increased the number of hypodiploid cells and induced DNA fragmentation in LNCaP cells[36]. More recently, curcumin was found to sensitize pancreatic cancer cells to gemcitabine-induced killing[37]. In cancer radiotherapy, curcumin at a low concentration also showed significant enhancement to radiation-induced clonogenic inhibition and apoptosis in PC-3 prostate cancer cells[38]. These results demonstrated that curcumin is a potent agent in cancer prevention and therapy.

Epidemiologic evidence showed that consumption of green tea, which contains EGCG, significantly decreased overall cancer incidence[39]. As an antioxidant and photoprotective agent, EGCG promotes cell cycle arrest and apoptosis in cancer cells through the modulations of cyclin/CDK and Bcl-2 family proteins[40]. EGCG in vivo also inhibits tumor promotion and metastasis in murine melanoma[41]. Importantly, it has been found that EGCG combined with tamoxifen significantly induced apoptosis and growth inhibition in MDA-MB-231 human breast cancer cells[42]. EGCG could also chemosensitize resistant tumor cells to doxorubicin in the human carcinoma xenograft model[43], suggesting its effects on cancer therapy in combination with chemo-therapeutics.

COX-2 inhibitor NSAIDs have been shown to decrease the risk of various cancers, including colon and lung cancers[44,45]. NSAIDs exert apoptotic effects in a variety of cancer cells, including esophageal, liver, colon, lung, oral, and bladder cancer cells in a COX-2-dependent or COX-2-independent manner through the regulation of other molecules in cellular signaling pathways[46]. Celecoxib, the first selective COX-2 inhibitor approved for the chemoprevention of colon cancer in patients with familial adenomatous polyposis, has also been found to decrease the incidence of various cancers in various animal models with no associated toxicity[47]. Interestingly, the forced expression of COX-2 caused enhancement in multiple drug resistance (MDR)1 expression and functional activity, suggesting the existence of a causal link between COX-2 activity and MDR1 expression[48]. Therefore, the use of COX-2 inhibitors to decrease MDR1 function may enhance the accumulation of chemotherapy agents and decrease the resistance of tumors to chemotherapeutic drugs. Altorki et al found that celecoxib enhanced the response to paclitaxel and carboplatin in early-stage, non-small-cell lung cancer[49]. Moreover, selective COX-2 inhibitors were found to enhance tumor response to radiotherapy or radiochemotherapy, suggesting that these agents can improve the response of various cancers to conventional cancer therapies[5052].

Resveratrol is a phytoalexin mainly found in grapes. It exhibits anticancer properties in a variety of cancer cells in vitro, including lymphoid, myeloid, breast, prostate, and colon cancers[53]. Importantly, resveratrol has been reported to sensitize non-Hodgkin’s lymphoma and multiple myeloma cells to paclitaxel-mediated apoptosis[54]. The oral administration of proanthocyanidin, another compound from grapes, has also been found to decrease the tumor progression and the size of cutaneous carcinomas in an animal study[55]. Another murine study showed that the administration of grape seed extract significantly reduced metastatic melanoma pulmonary nodules[56]. Moreover, proanthocyanidin has been found to enhance doxorubicin-induced antitumor effects and reverse drug resistance in doxorubicin-resistant K562/DOX cells, breast cancer cells, and mouse tumor xenograft models[57,58].

We and others have found that I3C from cruciferous vegetables combined with cisplatin or tamoxifen could inhibit the growth of PC-3 prostate and MCF-7 breast cancer cells more effectively than either agent alone[59,60]. Phenethyl-ITC (PEITC) is another compound from cruciferous vegetables and has generated a great deal of research interest due to its cancer chemopreventive activity. PEITC administration was shown to significantly inhibit carcinogen-induced oncogenesis in mouse models[6163]. More impor-tantly, PEITC has been found to inhibit angiogenesis in vitro and ex vivo, suggesting that it is not just a chemopreventive agent, but could also be used for cancer therapy[64]. Recent reports showed that sulforaphane, another compound from cruciferous vegetables, exerted its antiproliferative activity in Akt-overexpressing ovarian cancer cells[65]. These findings suggest the beneficial effects of cruciferous vegetables in the fight against cancer.

Vitamin D obtained from the diet and synthesized in the skin in response to UVB exposure has been advocated as a potential preventive agent for prostate, colon, and lung cancers[6668]. Analogs of vitamin D were also shown to potentiate the antiproliferative effect of doxorubicin, cisplatin, and genistein in vitro[69]. N-(4-hydroxyphenyl) retinamide (4HPR; fenretinide), a synthetic derivative of retinoic acid, has been found to exert potent pro-apoptotic effects on a variety of cancer cells[70]. A recent report showed that 4HPR combined with low doses of celecoxib more potently inhibited growth and induced the apoptosis of premalignant and tumorigenic bronchial epithelial cell lines[71].

These reports clearly demonstrate that chemopreventive agents (natural or synthetic agents, which inhibit the development of cancer) could be used in cancer treatment to further enhance the antitumor activities of conventional chemotherapeutics (which have been used in the treatment of cancer in clinics) and radiotherapy.


Targeting cellular signaling pathways by chemopreventive agents in cancer prevention and treatment

In this review, we present a succinct summary of the major signaling pathways that are regulated by chemopreventive agents. It has been known that chemopreventive agents exert their inhibitory effects on carcinogenesis through multiple signaling pathways, including Akt, NF-κB, mitogen-activated protein kinase (MAPK), p53, COX-2, Ras, and many other molecules that are known to regulate cell growth and apoptotic pathways. However, the molecular mechanisms by which chemopreventive agents potentiate the antitumor effects of cancer therapies have not been fully elucidated. It is known that chemotherapy and radiotherapy can induce drug resistance in cancer cells, resulting in treatment failure. Emerging evidence has demonstrated that MDR, NF-κB, Akt, and some molecules in the apoptotic pathway are involved in the development of drug resistance. Chemopreventive agents could sensitize cancer cells to cancer therapies through the regulation of Akt, c-Myc, NF-κB, COX-2, and apoptotic pathways, all of which are known to play important roles in the regulation of cell survival and cell growth (Figure 1).

