Abstract
The development of tumor drug resistance is one of the biggest obstacles on the way to achieve a favorable outcome of chemotherapy. Among various strategies that have been explored to overcome drug resistance, the combination of current chemotherapy with plant polyphenols as a chemosensitizer has emerged as a promising one. Plant polyphenols are a group of phytochemicals characterized by the presence of more than one phenolic group. Mechanistic studies suggest that polyphenols have multiple intracellular targets, one of which is the proteasome complex. The proteasome is a proteolytic enzyme complex responsible for intracellular protein degradation and has been shown to play an important role in tumor growth and the development of drug resistance. Therefore, proteasome inhibition by plant polyphenols could be one of the mechanisms contributing to their chemosensitizing effect. Plant polyphenols that have been identified to possess proteasome-inhibitory activity include (−)-epigallocatechins-3-gallate (EGCG), genistein, luteolin, apigenin, chrysin, quercetin, curcumin and tannic acid. These polyphenols have exhibited an appreciable effect on overcoming resistance to various chemotherapeutic drugs as well as multidrug resistance in a broad spectrum of tumors ranging from carcinoma and sarcoma to hematological malignances. The in vitro and in vivo studies on polyphenols with proteasome-inhibitory activity have built a solid foundation to support the idea that they could serve as a chemosensitizer for the treatment of cancer. In-depth mechanistic studies and identification of optimal regimen are needed in order to eventually translate this laboratory concept into clinical trials to actually benefit current chemotherapy.
Keywords: Polyphenols, flavonoids, EGCG, genistein, curcumin, structure-activity relationship, ubiquitin-proteasome pathway, proteasome inhibitors, bortezomib, chemotherapy, drug resistance, NF-κB, Bcl-2, chemosensitization, clinical trials
1. INTRODUCTION
The development of tumor drug resistance is the major obstacle to the success of chemotherapy in cancer treatment. A growing amount of studies suggest that plant polyphenols are able to sensitize drug-resistant tumors to chemotherapy via various mechanisms [1]. One of the involved mechanisms is targeting tumor ubiquitin-proteasome pathway which plays a critical role in tumor growth and drug resistance development [2, 3]. In this article, we will review natural polyphenols and their synthetic analogs, and the relationships between their chemical structures and biological activity, especially the proteasome-inhibitory activity. We will then discuss the molecular mechanisms underlying the ability of plant polyphenols to reverse drug resistance and enhance chemosensitivity. Lastly, we will summarize preclinical and clinical studies that combine plant polyphenols with conventional chemotherapy in the treatment of various human cancers, and discuss the issue of potential interaction of plant polyphenols with the first therapeutic proteasome inhibitor bortezomib.
2. PLANT POLYPHENOLS
2.1. Subclasses, structures and diet sources
Polyphenols are secondary metabolites of plants, protecting plants from ultraviolet radiation and pathogen assault [4, 5]. They constitute an important component of our daily diet, and are found in fruits, vegetables and beverages. Plant polyphenols can be divided into three classes based on their chemical structure: phenolic acids, flavonoids, and other non-flavonoid polyphenols [6]. Of note, the physiological action of the plant polyphenols is strongly affected by their chemical structure. For example, many isoflavones have the pseudohormonal property due to their structural similarity to the estradiol molecule, and they are consequently classified as phytoestrogens [4, 5].
Phenolic acids are a class of substances containing one or more hydroxyl functions and a carboxylic acid function at the benzene ring. Two subclasses of phenolic acids can be distinguished: derivatives of benzoic acid, whose polymer forms hydrolysable tannins (either gallotannins or ellagitannins), and derivatives of cinnamic acid, whose polymer forms lignins [4]. Flavonoids comprise the largest class of polyphenols, which are non-hydrolysable/condensed tannins with a common structure of two aromatic rings linked through three carbons (C6-C3-C6) [5]. Flavonoids can be further divided into six subclasses: flavones, flavonones, flavonols, flavanonols, flavanols and isoflavones [7]. For non-flavonoid polyphenols, two of the most well-known ones are curcumin and resveratrol [6].
The backbone structures, representative compounds and dietary sources of plant polyphenols in each class are summarized in Table 1.
Table 1.
Classes and subclasses of plant polyphenols.
Class | Backbone | Representative compounds | Dietary sources |
---|---|---|---|
Phenolic acid | |||
![]() |
Gallic acid | Blackberry | |
![]() |
Cinnamic acid | Cinnamon | |
| |||
Flavonoids | |||
Flavones |
![]() |
Luteolin (R′1=R′2=OH, R′3=H) Apigenin (R′1=R′3=H, R′2=OH) Chrysin (R′1=R′2=R′3=H) |
Parsley |
Flavonones |
![]() |
Eriodictyol (R′1=R′2=OH, R′3=H) Naringenin (R′1=R′3=H, R′2=OH) |
Orange juice, grapefruit juice |
Flavonols |
![]() |
Quercetin (R′1=R′2=OH, R′3=H) Kaempferol (R′1=R′3=H, R′2=OH) Myricetin (R′1=R′2=R′3=OH) |
Black and green tea, red onion |
Flavanonols |
![]() |
||
Flavanols |
![]() |
EC (R′1=R′2=OH, R′3=H; R1=H) ECG (R′1=R′2=OH, R′3=H; R1=gallate) EGC (R′1=R′2= R′3=OH; R1=H) EGCG (R′1=R′2= R′3=OH; R1=gallate) |
Green tea |
Isoflavones |
![]() |
Genistein (R2=OH) Daidzin (R2=H) |
Soybeans |
| |||
Non-flavonoid polyphenols | |||
e.g.![]() |
Curcumin | Turmeric | |
![]() |
Resveratrol | Red wine, grapes |
2.2. Bioavailability and bioactivity
Plant polyphenols are mainly absorbed in the small intestines or colon. Most plant polyphenols are present in the form of esters, glycosides, or polymers that cannot be absorbed directly. They need to be hydrolyzed by intestinal enzymes or colonic microflora before absorption [4, 8]. Once absorbed, polyphenols are subjected to intestinal and hepatic conjugations, mainly methylation, sulfation, and glucuronidation. O-methyltransferase (OMT) catalyzes the transfer of a methyl group from S-adenosyl-L-methionine (SAM) to polyphenols that have an O-phenolic moiety. The methylation occurs predominantly in the 3′ position of the polyphenols, with a minor proportion in the 4′ position. Sulfotransferase (SULT) catalyzes the transfer of a sulfate moiety from 3′-phosphoadenosine-5′-phosphosulfate to a hydroxyl group on various substrates including polyphenols, while UDP-glucuronosyltransferase (UGT) catalyzes the transfer of a glucuronic acid from UDP-glucuronic acid to its substrates including polyphenols [4, 8].
Not only protecting plants from ultraviolet radiation and pathogen assault, plant polyphenols are also protective against a variety of human diseases, particularly cardiovascular disease and neoplastic disease. The recorded pharmacological effects include anti-oxidation, anti-inflammation, anti-bacteria, anti-fungus, anti-virus and anti-neoplasm [4, 8]. These versatile bioactivities are achieved through modulating (either by activating or inhibiting) multiple intracellular targets and signaling pathways, including reactive oxygen species, nuclear receptors/transcription factors, NF-κB pathway, PI3K/Akt pathway, as well as the ubiquitin-proteasome pathway [1, 9]. In this review article, we will focus on polyphenols’ proteasome-inhibitory effect.
3. THE UBIQUITIN-PROTEASOME PATHWAY
3.1. Composition of proteasome
Eukaryotic 26S proteasome is a 2.4 MDa large complex consisting of one 20S core proteasome and two 19S regulatory caps [10] (Fig. 1). The 20S core proteasome harbors seven different α-subunits and seven different β-subunits in its twofold symmetrical α7β7β7α7 stacked complex, among which mainly three sets of β-subunits, β1 (caspase-like, or peptidyl-glutamyl peptide-hydrolyzing-like, PGPH-like), β2 (trypsin-like), and β5 (chymotrypsin-like) are proteolytically active [10] (Fig. 1). The 19S regulatory caps consist of the lid, which is responsible for recognition and docking of polyubiquitinylated proteins into the 20S core proteasome, and the base, which contains ATPase activity required for the unfolding and linearization of large proteins [10] (Fig. 1).
Fig. 1.
A schematic diagram of the 26S proteasome and its catalytic subunits inhibitable by various proteasome inhibitors.
Unlike common proteolytic enzymes, which contain a catalytic triad, proteasome catalytic subunits, namely β1, β2 and β5, belong to a special group termed N-terminal nucleophile hydrolases, which utilizes the side chain of the N-terminal residue as the catalytic nucleophile [11–13]. A variety of observations indicate that all three catalytic β-subunits indeed react with peptide bonds of substrates as well as with electrophilic functional groups of inhibitors through their –OH group of the N-terminal threonine (Thr1) [11–13] (Fig. 2A).
Fig. 2. Proteasome-inhibitory mechanisms of bortezomib and EGCG, as well as their direct interaction.
(A) Reversible inhibitory mechanism of bortezomib. (B) Irreversible inhibitory mechanism of EGCG. (C) Postulated interaction between bortezomib and EGCG.
3.2. Function of the ubiquitin-proteasome pathway
More than 90% of intracellular protein degradation is performed by the ubiquitin-proteasomal system. Proteins marked for degradation with polyubiquitin tags are hydrolyzed by the proteasome in an ATP-dependent manner. Differing from the lysosome, which is mainly responsible for degrading extracellular and transmembrane proteins, the proteasome is in charge of degrading intracellular proteins which are aberrantly folded or normally short-lived. A large and growing body of evidence indicates that the proteasome affects cell-cycle progression partially by regulating the turnover of cyclins and cyclin-dependent kinase inhibitors (CIP/KIP family). Inhibition of proteasome function could result in cell-cycle arrest [10]. In addition, the ubiquitin-proteasomal system can regulate the level of apoptotic activity by affecting the Bcl-2 family, NF-κB family, p53-MDM2 complex and others [10, 14]. Therefore, the proteasome is crucial for cell survival and proliferation.
3.3. Modulation of proteasome activity
Proteasome activity could be either up-regulated or down-regulated under in vivo conditions. In many cancer cases, proteasome activity is up-regulated by cellular oncogenic factors. Enhanced proteasome activity in turn promotes the degradation of tumor suppressor proteins, resulting in cancer cell survival and proliferation as well as the development of drug resistance [10, 15]. On the other hand, proteasome activity could be suppressed by several endogenous inhibitors as well as various exogenous inhibitors, including some synthetic compounds such as bortezomib (the first therapeutic proteasome inhibitor launched in 2003) and many natural products such as plant polyphenols [10, 15] (Fig. 1). In the following section, we will elaborate on how plant polyphenols modulate proteasome activity in drug-sensitive and drug-resistant cancer cells.
4. TARGETING TUMOR PROTEASOME BY PLANT POLYPHENOLS
4.1. EGCG
Tea is the second most popular beverage in the world after water. Green tea and, to a less extent, black tea are abundant in polyphenols. The major catechins in green tea are (−)-epigallocatechin-3-gallate [(−)-EGCG], (−)-epigallocatechin [(−)-EGC], (−)-epicatechin-3-gallate [(−)-ECG], and (−)-epicatechin [(−)-EC] [16] (Table 1). Among them, EGCG is the most abundant and active one, and has been extensively studied for its biological activities and cellular targets. One potentially important cellular target of EGCG is the proteasome [16]. Both naturally-occurring (−)-EGCG and its synthetic enantiomer (+)-EGCG are able to potently, specifically and irreversibly inhibit the chymotrypsin-like activity of proteasome β5 subunit in vitro (IC50 = 86–194 nM) and in vivo (1–10 μM) [17, 18] (Table 2). Of note, EGCG is able to interact with not only the β5 subunit in constitutive proteasome but also the β5i subunit in interferon-γ inducible immunoproteasome (referring to as BrAAP activity) with even higher affinity [19].
Table 2.
Median inhibitory concentration (IC50) for inhibition of the chymotrypsin-like activity of purified 20S proteasome and cellular 26S proteasome by plant polyphenols.