Figure 1 Chemopre-ventive agents enhance anti-tumor effects of chemotherapy through the regulation of Akt, NF-κB, c-Myc, COX-2, and apoptosis pathways.

Regulation of the Akt pathway It has been well known that the Akt signaling pathway plays important roles in the control of cell survival. Many chemopreventive agents have been found to inhibit cancer cell growth and induce apoptosis through the inhibition of the Akt pathway[72]. A component of green tea, EGCG, promoted apoptosis in T24 human bladder cancer cells via the modulation of the PI3K/Akt pathway and Bcl-2 family proteins[73]. Indomethacin, a NSAID, has been found to induce apoptosis in renal cell carcinoma cells by activating Akt and MAPK signaling[46]. Other COX-2 inhibitors, including celecoxib and SC236, have also been found to inhibit cell growth and induce apop-tosis through the regulation of Akt and the COX-2 signaling pathway[70,74]. It has been known that the PEITC-mediated inhibition of the angiogenic features of human umbilical vein endothelial cells in vitro is associated with the inactivation of Akt, the suppression of vascular endothelial growth factor (VEGF) secretion, and the downregulation of VEGF receptor 2 protein levels[64]. In a study on sulforaphane from cruciferous vegetables, PI3K and both total Akt protein and active phosphorylated Akt (Ser473) were significantly decreased in sulforaphane-treated ovarian cancer cells, suggesting the inhibitory effect of sulforaphane on the Akt pathway[65]. Deguelin, a member of the flavonoid family with chemopre-ventive activities, has been found to decrease tumor incidence in animal models for lung, colon, mammary, and skin carcinogenesis through Akt inhibition[75,76]. We and others have found that isoflavone genistein could inhibit cancer cell growth and induce apoptosis through the downregulation of Akt. These results demonstrate that Akt is a target of chemopreventive agents in cancer prevention.

Growing evidence has also shown that activated Akt is critical for acquiring drug resistance[7779], therefore the downregulation of Akt by chemopreventive agents could sensitize cancer cells to chemotherapeutics or radiotherapy. We and other investigators have found that activated Akt was inhibited by isoflavone genistein combined with gemcitabine or radiation in pancreatic, cervical, and esophageal cancer cells, suggesting that the enhancement of chemotherapeutic or radiation effects by isoflavone genistein may be partially mediated by the inhibition of Akt signaling[6,26,27]. It has been found that genistein also enhanced necrotic-like cell death with the significant inhibition of Akt activity in breast cancer cells treated with genis-tein and adriamycin, suggesting that the enhanced growth inhibition by combination treatment is through the inactivation of the Akt pathway[19]. Kamsteeg et al reported that phenoxo-diol, one of the synthetic derivatives of genistein, could inhibit Akt signaling transduction and subsequently activate the caspase system, inhibiting X-linked inhibitor of apoptosis protein (XIAP) and in turn leading to increased chemosen-sitization[14]. It has been reported that curcumin downregul-ated the Taxol-induced phosphorylation of Akt, which interacts with NF-κB, suggesting that enhanced antitumor activity by curcumin is through the inactivation of the Akt and NF-κB pathways[9].

Regulation of the c-Myc/cyclin D/CDK pathway It is known that activated Akt can upregulate c-Myc through the activation of IkappaB kinase complex (IKK)/NF-κB and the inhibition of cyclin D1 and c-Myc proteolysis[80,81]. Almost all types of human cancers show high frequencies of c-Myc amplification or overexpression of its protein product, c-Myc. c-Myc can induce cyclin D1 which interacts with CDK4 and CDK6 to promote cell cycle progression[82,83]. It has been found that curcumin inhibited the expression of c-Myc and tumorigenesis[84,85]. Other chemo-preventive agents, including EGCG, also showed their ability to downregulate c-Myc[86,87]. It has been reported that chemotherapeutics, including cisplatin, doxorubicin, paclitaxel, and 5-FU can induce c-Myc expression[88]. Interes-tingly, the surviving cancer cells from cisplatin treatment display a significant elevation in c-Myc expression[89] and the enhanced antitumor activity of chemotherapeutics can be achieved by the combination treatment with low-dose c-Myc antisense oligonucleotides[90], suggesting that c-Myc could cause chemoresistance of cancer cells to chemothera-peutics. Experimental studies have also shown that the overexpression of cyclin D1 contributed to the chemoresistance of pancreatic cancer cells because of the dual roles of cyclin D1 in promoting cell proliferation and inhibiting drug-induced apoptosis[91]. Therefore, the chemopreventive agent, which downregulates c-Myc, cyclin D, and CDK, could be used in combination with chemotherapeutics to improve the treatment outcome in cancer therapy. It has been reported that the combination of I3C and tamoxifen caused a more pronounced decrease in CDK2-specific enzymatic activity, CDK6 expression, and the level of phosphorylated retinoblastoma protein, leading to the more effective inhibition of the growth of human MCF-7 breast cancer cells compared to either agent alone[59].

Regulation of the NF-κB pathway NF-κB is an inducible and ubiquitously expressed transcription factor which regulates cell survival, inflammation, and differentiation[92]. It is becoming increasingly clear that NF-κB signaling plays critical roles in cancer development and progression. A large portion of cancer cells, especially poorly differentiated cancer cells, shows activated NF-κB in the nucleus, suggesting that activated NF-κB regulates its downstream genes to promote cancer cell growth. Therefore, NF-κB has long been believed to be a target for the prevention and treatment of cancer. Several natural and synthetic chemopreventive agents that are able to inhibit the activation of NF-κB have been shown to either prevent cancer or to inhibit cell growth in animal models[93]. It has been reported that curcumin inhibited IKK, suppressed both constitutive and inducible NF-κB activation, and potentiated tumor necrosis factor (TNF)-induced apoptosis[94]. EGCG was also shown to inhibit the activation of IKK, the phosphorylation of IκBα, and the activation of NF-κB[95]. It has been found that deguelin, a member of the flavonoid family, also exerted its chemopreventive effects through the inhibition of NF-κB activity, even in the presence of inflammatory stimuli, such as TNF-α[96]. Other preventive agents, such as Ganoderma lucidum from an oriental medical mushroom and silibinin from the seeds of milk thistle, also inhibited cell growth and induced apoptosis through the inhibition of the NF-κB pathway[97,98]. We and others have reported that isoflavone genistein significantly inhibited cancer cell growth and induced apoptosis through the downregulation of NF-κB activity[99,100]. We also found that DIM from cruciferous vegetables inhibited NF-κB and its downstream genes VEGF, urokinase-plasminogen activator (uPA), and matrix metallo-proteinase (MMP)-9, leading to the inhibition of angio-genesis, invasion, and metastasis in prostate cancer cells[101]. These results demonstrate that NF-κB is a target of chemopreventive agents in cancer prevention.

More importantly, it has been well known that many chemotherapeutic agents induce the activity of NF-κB, which causes drug resistance in cancer cells[102]. Therefore, targeting NF-κB by chemopreventive agents could be a promising strategy to enhance the antitumor activity of chemotherapeutics in cancer treatment. From in vitro and in vivo experimental studies, we observed that NF-κB activity was significantly increased by cisplatin, docetaxel, gemcitabine, and radiation treatment and that the NF-κB-inducing activity of these agents was completely abrogated by isoflavone genistein treatment in prostate, breast, lung, and pancreatic cancer cells, suggesting that isoflavone genistein pretreatment inactivates NF-κB and thus contributes to increased growth inhibition and apoptosis induced by these agents[6,7,12,24,103]. We also found that isoflavone genistein enhanced the antitumor activity of CHOP by the inhibition of NF-κB in lymphoma cells[18], suggesting that the inhibition of NF-κB is the most important event for chemosensitization. Studies also showed that increased cell death by genistein and radiation occurred via the inhibition of NF-κB, leading to the altered expression of regulatory cell cycle proteins, cyclin B and p21WAF1/Cip1, thus promoting G2/M arrest and increased radiosensitivity[103]. Similarly, curcumin has been found to inhibit the activity of NF-κB, resulting in the sensitization of cancer cells to cisplatin or Taxol-induced apoptosis[9,104], suggesting its beneficial effects in cancer treatment.

Regulation of the COX-2 pathway COX plays an important role in the biosynthesis of prostanoids. COX-1 is constitutively expressed in many tissues and is involved in the housekeeping function of prostanoids, while COX-2, the inducible isoform, accounts for the elevated production of prostaglandins in response to various inflammatory stimuli, hormones, and growth factors. COX-2 has received more attention than COX-1 in cancer research because COX-2 expression is associated with cell growth regulation, tissue remodeling, and carcinogenesis. In recent years, COX-2 inhibitors and NSAIDs have been shown to decrease the risk of various cancers, including colon and lung cancers, suggesting that the downregulation of COX-2 could be one of the molecular mechanisms by which chemopreventive agents prevent and inhibit tumor growth. It has been found that curcumin or EGCG inhibited the expression of COX-2 along with the growth of colorectal or prostate cancer cells[35,105]. Experimental studies have also shown that curcumin and EGCG could downregulate COX-2 expression without any change in the expression of COX-1 at both the mRNA and protein levels in colorectal or prostate cancer cells, suggesting that a combination of chemopreventive agent, such as curcumin or EGCG, with chemotherapeutic agents could be an improved strategy for the treatment of colorectal or prostate cancer[35,105]. In support of this suggestion, the synergistic growth inhibitory effect of curcumin and celecoxib has been demonstrated in colorectal cancer cells through the inhibition of the COX-2 pathway[35]. The combination of 5-FU and isoflavone genistein also enhanced therapeutic effects in colon cancer through the COX-2 pathway[13], although the inhibition of tumor growth by some COX-2 inhibitors could also be mediated through the COX-2-independent pathway. Studies have shown that celecoxib at clinically feasible concentrations (≤5.6 µmol/L) markedly suppresses the biosynthesis of prostaglandin E2 (PGE2) in COX-2-expressing lung cancer cells[106]. However, much higher doses of celecoxib (≥25 µmol/L) are required for growth inhibition and apoptosis induction in cell culture systems, suggesting its COX-2-independent activity[107]. A recent report also showed that even at a low concentration, celecoxib combined with 4HPR inhibited cell growth and induced apoptosis though COX-2-independent mecha-nisms[70], and as such, suggest that further studies are needed to fully elucidate the mechanism of action of COX-2 inhibitors toward cancer prevention and therapy.

Regulation of the apoptotic pathway It has been well known that Akt, c-Myc, NF-κB, and COX-2 signaling could mediate apoptotic processes through the regulation of molecules in the apoptotic pathway. Therefore, the regulation of these signaling molecules by chemopreventive agents could lead to alterations in the levels of important molecules in the apoptotic pathway. EGCG, which inhibits Akt and NF-κB signaling, has been found to promote apoptosis in T24 human bladder cancer cells via the modulation of proteins in the Bcl-2 family[73]. It has also been reported that the genistein derivative phenoxodiol can bind to the tumor-associated NOX (tNOX) receptor, block its function, and subsequently inhibit the anti-apoptotic proteins XIAP and FADD-like ICE (FLICE) inhibitory protein, eventually inducing apoptotic cell death[14]. We and others have also found that isoflavone genistein combined with docetaxel or gemcitabine significantly inhibited Bcl-2, Bcl-XL, and survivin, and induced p21WAF1, suggesting that the enhanced antitumor effect in combination treatment is through the regulation of these important molecules in the apoptotic pathway[6,7]. It has been found that curcumin combined with cisplatin decreased the expression of several apoptosis-related genes, including c-Myc, Bcl-XL, c-IAP-2, neuronal apoptosiainhibitory protein (NAIP), and XIAP[8]. The combination of curcumin and TRAIL also induced the cleavage of procaspase-3, procaspase-8, and procaspase-9, the truncation of Bid, and the release of cytochrome c from the mitochondria in prostate cancer cells, indicating that the apoptotic pathway is triggered in prostate cancer cells treated with a combination of curcumin and TRAIL[36]. These findings suggest that chemopreventive agents also regulate the apoptotic pathway during cancer prevention and treatment.