Polyphenols | IC50 in 20S (μM) | IC50 in 26S (μM) | References |
---|---|---|---|
( −)-EGCG | 0.086 ~ 0.194 | 1~10 * | [17] |
Luteolin | 1.5 ± 0.10 | 1.3 ± 0.11* | [36] |
Apigenin | 2.3 ± 0.20 | 1.9 ± 0.17* | [36] |
Chrysin | 4.9 ± 0.15 | 6.1 ± 0.44* | [36] |
Quercetin | 3.5 ± 0.05 | 2 ± 0.09* | [38] |
Genistein | 9.26 ± 1.2 | N/A | [107] |
Curcumin | 1.85 | 10~20# | [44] |
data from a leukemia cell line Jurkat cells;
data from two colon cancer cell lines, HCT-116 and SW480.
Similar to another proteasome inhibitor β-lactone, molecular docking and structure-activity relationship (SAR) analysis suggest that the ester carbon linking the C and D rings of EGCG is important for mediating its proteasome-inhibitory activity, which could be nucleophilically attacked by the –OH group of the catalytic Thr1 residue in β5 subunit [17] (Fig. 2B). In addition, the A ring mimics the Tyr residue and binds to the hydrophobic S1 pocket of the β5 subunit [20]. The B and D rings are oriented towards the water phase, and the number of –OH groups on them is positively correlated with the proteasome-inhibitory activity [21].
However, the vicinal trihydroxy groups of EGCG not only contribute to its proteasome-inhibitory activity, but also cause its susceptibility to autooxidation even under physiological conditions [22]. In an effort to increase the stability of EGCG, a series of analogs with different replacements on benzene rings were synthesized and examined. Among them, the peracetate-protected prodrug of EGCG (with all –OH groups being peracetylated) was found to have improved stability, bioavailability, and intracellular proteasome-inhibitory activity [23, 24]. More recently, a para-amino substituent at 4″ position and a fluoro-substituent at 3″ and 4″ positions are found to have increased stability as well and are worthy to be further investigated [25, 26].
Interestingly, among all oxidative metabolites of EGCG, one metabolite resulted from the oxidative decarboxylation of the A ring and the hydrolysis of the D ring (the gallate group) was recently identified to possess a similar ability to EGCG in modulating proteasome functionality and apoptotic pathways. However, differing from EGCG, which has covalent interaction with the catalytic Thr1, this metabolite is most likely to have non-covalent interaction with Thr1, resulting in the hindering of Thr1 [27].
Besides the autooxidation issue, methylation is another issue impeding the application of EGCG. As mentioned previously, once absorbed, EGCG is readily methylated by catechol-O-methyltransferase (COMT) at the 4′ and 4″ positions [28]. COMT is a mammalian OMT mainly involved in the inactivation of the catecholamine neurotransmitters. Methylation of EGCG significantly affects its binding to the proteasome β5 subunit; as the number of methyl groups increases, the inhibitory potency decreases accordingly [29]. Thus, cancer cells with lower COMT activity are more vulnerable to EGCG-mediated proteasome inhibition and apoptosis induction than those with higher COMT activity [30]. These findings raise the exciting possibility of a combinational treatment of cancer with EGCG and a COMT inhibitor [31].
Regarding the application of EGCG, one fact that needs to be kept in mind is that EGCG is found to have the potential to negate the therapeutic efficacy of bortezomib and other boronic acid-based proteasome inhibitors [32]. This neutralizing effect is due mainly to the direct reaction between the pyrocatechol moieties on EGCG and the boronic acid moiety on bortezomib, which confers its proteasome-inhibitory activity [32] (Fig. 2C). However, this issue is still disputable. Wang et al. reported that EGCG-induced apoptosis was potentiated by bortezomib in KM3 multiple myeloma cells [33]. More recently, a group from Millennium Pharmaceuticals, Inc. reported that no antagonism of bortezomib was seen where EGCG plasma concentrations were commensurate with dietary or supplemental intake in their preclinical study using CWR22 human prostate cancer cell xenograft mouse model [34].
4.2. Other dietary flavonoids: genistein, luteolin, apigenin, chrysin, and quercetin
After the discovery of the proteasome-inhibitory activity of EGCG, several other dietary flavonoids, including genistein [35], luteolin [36], apigenin [36–38], chrysin [36] and quercetin [38], were subsequently found to possess proteasome-inhibitory activity too. Table 2 listed the median inhibitory concentration (IC50) of these flavonoids for the inhibition of the chymotrypsin-like activity of both purified 20S proteasome and intracellular 26S proteasome. Docking studies suggested that the carbonyl carbon on the C ring might be the site of nucleophilic attack by the –OH group of Thr1 in proteasome β5 subunit [38]. In addition, SAR analysis found that flavonoids with a hydroxylated B ring and/or unsaturated C ring tend to be potent proteasome inhibitors and tumor cell apoptosis inducers [36]. Flavonoids with a pyrocatechol structure such as quercetin could also be methylated by COMT, but less efficiently than EGCG [39, 40]. Although methylated flavonoids have been suggested to be metabolically more stable than unmethylated ones, their proteasome-inhibitory activity and cytotoxicity against tumor cells are largely compromised [41]. Furthermore, similar to EGCG, those flavonoids with a vicinal diol structure could also antagonize the anticancer effect of bortezomib. Theoretically, all flavonoids with pyrocatechol or pyrogallol structure are potentially able to chemically react with the boric acid group in bortezomib and other boronic acid-based proteasome inhibitors [42].
4.3. Curcumin
Curcumin is extracted from turmeric, which is commonly used as spice. It falls into the non-flavonoid polyphenol group. Due to the observation that the curcumin-induced cell death is very similar to that of the proteasome inhibitors, the proteasome-inhibitory property of curcumin was proposed and verified in proliferating neuroblastoma cells [43]. Proteasome inhibition, as one of the mechanisms responsible for the curcumin-induced cell death, was further supported by local and metastatic colon cancer models both in vitro and in vivo [44]. Consistent with previous findings that the ester bond carbon of β-lactone and tea polyphenols as well as the carbonyl carbon of flavonoids confer their proteasome-inhibitory potencies, two carbonyl carbons on curcumin were found to be susceptible to nucleophilic attack by the –OH group of Thr1 in proteasome β5 subunit [44]. Although most polyphenols tend to be water-soluble, curcumin is an exception. Its poor solubility in water leads to its poor bioavailability in tissues remote from the gastrointestinal tract, which largely compromises its anticancer effect [45]. Among different analogs synthesized to improve the water solubility, amino acid conjugates of curcumin were found to be soluble in water and showed a potent antiproliferative effect in cell models, indicating its potential as a curcumin substituent [45]. Of note, in contrast to above-discussed flavonoids that contains pyrocatechol or pyrogallol structure, curcumin was reported to potentiate the effect of bortezomib against human multiple myeloma in nude mice model [46].