Regulation of other pathways We have reported that the antitumor and antimetastatic activities of docetaxel are enhanced by isoflavone genistein through the regulation of osteoprotegerin/receptor activator of NF-κB (RANK)/RANK ligand/MMP-9 signaling in prostate cancer, suggesting that isoflavone genistein could be a promising non-toxic agent to improve the treatment outcome of metastatic prostate cancer with docetaxel[11]. Soy isoflavone also enhanced prostate cancer radiotherapy through the downregulation of apurinic/apyrimidinic endonuclease 1/redox factor-1 expression[108]. In addition, isoflavone genistein and its isoflavone analogs also showed the potential to decrease the side-effects of tamoxifen through metabolic interactions that inhibit the formation of α-hydroxytamoxifen via the inhibition of CYP1A2[109], resulting in the beneficial effects of isoflavone genistein in combination with tamoxifen.

It has been found that phenoxodiol, another analog of isoflavone, exerts its inhibitory effects on cancer through pleiotropic molecular mechanisms. In addition to the regulation of Akt and the caspase-dependent apoptotic pathway, phenoxodiol also induced G1 arrest by specific loss in cyclin-dependent kinase 2 activity through the p53-independent induction of p21WAF1/CIP1 [110]. In addition, phenoxodiol also inhibits the catalytic activity of topo II in a dose-dependent manner and stabilizes the topo II-mediated cleavable complex[111], demonstrating the pleiotropic effects of this agent in cancer prevention and treatment. The enhanced effects of chemotherapy by chemopreventive agents may also be related to immunopotentiating activities through the reduction of interleukin (IL)-6[10] and the enhancements of lymphocyte proliferation, NK cell cytotoxicity, the CD4+/CD8+ ratio, IL-2, and interferon (IFN)-γ productions[57]. These results clearly suggest that chemopreventive agents are pleiotropic and thus could be considered as multitarget agents that are likely to revolutionize our approach for the prevention and treatment of cancer.


Conclusion and perspective

The emerging evidence from in vitro and in vivo studies reviewed above demonstrate that chemopreventive agents could inhibit the development and progression of cancer by targeting multiple cell signaling pathways for cancer prevention and treatment. It is important to note that one chemopreventive agent could target several cell signaling pathways which crosstalk in a complex cellular signal transduction network that are responsible for the development and progression of cancer. Further in-depth mechanistic studies, in vivo animal experiments, and novel clinical trials are needed to investigate the effects of chemopreventive agents in the combination treatment of cancer with conventional cancer therapies. Moreover, further development of potent natural and synthetic chemopreventive agents are also needed to improve the efficacy of mechanism-based and targeted therapeutic strategies to win the battle against cancer.