4.4. Tannic acid
Tannic acid (TA) refers to hydrolysable tannins, composed of either gallotannins or ellagitannins. Upon hydrolysis, gallotannin yields a D-glucose and 6 to 9 gallate groups which link to each other or the glucose core through ester bonds. Since ester bond-containing tea polyphenols are potent proteasome inhibitors, the ability of TA to inhibit the proteasome activity was tested and verified in purified 20S proteasome [47] and cellular 26S proteasome in different cell types [47, 48] as well as in tumor-bearing mouse models [48]. Inhibition of the proteasome function by TA resulted in increased p27 and Bax expression, and impaired cell cycle progression [47].
5. DRUG RESISTANCE OF CANCER
5.1. Mechanisms of tumor drug resistance
Development of tumor drug resistance is one of the major limiting factors of current chemotherapy. Drug resistance can occur through different mechanisms, some of which are intrinsic while others are acquired after chemotherapy. Acquired resistance is particularly critical, as tumors not only become resistant to the drugs originally used to treat them, but may also become cross-resistant to other drugs with different mechanisms of action [49]. The molecular mechanisms involved in drug resistance include evasion of apoptosis, increase in drug efflux, increase in drug detoxification, alterations in drug targets, loss of dependence on drug targets, as well as enhanced processing of drug-induced damage [49]. It has been well documented that the ubiquitin-proteasome pathway extensively participates in the development of drug resistance, as discussed in details below.
5.2. Involvement of the ubiquitin-proteasome pathway in drug resistance
A large amount of chemotherapeutic drugs exert antitumor effect via induction of apoptosis in tumor cells. Drug-resistant tumor cells evade apoptosis by enhancing survival signals and/or suppressing apoptotic signals. Up-regulated proteasome activity in tumor cells helps them evade apoptosis on at least two levels. First, increased proteasome activity could facilitate NF-κB survival pathway in tumor cells by accelerating I-κB turnover. Consistently, proteasome inhibitors were reported to enhance tumoricidal response to conventional chemotherapy in colon cancer model and others [50]. Increased proteasome activity in cancer cells could also interfere with the balance between pro- and anti-apoptotic Bcl-2 family members by accelerating Bax degradation and increasing Bcl-2 expression (which is a target gene of the transcription factor NF-κB). Therefore, proteasome inhibition not only stabilizes/activates Bax, but also down-regulates Bcl-2 expression [51–53].
Another important cause of drug resistance is the increased drug efflux. Drug efflux is mediated by some ATP-binding cassette (ABC) transporters, including ABCB1 (MDR1/P-glycoprotein/P-gp), ABCC subfamily members (MRPs) and ABCG2 (BCRP) [54, 55]. Newly synthesized P-gp is immature (only core-glycosylated, associated with chaperones) and trapped in endoplasmic reticulum (ER) [56]. Proteasome is believed to be involved in its maturation (full-glycosylation) as proteasome inhibition significantly suppressed P-gp maturation and exportation from ER to plasma membrane [56]. Proteasome inhibitors successfully reversed multidrug resistance phenotype in P-gp-overexpressing cancer cells through down-regulating P-gp expression [52, 57].
In addition to the involvement in the above-mentioned two major mechanisms of drug resistance, increased proteasome activity also facilitates the elimination of oxidatively damaged proteins that are caused by cytotoxic drugs so that relieves the drug-induced oxidative stress [58]. Proteasome inhibition led to considerable accumulation of oxidative proteins [58] and hypersensitized tumor cells to oxidative stress inducer [59].
Also, a crucial role of the ubiquitin-proteasomal system in regulating DNA repair has been unveiled. Among different DNA repair pathways, the ubiquitin-proteasomal system is involved in at least four pathways [60], namely nucleotide excision repair [61], post-replication repair [62], homologous recombination [63] and the fanconi anemia pathway [64]. Proteasome inhibitors significantly potentiated the cytotoxicity of cisplatin, a DNA damage agent that causes the formation of cisplatin-DNA adducts, by preventing nucleotide excision repair [65].
6. REVERSING DRUG RESISTANCE BY PLANT POLYPHENOLS
6.1. EGCG and overcoming drug resistance
Many studies have demonstrated the chemosensitizing effect of EGCG and other catechins in vitro and in vivo. Most of them found that EGCG reversed drug resistance by inhibiting the drug efflux transporter P-gp. For example, Kitagawa et al reported that catechins increased the cellular accumulation of P-gp substrates in the order of EGCG > ECG > EGC, while EC did not have any effect on substrates accumulation in P-gp-overexpressing KB-C2 human cervical epidermal carcinoma cells [66]. The chemosensitization by EGCG through the inhibition of P-gp expression/activity has also been validated in Caco-2 human intestinal adenocarcinoma cells (which are often used as a model for intestinal transport studies mediated by P-gp) [67] and tamoxifen-resistant MCF-7 human breast cancer cells [68], as well as in the xenograft mouse models using doxorubicin-resistant KB-A1 cells [69, 70] or doxorubicin-resistant BEL-7404/DOX hepatocarcinoma cells [71]. The study with tamoxifen-resistant MCF-7 cells also found that EGCG was able to inhibit the activity of the BCRP transporter [68]. EGCG was also reported to enhance the sensitivity of glioblastoma to temozolomide [72], of prostate carcinoma to doxorubicin [73], and of breast carcinoma to paclitaxel [74] in corresponding ectopic or orthotopic xenograft mouse models. Furthermore, the peracetate-protected prodrug of EGCG also exhibited a chemosensitizing effect in the treatment of leukemia cells by augmenting the efficacy of conventional chemotherapy daunorubicin and cytosine arabinoside [75]. A very recent study reported that polymer-based nanoparticle of polyphenols EGCG and theaflavin retained biological effectiveness with over 20-fold dose advantage than EGCG/theaflavin in exerting anti-cancer effects and also enhanced the potential of cisplatin in several different tumor cell types [76].
6.2. Genistein and overcoming drug resistance
Genistein has been widely studied as a promising agent for chemoprevention, chemotherapy, and chemosensitization. Genistein enhances the efficacy of chemotherapy mainly by inhibiting survival signals and/or enhancing apoptotic signals. Dr. Sarkar’s group reported that genistein pretreatment inactivated NF-κB and contributed to increased growth inhibition and apoptosis induced by cisplatin, docetaxel, doxorubicin or gemcitabine in various cancer cells including prostate, breast, lung, pancreatic and ovarian cancer cells [77–79]. Inactivation of Akt/NF-κB by genistein not only potentiated the proliferation-inhibiting and apoptosis-inducing effect of arsenic trioxide on human hepatocellular carcinoma cells in vitro, but also dramatically augmented its suppressive effect on tumor growth and angiogenesis in tumor-bearing nude mice [80]. Genistein also restored the sensitivity of platinum-resistant epithelial ovarian cancer cells to cisplatin, taxotere or gemcitabine via the inhibition of NF-κB and its target genes (c-IAP1, Bcl-2, Bcl-xL and survivin) [78]. Similarly, genistein restored the sensitivity to oxaliplatin in gemcitabine-resistant pancreatic cancer cells both in vitro and in vivo [81]. In addition to the effect in carcinoma cells, the combination of gemcitabine and genistein also showed enhanced antitumor efficacy in osteosarcoma cells through the same mechanism (abrogating Akt/NF-κB pathway) [82]. Genistein also enhanced the cytotoxicity of cisplatin and, to a lesser extent, vincristine in medulloblastoma cells [83]. Moreover, in acute myeloid leukemia cells, genistein exhibited synergistic anti-leukemia effect with cytosine arabinoside in both in vitro and in vivo models via affecting MAPK signaling pathway [84].
6.3. Curcumin and overcoming drug resistance
Extensive research has unveiled that curcumin can sensitize tumors to different chemotherapeutic drugs including doxorubicin, 5-FU, paclitaxel, vincristine, vinorelbine, melphalan, butyrate, cisplatin, oxaliplatin, celecoxib, gemcitabine, etoposide, sulfinosine and thalidomide [85]. Chemosensitization by curcumin has been observed in solid tumors of the lung, liver, pancreas, stomach, colon, head and neck, brain, breast, prostate, bladder, cervix, and ovary, as well as in hematological tumors of multiple myeloma, leukemia, and lymphoma [85]. Curcumin remarkably increased sensitivity to vinblastine/vincristine and reversed multidrug-resistance via modulation of P-gp expression and function in vincristine-selected P-gp overexpressing KB-V1 cervical epidermal carcinoma cells [86] and SGC7901/VCR gastric carcinoma cells [87]. Curcumin also sensitized the BRCP-expressing human breast cancer cells to different chemotherapeutic drugs including mitoxantrone, topotecan, SN-38 and doxorubicin by down-regulating the function of the BRCP transporter [88], which was further confirmed in a transgenic mice model [89]. Not only curcumin but also its major metabolite tetrahydrocurcumin was able to inhibit the efflux function of P-gp, MRP1 and BRCP and thereby reverse multidrug resistance [90]. Furthermore, curcumin was able to improve the sensitivity of drug resistant cancer cells by suppressing NF-κB signal. Dhandapani et al reported that curcumin sensitized glioma cells to clinically used chemotherapeutic agents (cisplatin, etoposide, camptothecin and doxorubicin) via inhibition of AP-1 and NF-κB, which correlated with reduced expression of Bcl-2 and IAP family members and some DNA repair enzymes [91]. Blockage of the NF-κB pathway by curcumin also potentiated the effect of paclitaxel in breast cancer xenograft [92] and the effect of cisplatin in head and neck squamous cell carcinoma xenograft [93]. Whether inhibition of the proteasome activity by curcumin is responsible for or contributes to the blockage of NF-κB pathway needs to be studied. Also, in a recent study, Yoon et al showed for the first time that curcumin effectively induced paraptosis, a new type of nonapoptotic cell death, in malignant breast cancer cell lines. Proteasomal dysfunction and superoxide anion contributed to those paraptotic changes characterized by vacuolation that begins with physical enlargement of mitochondria and ER. These findings suggest that curcumin could be beneficial to the treatment of apoptosis-resistant tumors [94].
7. CLINICAL TRIALS USING POLYPHENOLS AND CHEMOTHERAPY
7.1. EGCG in clinical trials
A few studies were conducted using green tea extract (GTE) as monotherapy in cancer patients. Although the oral consumption of GTE was well tolerated in these studies, its anticancer activity was unsatisfactory. For example, a phase I trial in patients with advanced lung cancer suggested that while relatively nontoxic at a dose of 3 g/m2 per day, GTE likely has limited activity as a cytotoxic agent [95]; a phase II trial of green tea suggested that oral administration of green tea powder 6 g/d carried limited antineoplastic activity, as defined by a decline in PSA level, among patients with androgen independent prostate cancer [96].
Therefore, a prospective clinical trial using green tea as a complementary/alternative medicine (CAM) was elicited in patients with hormone refractory prostate cancer. However, the results suggested that green tea, as CAM therapy, was found to have minimal clinical activity against hormone refractory prostate cancer [97]. In view of the greater efficacy of the prodrugs of EGCG (peracetate-protected or fluoro-substituted) than EGCG itself in xenograft mouse models of both estrogen-independent human breast cancer and androgen-independent human prostate cancer [98–100], our future goal is to translate these EGCG prodrugs into clinical studies.
Despite the excellent safety records of EGCG, in 2006, the US Food and Drug Administration (FDA) temporarily suspended all human trials of EGCG for additional review of toxicity data, and later allowed trials of Polyphenon E to resume. In a phase I trial in patients with asymptomatic Rai stage 0 to II chronic lymphocytic leukemia, daily oral consumption of Polyphenon E was well tolerated, and declines in absolute lymphocyte count and /or lymphadenopathy were observed in the majority of patients. Based on the encouraging phase I result, a phase II trial using 2 g Polyphenon E twice a day to evaluate the efficacy started in 2007 [101].