References

  1. American Cancer Society. Cancer facts & figures 2007. Atlanta: American Cancer Society; 2007.
  2. Leung WK, Sung JJ. Chemoprevention of gastric cancer. Eur J Gastroenterol Hepatol 2006;18:867-71.
  3. Mokbel K. Risk-reducing strategies for breast cancer — a review of recent literature. Int J Fertil Womens Med 2003;48:274-7.
  4. McWilliams A, Lam S. New approaches to lung cancer prevention. Curr Oncol Rep 2002;4:487-94.
  5. Kelloff GJ, Lippman SM, Dannenberg AJ, Sigman CC, Pearce HL, Reid BJ, et al. Progress in chemoprevention drug development: the promise of molecular biomarkers for prevention of intraepithelial neoplasia and cancer-a plan to move forward. Clin Cancer Res 2006;12:3661-97.
  6. Banerjee S, Zhang Y, Ali S, Bhuiyan M, Wang Z, Chiao PJ, et al. Molecular evidence for increased antitumor activity of gemcitabine by genistein in vitro and in vivo using an orthotopic model of pancreatic cancer. Cancer Res 2005;65:9064-72.
  7. 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;65:6934-42.
  8. Notarbartolo M, Poma P, Perri D, Dusonchet L, Cervello M, D’Alessandro N. Antitumor effects of curcumin, alone or in combination with cisplatin or doxorubicin, on human hepatic cancer cells. Analysis of their possible relationship to changes in NF-kappaB activation levels and in IAP gene expression. Cancer Lett 2005;224:53-65.
  9. Bava SV, Puliappadamba VT, Deepti A, Nair A, Karunagaran D, Anto RJ. Sensitization of taxol-induced apoptosis by curcumin involves down-regulation of nuclear factor-kappaB and the serine/threonine kinase Akt and is independent of tubulin poly-merization. J Biol Chem 2005;280:6301-8.
  10. Chan MM, Fong D, Soprano KJ, Holmes WF, Heverling H. Inhibition of growth and sensitization to cisplatin-mediated killing of ovarian cancer cells by polyphenolic chemopreventive agents. J Cell Physiol 2003;194:63-70.
  11. 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;66:4816-25.
  12. Li Y, Ellis KL, Ali S, El-Rayes BF, Nedeljkovic-Kurepa A, Kucuk O, et al. Apoptosis-inducing effect of chemotherapeutic agents is potentiated by soy isoflavone genistein, a natural inhibitor of NF-kappaB in BxPC-3 pancreatic cancer cell line. Pancreas 2004;28:e90-e95.
  13. Hwang JT, Ha J, Park OJ. Combination of 5-fluorouracil and genistein induces apoptosis synergistically in chemo-resistant cancer cells through the modulation of AMPK and COX-2 signaling pathways. Biochem Biophys Res Commun 2005;332:433-40.
  14. 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;22:2611-20.
  15. Kelloff GJ, Crowell JA, Steele VE, Lubet RA, Malone WA, Boone CW, et al. Progress in cancer chemoprevention: development of diet-derived chemopreventive agents. J Nutr 2000;130:467S-71S.
  16. Pezzuto JM. Plant-derived anticancer agents. Biochem Pharmacol 1997;53:121-33.
  17. Messina MJ, Persky V, Setchell KD, Barnes S. Soy intake and cancer risk: a review of the in vitro and in vivo data. Nutr Cancer 1994;21:113-31.
  18. Mohammad RM, Al-Katib A, Aboukameel A, Doerge DR, Sarkar F, Kucuk O. Genistein sensitizes diffuse large cell lymphoma to CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone) chemotherapy. Mol Cancer Ther 2003;2:1361-8.
  19. Satoh H, Nishikawa K, Suzuki K, Asano R, Virgona N, Ichikawa T, et al. Genistein, a soy isoflavone, enhances necrotic-like cell death in a breast cancer cell treated with a chemotherapeutic agent. Res Commun Mol Pathol Pharmacol 2003;113–114:149-58.
  20. Tanos V, Brzezinski A, Drize O, Strauss N, Peretz T. Synergistic inhibitory effects of genistein and tamoxifen on human dysplastic and malignant epithelial breast cells in vitro. Eur J Obstet Gynecol Reprod Biol 2002;102:188-94.
  21. Lee R, Kim YJ, Lee YJ, Chung HW. The selective effect of genistein on the toxicity of bleomycin in normal lymphocytes and HL-60 cells. Toxicology 2004;195:87-95.
  22. Khoshyomn S, Manske GC, Lew SM, Wald SL, Penar PL. Synergistic action of genistein and cisplatin on growth inhibition and cytotoxicity of human medulloblastoma cells. Pediatr Neurosurg 2000;33:123-31.
  23. Khoshyomn S, Nathan D, Manske GC, Osler TM, Penar PL. Synergistic effect of genistein and BCNU on growth inhibition and cytotoxicity of glioblastoma cells. J Neurooncol 2002;57:193-200.
  24. Hillman GG, Forman JD, Kucuk O, Yudelev M, Maughan RL, Rubio J, et al. Genistein potentiates the radiation effect on prostate carcinoma cells. Clin Cancer Res 2001;7:382-90.
  25. Hillman GG, Wang Y, Kucuk O, Che M, Doerge DR, Yudelev M, et al. Genistein potentiates inhibition of tumor growth by radiation in a prostate cancer orthotopic model. Mol Cancer Ther 2004;3:1271-9.
  26. Yashar CM, Spanos WJ, Taylor DD, Gercel-Taylor C. Potentiation of the radiation effect with genistein in cervical cancer cells. Gynecol Oncol 2005;99:199-205.
  27. Akimoto T, Nonaka T, Ishikawa H, Sakurai H, Saitoh JI, Takahashi T, et al. Genistein, a tyrosine kinase inhibitor, enhanced radiosensitivity in human esophageal cancer cell lines in vitro: possible involvement of inhibition of survival signal transduction pathways. Int J Radiat Oncol Biol Phys 2001;50:195-201.
  28. 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;39:1012-8.
  29. 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.
  30. 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;14:567-78.
  31. Choueiri TK, Wesolowski R, Mekhail TM. Phenoxodiol: isoflavone analog with antineoplastic activity. Curr Oncol Rep 2006;8:104-7.