Furthermore, two clinical studies using Polyphenon E in combination with erlotinib are currently ongoing in patients with advanced non-small cell lung cancer and premalignant lesions of the head and neck, respectively (Table 3).
Table 3.
Combination of polyphenols and chemotherapy in clinical trials*
Polyphenols | Drugs | Tumor Type | Phase Stage | Country | NCT ID |
---|---|---|---|---|---|
EGCG (Polyphenon E) | Erlotinib | Advanced Non-Small Cell Lung Cancer | I/II | US | NCT00707252 |
Erlotinib | Premalignant Lesions of the Head and Neck | I | US | NCT01116336 | |
Genistein | Gemcitabine | Stage IV Breast Cancer | II | US | NCT00244933 |
Erlotinib/Gemcitabine | Locally Advanced or Metastatic Pancreatic Cancer | II | US | NCT00376948 | |
Gemcitabine | Pancreatic Cancer | I/II | SE | NCT01182246 | |
Genistein derivative (Phenoxodiol) | Docetaxel | Recurrent advanced ovarian epithelial cancer, fallopian tube cancer, or peritoneal cavity cancer | I/II | US | NCT00303888 |
Carboplatin | Recurrent platinum-resistant ovarian epithelial caner, fallopian tube cancer, or peritoneal cavity cancer | III | US | NCT00382811 | |
Curcumin | Gemcitabine | Pancreatic Cancer | II | IL | NCT00192842 |
Gemcitabine and Celebrex | Metastatic Colon Cancer | III | IL | NCT00295035 | |
Gemcitabine and Celebrex | Advance or Inoperable Pancreatic Cancer | III | IL | NCT00486460 |
data from www.clinicaltrials.gov.
7.2. Genistein in clinical trials
Genistein has been investigated as a sensitizer to gemcitabine and erlotinib in patients with breast cancer or pancreatic cancer (Table 3). In a phase II study in US, 20 patients with locally advanced or metastatic pancreatic cancer received gemcitabine and erlotinib-based chemotherapy, along with soy isoflavones (Novasoy@, 0.531 g twice daily, genistein is the major component). According to the RECIST criteria, 1 patient had a partial response (PR) and 6 had a stable disease (SD); the 6-month survival rate was 50% and the median survival time was 5.2 months. Since these results were comparable to the outcome observed in gemcitabine/erlotinib bitherapy without genistein, the investigators concluded that the antitumor activity of gemcitabine and erlotinib was not improved by soy isoflavones [102].
Although the result from prototype genistein is unfavorable, phenoxodiol, a synthetic derivative of the genistein, has been granted ‘fast track’ status by the US FDA for being developed as a chemosensitizer for platinum and taxane drugs used in the treatment of recurrent ovarian cancer [103]. A phase III study treating recurrent platinum-resistant ovarian cancer patients with phenoxodiol (400 mg/8h) and carboplatin (weekly) is currently ongoing (Table 3). Of note, phenoxodiol could be administered not only orally but also via intravenous infusion which would greatly enhance the bioavailability of the drug. In a phase II study, patients with platinum/taxane-refractory/resistant ovarian cancer received phenoxodiol (intravenously 3 mg/kg on days 1 and 2) and cisplatin or paclitaxel (on day 2) each cycle (7 days per cycle). Among 16 platinum-resistant patients, 3 patients had PR and 9 had SD. Among 15 taxane-resistant patients, one patient had a complete response, 2 had PR and 8 had SD. These results, particularly the result from the cisplatin-phenoxodiol combination, are quite encouraging and warrant further investigation [104].
7.3. Curcumin in clinical trials
Clinical studies using the combination of chemotherapy and curcumin have been conducted around the world. In a phase I/II trial in Japan, 21 patients with gemcitabine-resistant pancreatic cancer received curcumin (8 g/d) in a combination with gemcitabine-based chemotherapy (gemcitabine/S-1 bitherapy or gemcitabine monotherapy). No dose-limiting toxicity was observed in the Phase I study and oral curcumin 8 g/d was selected for the Phase II study. Among 18 evaluable patients, no patient had a PR and 5 had SD. These results warranted further investigation into the efficacy of curcumin in combined with gemcitabine-based chemotherapy [105].
In a phase I trial in France, 14 patients with advanced and metastatic breast cancer were treated with docetaxel, along with curcumin (does started from 0.5 g/d for 7 consecutive days and escalated until a dose-limiting toxicity occurred). The maximal tolerated dose of curcumin ended up as 8 g/d, and a dose of 6 g/d in combination with docetaxel was recommended. Among 8 evaluable patients, 5 patients had PR and 3 had SD. Based on the encouraging efficacy results, a comparative Phase II trial is ongoing [106].
Furthermore, a combined regimen of gemcitabine, Celebrex and curcumin in the treatment of colon cancer and pancreatic cancer has entered phase III trial in Israel (Table 3).
8. CONCLUSION AND PERSPECTIVES
The in vitro and in vivo studies reviewed above all suggest that plant polyphenols with proteasome-inhibitory activity (especially EGCG, genistein and curcumin) may serve as powerful agents for reversing tumor drug resistance and enhancing the efficacy of chemotherapy. Further in-depth mechanistic studies and high-throughput screening could help find the best combination to achieve the most favorable outcome. Most importantly, clinical trials are urgently needed to eventually translate this laboratory concept into clinical benefits.
Acknowledgments
This work was partially supported by grants from the National Cancer Institute (1R01CA120009 and 3R01CA120009-04S1, to QPD).
References
- 1.Garg AK, Buchholz TA, Aggarwal BB. Chemosensitization and radiosensitization of tumors by plant polyphenols. Antioxid Redox Signal. 2005;7(11–12):1630–47. doi: 10.1089/ars.2005.7.1630. [DOI] [PubMed] [Google Scholar]
- 2.Landis-Piwowar KR, Milacic V, Chen D, Yang H, Zhao Y, Chan TH, Yan B, Dou QP. The proteasome as a potential target for novel anticancer drugs and chemosensitizers. Drug Resist Updat. 2006;9(6):263–73. doi: 10.1016/j.drup.2006.11.001. [DOI] [PubMed] [Google Scholar]
- 3.McConkey DJ, Zhu K. Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist Updat. 2008;11(4–5):164–79. doi: 10.1016/j.drup.2008.08.002. [DOI] [PubMed] [Google Scholar]
- 4.Manach C, Scalbert A, Morand C, Remesy C, Jimenez L. Polyphenols: food sources and bioavailability. Am J Clin Nutr. 2004;79(5):727–47. doi: 10.1093/ajcn/79.5.727. [DOI] [PubMed] [Google Scholar]
- 5.Ross JA, Kasum CM. Dietary flavonoids: bioavailability, metabolic effects, and safety. Annu Rev Nutr. 2002;22:19–34. doi: 10.1146/annurev.nutr.22.111401.144957. [DOI] [PubMed] [Google Scholar]
- 6.Rossi L, Mazzitelli S, Arciello M, Capo CR, Rotilio G. Benefits from dietary polyphenols for brain aging and Alzheimer’s disease. Neurochem Res. 2008;33(12):2390–400. doi: 10.1007/s11064-008-9696-7. [DOI] [PubMed] [Google Scholar]
- 7.Narayana KR, Reddy MS, Chaluvadi M, Krishna D. Bioflavonoids classification, pharmacological, biochemical effects and therapeutic potential. Indian Journal of Pharmacology. 2001;33(1):2–16. [Google Scholar]
- 8.D’Archivio M, Filesi C, Di Benedetto R, Gargiulo R, Giovannini C, Masella R. Polyphenols, dietary sources and bioavailability. Ann Ist Super Sanita. 2007;43(4):348–61. [PubMed] [Google Scholar]
- 9.Aggarwal BB, Takada Y, Oommen OV. From chemoprevention to chemotherapy: common targets and common goals. Expert Opin Investig Drugs. 2004;13(10):1327–38. doi: 10.1517/13543784.13.10.1327. [DOI] [PubMed] [Google Scholar]
- 10.Adams J. The proteasome: a suitable antineoplastic target. Nat Rev Cancer. 2004;4(5):349–60. doi: 10.1038/nrc1361. [DOI] [PubMed] [Google Scholar]
- 11.Kisselev AF, Songyang Z, Goldberg AL. Why does threonine, and not serine, function as the active site nucleophile in proteasomes? J Biol Chem. 2000;275(20):14831–7. doi: 10.1074/jbc.275.20.14831. [DOI] [PubMed] [Google Scholar]
- 12.Moore BS, Eustaquio AS, McGlinchey RP. Advances in and applications of proteasome inhibitors. Curr Opin Chem Biol. 2008;12(4):434–40. doi: 10.1016/j.cbpa.2008.06.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Borissenko L, Groll M. 20S proteasome and its inhibitors: crystallographic knowledge for drug development. Chem Rev. 2007;107(3):687–717. doi: 10.1021/cr0502504. [DOI] [PubMed] [Google Scholar]
- 14.Jesenberger V, Jentsch S. Deadly encounter: ubiquitin meets apoptosis. Nat Rev Mol Cell Biol. 2002;3(2):112–21. doi: 10.1038/nrm731. [DOI] [PubMed] [Google Scholar]
- 15.Voorhees PM, Orlowski RZ. The proteasome and proteasome inhibitors in cancer therapy. Annu Rev Pharmacol Toxicol. 2006;46:189–213. doi: 10.1146/annurev.pharmtox.46.120604.141300. [DOI] [PubMed] [Google Scholar]
- 16.Dou QP, Landis-Piwowar KR, Chen D, Huo C, Wan SB, Chan TH. Green tea polyphenols as a natural tumour cell proteasome inhibitor. Inflammopharmacology. 2008;16(5):208–12. doi: 10.1007/s10787-008-8017-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nam S, Smith DM, Dou QP. Ester bond-containing tea polyphenols potently inhibit proteasome activity in vitro and in vivo. J Biol Chem. 2001;276(16):13322–30. doi: 10.1074/jbc.M004209200. [DOI] [PubMed] [Google Scholar]
- 18.Smith DM, Wang Z, Kazi A, Li LH, Chan TH, Dou QP. Synthetic analogs of green tea polyphenols as proteasome inhibitors. Mol Med. 2002;8(7):382–92. [PMC free article] [PubMed] [Google Scholar]
- 19.Mozzicafreddo M, Cuccioloni M, Cecarini V, Eleuteri AM, Angeletti M. Homology modeling and docking analysis of the interaction between polyphenols and mammalian 20S proteasomes. J Chem Inf Model. 2009;49(2):401–9. doi: 10.1021/ci800235m. [DOI] [PubMed] [Google Scholar]
- 20.Smith DM, Daniel KG, Wang Z, Guida WC, Chan TH, Dou QP. Docking studies and model development of tea polyphenol proteasome inhibitors: applications to rational drug design. Proteins. 2004;54(1):58–70. doi: 10.1002/prot.10504. [DOI] [PubMed] [Google Scholar]
- 21.Wan SB, Landis-Piwowar KR, Kuhn DJ, Chen D, Dou QP, Chan TH. Structure-activity study of epi-gallocatechin gallate (EGCG) analogs as proteasome inhibitors. Bioorg Med Chem. 2005;13(6):2177–85. doi: 10.1016/j.bmc.2004.12.056. [DOI] [PubMed] [Google Scholar]
- 22.Sang S, Lambert JD, Yang CS. Bioavailability and stability issues in understanding the cancer preventive effects of tea polyphenols. Journal of the Science of Food and Agriculture. 2006;86(14):2256–2265. [Google Scholar]
- 23.Lam WH, Kazi A, Kuhn DJ, Chow LM, Chan AS, Dou QP, Chan TH. A potential prodrug for a green tea polyphenol proteasome inhibitor: evaluation of the peracetate ester of (−)-epigallocatechin gallate [(−)-EGCG] Bioorg Med Chem. 2004;12(21):5587–93. doi: 10.1016/j.bmc.2004.08.002. [DOI] [PubMed] [Google Scholar]