  32. Pereira MA, Grubbs CJ, Barnes LH, Li H, Olson GR, Eto I, et al. Effects of the phytochemicals, curcumin and quercetin, upon azoxymethane-induced colon cancer and 7,12-dimethylbenz[a]anthracene-induced mammary cancer in rats. Carcinogenesis 1996;17:1305-11.
  33. Li N, Chen X, Liao J, Yang G, Wang S, Josephson Y, et al. Inhibition of 7,12-dimethylbenz[a]anthracene (DMBA)-induced oral carcinogenesis in hamsters by tea and curcumin. Carcinogenesis 2002;23:1307-13.
  34. Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies. Anticancer Res 2003;23:363-98.
  35. Lev-Ari S, Strier L, Kazanov D, Madar-Shapiro L, Dvory-Sobol H, Pinchuk I, et al. Celecoxib and curcumin synergistically inhibit the growth of colorectal cancer cells. Clin Cancer Res 2005;11:6738-44.
  36. Deeb D, Xu YX, Jiang H, Gao X, Janakiraman N, Chapman RA, et al. Curcumin (diferuloyl-methane) enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in LNCaP prostate cancer cells. Mol Cancer Ther 2003;2:95-103.
  37. Kunnumakkara AB, Guha S, Krishnan S, Diagaradjane P, Gelovani J, Aggarwal BB. Curcumin potentiates antitumor activity of gemcitabine in an orthotopic model of pancreatic cancer through suppression of proliferation, angiogenesis, and inhibition of nuclear factor-{kappa}B-regulated gene products. Cancer Res 2007;67:3853-61.
  38. Chendil D, Ranga RS, Meigooni D, Sathishkumar S, Ahmed MM. Curcumin confers radiosensitizing effect in prostate cancer cell line PC-3. Oncogene 2004;23:1599-607.
  39. Nakachi K, Matsuyama S, Miyake S, Suganuma M, Imai K. Preventive effects of drinking green tea on cancer and cardiovascular disease: epidemiological evidence for multiple targeting prevention. Biofactors 2000;13:49-54.
  40. Nihal M, Ahmad N, Mukhtar H, Wood GS. Anti-proliferative and proapoptotic effects of (-)-epigallocatechin-3-gallate on human melanoma: possible implications for the chemoprevention of melanoma. Int J Cancer 2005;114:513-21.
  41. Taniguchi S, Fujiki H, Kobayashi H, Go H, Miyado K, Sadano H, et al. Effect of (-)-epigallocatechin gallate, the main constituent of green tea, on lung metastasis with mouse B16 melanoma cell lines. Cancer Lett 1992;65:51-4.
  42. Chisholm K, Bray BJ, Rosengren RJ. Tamoxifen and epigallocatechin gallate are synergistically cytotoxic to MDA-MB-231 human breast cancer cells. Anticancer Drugs 2004;15:889-97.
  43. Zhang Q, Wei D, Liu J. In vivo reversal of doxorubicin resistance by (-)-epigallocatechin gallate in a solid human carcinoma xenograft. Cancer Lett 2004;208:179-86.
  44. Rostom A, Dube C, Lewin G, Tsertsvadze A, Barrowman N, Code C, et al. Nonsteroidal anti-inflammatory drugs and cyclooxy-genase-2 inhibitors for primary prevention of colorectal cancer: a systematic review prepared for the U.S. Preventive Services Task Force. Ann Intern Med 2007;146:376-89.
  45. Sandler AB, Dubinett SM. COX-2 inhibition and lung cancer. Semin Oncol 2004;31:45-52.
  46. Ou YC, Yang CR, Cheng CL, Raung SL, Hung YY, Chen CJ. Indomethacin induces apoptosis in 786-O renal cell carcinoma cells by activating mitogen-activated protein kinases and AKT. Eur J Pharmacol 2007. in press.
  47. Grosch S, Maier TJ, Schiffmann S, Geisslinger G. Cyclooxy-genase-2 (COX-2)-independent anticarcinogenic effects of selective COX-2 inhibitors. J Natl Cancer Inst 2006;98:736-47.
  48. Sorokin A. Cyclooxygenase-2: potential role in regulation of drug efflux and multidrug resistance phenotype. Curr Pharm Des 2004;10:647-57.
  49. Altorki NK, Keresztes RS, Port JL, Libby DM, Korst RJ, Flieder DB, et al. Celecoxib, a selective cyclooxygenase-2 inhibitor, enhances the response to preoperative paclitaxel and carboplatin in early-stage non-small-cell lung cancer. J Clin Oncol 2003;21:2645-50.
  50. Komaki R, Liao Z, Milas L. Improvement strategies for molecular targeting: cyclooxygenase-2 inhibitors as radiosensitizers for non-small cell lung cancer. Semin Oncol 2004;31:47-53.
  51. Shin YK, Park JS, Kim HS, Jun HJ, Kim GE, Suh CO, et al. Radiosensitivity enhancement by celecoxib, a cyclooxygenase (COX)-2 selective inhibitor, via COX-2-dependent cell cycle regulation on human cancer cells expressing differential COX-2 levels. Cancer Res 2005;65:9501-9.
  52. Liao Z, Milas L, Komaki R, Stevens C, Cox JD. Combination of a COX-2 inhibitor with radiotherapy or radiochemotherapy in the treatment of thoracic cancer. Am J Clin Oncol 2003;26:S85-S91.
  53. Aggarwal BB, Bhardwaj A, Aggarwal RS, Seeram NP, Shishodia S, Takada Y. Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Res 2004;24:2783-840.
  54. Jazirehi AR, Bonavida B. Resveratrol modifies the expression of apoptotic regulatory proteins and sensitizes non-Hodgkin’s lymphoma and multiple myeloma cell lines to paclitaxel-induced apoptosis. Mol Cancer Ther 2004;3:71-84.
  55. Mittal A, Elmets CA, Katiyar SK. Dietary feeding of proantho-cyanidins from grape seeds prevents photocarcinogenesis in SKH-1 hairless mice: relationship to decreased fat and lipid peroxidation. Carcinogenesis 2003;24:1379-88.
  56. Martinez C, Vicente V, Yanez J, Alcaraz M, Castells MT, Canteras M, et al. The effect of the flavonoid diosmin, grape seed extract and red wine on the pulmonary metastatic B16F10 melanoma. Histol Histopathol 2005;20:1121-9.
  57. Zhang XY, Li WG, Wu YJ, Zheng TZ, Li W, Qu SY, et al. Proanthocyanidin from grape seeds potentiates anti-tumor activity of doxorubicin via immunomodulatory mechanism. Int Immunopharmacol 2005;5:1247-57.
  58. Sharma G, Tyagi AK, Singh RP, Chan DC, Agarwal R. Synergistic anti-cancer effects of grape seed extract and conventional cytotoxic agent doxorubicin against human breast carcinoma cells. Breast Cancer Res Treat 2004;85:1-12.
  59. Cover CM, Hsieh SJ, Cram EJ, Hong C, Riby JE, Bjeldanes LF, et al. Indole-3-carbinol and tamoxifen cooperate to arrest the cell cycle of MCF-7 human breast cancer cells. Cancer Res 1999;59:1244-51.
  60. Sarkar FH, Li Y. Indole-3-carbinol and prostate cancer. J Nutr 2004;134:3493S-8S.
  61. Morse MA, Amin SG, Hecht SS, Chung FL. Effects of aromatic isothiocyanates on tumorigenicity, O6-methylguanine forma-tion, and metabolism of the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone in A/J mouse lung. Cancer Res 1989;49:2894-7.
  62. Stoner GD, Morrissey DT, Heur YH, Daniel EM, Galati AJ, Wagner SA. Inhibitory effects of phenetyl isothiocyanate on N-nitrosobenzylmethylamine carcinogenesis in the rat esophagus. Cancer Res 1991;51:2063-8.
  63. Hecht SS, Kenney PM, Wang M, Trushin N, Upadhyaya P. Effects of phenethyl isothiocyanate and benzyl isothiocyanate, individually and in combination, on lung tumorigenesis induced in A/J mice by benzo[a]pyrene and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Cancer Lett 2000;150:49-56.
  64. Xiao D, Singh SV. Phenethyl isothiocyanate inhibits angiogenesis in vitro and ex vivo. Cancer Res 2007;67:2239-46.
  65. Chaudhuri D, Orsulic S, Ashok BT. Antiproliferative activity of sulforaphane in Akt-overexpressing ovarian cancer cells. Mol Cancer Ther 2007;6:334-45.
  66. Jacobs ET, Giuliano AR, Martinez ME, Hollis BW, Reid ME, Marshall JR. Plasma levels of 25-hydroxyvitamin D, 1,25-dihydroxyvitamin D and the risk of prostate cancer. J Steroid Biochem Mol Biol 2004;89-90:533-7.
  67. Kallay E, Bises G, Bajna E, Bieglmayer C, Gerdenitsch W, Steffan I, et al. Colon-specific regulation of vitamin D hydroxylases —a possible approach for tumor prevention. Carcinogenesis 2005;26:1581-9.
  68. Nakagawa K, Kawaura A, Kato S, Takeda E, Okano T. 1 alpha,25-Dihydroxyvitamin D(3) is a preventive factor in the metastasis of lung cancer. Carcinogenesis 2005;26:429-40.
  69. Siwinska A, Opolski A, Chrobak A, Wietrzyk J, Wojdat E, Kutner A, et al. Potentiation of the antiproliferative effect in vitro of doxorubicin, cisplatin and genistein by new analogues of vitamin D. Anticancer Res 2001;21:1925-9.
  70. Zou CP, Kurie JM, Lotan D, Zou CC, Hong WK, Lotan R. Higher potency of N-(4-hydroxyphenyl)retinamide than all-trans-retinoic acid in induction of apoptosis in non-small cell lung cancer cell lines. Clin Cancer Res 1998;4:1345-55.
  71. Schroeder CP, Kadara H, Lotan D, Woo JK, Lee HY, Hong WK, et al. Involvement of mitochondrial and Akt signaling pathways in augmented apoptosis induced by a combination of low doses of celecoxib and N-(4-hydroxyphenyl) retinamide in premalignant human bronchial epithelial cells. Cancer Res 2006;66:9762-70.
  72. Aggarwal BB, Shishodia S. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem Pharmacol 2006;71:1397-421.
  73. Qin J, Xie LP, Zheng XY, Wang YB, Bai Y, Shen HF, et al. A component of green tea, (-)-epigallocatechin-3-gallate, promotes apoptosis in T24 human bladder cancer cells via modulation of the PI3K/Akt pathway and Bcl-2 family proteins. Biochem Biophys Res Commun 2007;354:852-7.
  74. Fan XM, Jiang XH, Gu Q, Ching YP, He H, Xia HH, et al. Inhibition of Akt/PKB by a COX-2 inhibitor induces apoptosis in gastric cancer cells. Digestion 2006;73:75-83.
  75. Nair AS, Shishodia S, Ahn KS, Kunnumakkara AB, Sethi G, Aggarwal BB. Deguelin, an Akt inhibitor, suppresses Ikappa-Balpha kinase activation leading to suppression of NF-kappaB-regulated gene expression, potentiation of apoptosis, and inhibition of cellular invasion. J Immunol 2006;177:5612-22.
  76. Lee HY, Oh SH, Woo JK, Kim WY, Van Pelt CS, Price RE, et al. Chemopreventive effects of deguelin, a novel Akt inhibitor, on tobacco-induced lung tumorigenesis. J Natl Cancer Inst 2005;97:1695-9.
  77. McCubrey JA, Steelman LS, Franklin RA, Abrams SL, Chappell WH, Wong EW, et al. Targeting the RAF/MEK/ERK, PI3K/AKT and P53 pathways in hematopoietic drug resistance. Adv Enzyme Regul 2007. [Epub ahead of print].
  78. Han Z, Hong L, Wu K, Han S, Shen H, Liu C, et al. Reversal of multidrug resistance of gastric cancer cells by downregulation of Akt1 with Akt1 siRNA. J Exp Clin Cancer Res 2006;25:601-6.
  79. Tazzari PL, Cappellini A, Ricci F, Evangelisti C, Papa V, Grafone T, et al. Multidrug resistance-associated protein 1 expression is under the control of the phosphoinositide 3 kinase/Akt signal transduction network in human acute myelogenous leukemia blasts. Leukemia 2007;21:427-38.
  80. Romashkova JA, Makarov SS. NF-kappaB is a target of AKT in anti-apoptotic PDGF signalling. Nature 1999;401:86-90.
  81. Liang J, Slingerland JM. Multiple roles of the PI3K/PKB (Akt) pathway in cell cycle progression. Cell Cycle 2003;2:339-45.
  82. Daksis JI, Lu RY, Facchini LM, Marhin WW, Penn LJ. Myc induces cyclin D1 expression in the absence of de novo protein synthesis and links mitogen-stimulated signal transduction to the cell cycle. Oncogene 1994;9:3635-45.
  83. Mateyak MK, Obaya AJ, Sedivy JM. c-Myc regulates cyclin D-Cdk4 and -Cdk6 activity but affects cell cycle progression at multiple independent points. Mol Cell Biol 1999;19:4672-83.
  84. Lin JK. Suppression of protein kinase C and nuclear oncogene expression as possible action mechanisms of cancer chemo-prevention by Curcumin. Arch Pharm Res 2004;27:683-92.
  85. Wilson AJ, Velcich A, Arango D, Kurland AR, Shenoy SM, Pezo RC, et al. Novel detection and differential utilization of a c-myc transcriptional block in colon cancer chemoprevention. Cancer Res 2002;62:6006-10.
  86. Lin JK. Cancer chemoprevention by tea polyphenols through modulating signal transduction pathways. Arch Pharm Res 2002;25:561-71.
  87. Tao L, Kramer PM, Wang W, Yang S, Lubet RA, Steele VE, et al. Altered expression of c-myc, p16 and p27 in rat colon tumors and its reversal by short-term treatment with chemo-preventive agents. Carcinogenesis 2002;23:1447-54.
  88. Park JK, Chung YM, Kang S, Kim JU, Kim YT, Kim HJ, et al. c-Myc exerts a protective function through ornithine decarboxylase against cellular insults. Mol Pharmacol 2002;62:1400-8.
  89. Walker TL, White JD, Esdale WJ, Burton MA, DeCruz EE. Tumour cells surviving in vivo cisplatin chemotherapy display elevated c-myc expression. Br J Cancer 1996;73:610-4.
  90. Walker TL, Dass CR, Burton MA. Enhanced in vivo tumour response from combination of carboplatin and low-dose c-myc antisense oligonucleotides. Anticancer Res 2002;22:2237-45.
  91. Biliran H Jr, Wang Y, Banerjee S, Xu H, Heng H, Thakur A, et al. Overexpression of cyclin D1 promotes tumor cell growth and confers resistance to cisplatin-mediated apoptosis in an elastase-myc transgene-expressing pancreatic tumor cell line. Clin Cancer Res 2005;11:6075-86.
  92. Karin M. Nuclear factor-kappaB in cancer development and progression. Nature 2006;441:431-6.
  93. Greten FR, Karin M. The IKK/NF-kappaB activation pathway-a target for prevention and treatment of cancer. Cancer Lett 2004;206:193-9.
  94. 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;101:1053-62.
  95. Yang F, Oz HS, Barve S, de Villiers WJ, McClain CJ, Varilek GW. The green tea polyphenol (-)-epigallocatechin-3-gallate blocks nuclear factor-kappa B activation by inhibiting I kappa B kinase activity in the intestinal epithelial cell line IEC-6. Mol Pharmacol 2001;60:528-33.
  96. Dell’Eva R, Ambrosini C, Minghelli S, Noonan DM, Albini A, Ferrari N. The Akt inhibitor deguelin, is an angiopreventive agent also acting on the NF-kappaB pathway. Carcinogenesis 2007;28:404-13.
  97. Jiang J, Slivova V, Sliva D. Ganoderma lucidum inhibits proliferation of human breast cancer cells by down-regulation of estrogen receptor and NF-kappaB signaling. Int J Oncol 2006;29:695-703.
  98. Singh RP, Agarwal R. Prostate cancer chemoprevention by silibinin: bench to bedside. Mol Carcinog 2006;45:436-42.
  99. Davis JN, Kucuk O, Sarkar FH. Genistein inhibits NF-kappa B activation in prostate cancer cells. Nutr Cancer 1999;35:167-74.
  100. Singh AV, Franke AA, Blackburn GL, Zhou JR. Soy phyto-chemicals prevent orthotopic growth and metastasis of bladder cancer in mice by alterations of cancer cell proliferation and apoptosis and tumor angiogenesis. Cancer Res 2006;66:1851-8.
  101. Kong D, Li Y, Wang Z, Banerjee S, Sarkar FH. Inhibition of angiogenesis and invasion by 3,3'-diindolylmethane is mediated by the nuclear factor-{kappa}B downstream target genes MMP-9 and uPA that regulated bioavailability of vascular endothelial growth factor in prostate cancer. Cancer Res 2007;67:3310-9.
  102. Chuang SE, Yeh PY, Lu YS, Lai GM, Liao CM, Gao M, et al. Basal levels and patterns of anticancer drug-induced activation of nuclear factor-kappaB (NF-kappaB), and its attenuation by tamoxifen, dexamethasone, and curcumin in carcinoma cells. Biochem Pharmacol 2002;63:1709-16.
  103. Raffoul JJ, Wang Y, Kucuk O, Forman JD, Sarkar FH, Hillman GG. Genistein inhibits radiation-induced activation of NF-kappaB in prostate cancer cells promoting apoptosis and G2/M cell cycle arrest. BMC Cancer 2006;6:107.
  104. Venkatraman M, Anto RJ, Nair A, Varghese M, Karunagaran D. Biological and chemical inhibitors of NF-kappaB sensitize SiHa cells to cisplatin-induced apoptosis. Mol Carcinog 2005;44:51-9.
  105. Hussain T, Gupta S, Adhami VM, Mukhtar H. Green tea constituent epigallocatechin-3-gallate selectively inhibits COX-2 without affecting COX-1 expression in human prostate carcinoma cells. Int J Cancer 2005;113:660-9.
  106. Mao JT, Cui X, Reckamp K, Liu M, Krysan K, Dalwadi H, et al. Chemoprevention strategies with cyclooxygenase-2 inhibitors for lung cancer. Clin Lung Cancer 2005;7:30-9.
  107. Elle N, Irmg T, Greg V, Achi S, Helm S, Chri E, et al. Celecoxib loses its anti-inflammatory efficacy at high doses through activation of NF-{kappa}B. FASEB J 2001;15:1622-4.
  108. Raffoul JJ, Banerjee S, Singh-Gupta V, Knoll ZE, Fite A, Zhang H, et al. Down-regulation of apurinic/apyrimidinic endonuclease 1/redox factor-1 expression by soy isoflavones enhances prostate cancer radiotherapy in vitro and in vivo. Cancer Res 2007;67:2141-9.
  109. Chen J, Halls SC, Alfaro JF, Zhou Z, Hu M. Potential beneficial metabolic interactions between tamoxifen and isoflavones via cytochrome P450-mediated pathways in female rat liver microsomes. Pharm Res 2004;21:2095-104.
  110. Aguero MF, Facchinetti MM, Sheleg Z, Senderowicz AM. Phenoxodiol, a novel isoflavone, induces G1 arrest by specific loss in cyclin-dependent kinase 2 activity by p53-independent induction of p21WAF1/CIP1. Cancer Res 2005;65:3364-73.
  111. Constantinou AI, Husband A. Phenoxodiol (2H-1-benzopyran-7-0,1,3-(4-hydroxyphenyl)), a novel isoflavone derivative, inhibits DNA topoisomerase II by stabilizing the cleavable complex. Anticancer Res 2002;22:2581-5.
Cite this article as: Sarkar FH, Li Yw. Targeting multiple signal pathways by chemopreventive agents for cancer prevention and therapy1. Acta Pharmacologica Sinica 2007;28(9):1305-1315. doi: 10.1111/j.1745-7254.2007.00689.x