- 24.Kuhn D, Lam WH, Kazi A, Daniel KG, Song S, Chow LM, Chan TH, Dou QP. Synthetic peracetate tea polyphenols as potent proteasome inhibitors and apoptosis inducers in human cancer cells. Front Biosci. 2005;10:1010–23. doi: 10.2741/1595. [DOI] [PubMed] [Google Scholar]
- 25.Osanai K, Landis-Piwowar KR, Dou QP, Chan TH. A para-amino substituent on the D-ring of green tea polyphenol epigallocatechin-3-gallate as a novel proteasome inhibitor and cancer cell apoptosis inducer. Bioorg Med Chem. 2007;15(15):5076–82. doi: 10.1016/j.bmc.2007.05.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yu Z, Qin XL, Gu YY, Chen D, Cui QC, Jiang T, Wan SB, Dou QP. Prodrugs of fluoro-substituted benzoates of EGC as tumor cellular proteasome inhibitors and apoptosis inducers. Int J Mol Sci. 2008;9(6):951–61. doi: 10.3390/ijms9060951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bonfili L, Cuccioloni M, Mozzicafreddo M, Cecarini V, Angeletti M, Eleuteri AM. Identification of an EGCG oxidation derivative with proteasome modulatory activity. Biochimie. 2011;93(5):931–40. doi: 10.1016/j.biochi.2011.02.003. [DOI] [PubMed] [Google Scholar]
- 28.Lu H, Meng X, Yang CS. Enzymology of methylation of tea catechins and inhibition of catechol-O-methyltransferase by (−)-epigallocatechin gallate. Drug Metab Dispos. 2003;31(5):572–9. doi: 10.1124/dmd.31.5.572. [DOI] [PubMed] [Google Scholar]
- 29.Landis-Piwowar KR, Wan SB, Wiegand RA, Kuhn DJ, Chan TH, Dou QP. Methylation suppresses the proteasome-inhibitory function of green tea polyphenols. J Cell Physiol. 2007;213(1):252–60. doi: 10.1002/jcp.21124. [DOI] [PubMed] [Google Scholar]
- 30.Huo C, Yang H, Cui QC, Dou QP, Chan TH. Proteasome inhibition in human breast cancer cells with high catechol-O-methyltransferase activity by green tea polyphenol EGCG analogs. Bioorg Med Chem. 2010;18(3):1252–8. doi: 10.1016/j.bmc.2009.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Landis-Piwowar K, Chen D, Chan TH, Dou QP. Inhibition of catechol-Omicron-methyltransferase activity in human breast cancer cells enhances the biological effect of the green tea polyphenol (−)-EGCG. Oncol Rep. 2010;24(2):563–9. doi: 10.3892/or_00000893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Golden EB, Lam PY, Kardosh A, Gaffney KJ, Cadenas E, Louie SG, Petasis NA, Chen TC, Schonthal AH. Green tea polyphenols block the anticancer effects of bortezomib and other boronic acid-based proteasome inhibitors. Blood. 2009;113(23):5927–37. doi: 10.1182/blood-2008-07-171389. [DOI] [PubMed] [Google Scholar]
- 33.Wang Q, Li J, Gu J, Huang B, Zhao Y, Zheng D, Ding Y, Zeng L. Potentiation of (−)-epigallocatechin-3-gallate-induced apoptosis by bortezomib in multiple myeloma cells. Acta Biochim Biophys Sin (Shanghai) 2009;41(12):1018–26. doi: 10.1093/abbs/gmp094. [DOI] [PubMed] [Google Scholar]
- 34.Bannerman B, Xu L, Jones M, Tsu C, Yu J, Hales P, Monbaliu J, Fleming P, Dick L, Manfredi M, Claiborne C, Bolen J, Kupperman E, Berger A. Preclinical evaluation of the antitumor activity of bortezomib in combination with vitamin C or with epigallocatechin gallate, a component of green tea. Cancer Chemother Pharmacol. 2011 doi: 10.1007/s00280-011-1591-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kazi A, Daniel KG, Smith DM, Kumar NB, Dou QP. Inhibition of the proteasome activity, a novel mechanism associated with the tumor cell apoptosis-inducing ability of genistein. Biochem Pharmacol. 2003;66(6):965–76. doi: 10.1016/s0006-2952(03)00414-3. [DOI] [PubMed] [Google Scholar]
- 36.Chen D, Chen MS, Cui QC, Yang H, Dou QP. Structure-proteasome-inhibitory activity relationships of dietary flavonoids in human cancer cells. Front Biosci. 2007;12:1935–45. doi: 10.2741/2199. [DOI] [PubMed] [Google Scholar]
- 37.Chen D, Landis-Piwowar KR, Chen MS, Dou QP. Inhibition of proteasome activity by the dietary flavonoid apigenin is associated with growth inhibition in cultured breast cancer cells and xenografts. Breast Cancer Res. 2007;9(6):R80. doi: 10.1186/bcr1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chen D, Daniel KG, Chen MS, Kuhn DJ, Landis-Piwowar KR, Dou QP. Dietary flavonoids as proteasome inhibitors and apoptosis inducers in human leukemia cells. Biochem Pharmacol. 2005;69(10):1421–32. doi: 10.1016/j.bcp.2005.02.022. [DOI] [PubMed] [Google Scholar]
- 39.van Duursen MB, Sanderson JT, de Jong PC, Kraaij M, van den Berg M. Phytochemicals inhibit catechol-O-methyltransferase activity in cytosolic fractions from healthy human mammary tissues: implications for catechol estrogen-induced DNA damage. Toxicol Sci. 2004;81(2):316–24. doi: 10.1093/toxsci/kfh216. [DOI] [PubMed] [Google Scholar]
- 40.Chen D, Wang CY, Lambert JD, Ai N, Welsh WJ, Yang CS. Inhibition of human liver catechol-O-methyltransferase by tea catechins and their metabolites: structure-activity relationship and molecular-modeling studies. Biochem Pharmacol. 2005;69(10):1523–31. doi: 10.1016/j.bcp.2005.01.024. [DOI] [PubMed] [Google Scholar]
- 41.Landis-Piwowar KR, Milacic V, Dou QP. Relationship between the methylation status of dietary flavonoids and their growth-inhibitory and apoptosis-inducing activities in human cancer cells. J Cell Biochem. 2008;105(2):514–23. doi: 10.1002/jcb.21853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liu FT, Agrawal SG, Movasaghi Z, Wyatt PB, Rehman IU, Gribben JG, Newland AC, Jia L. Dietary flavonoids inhibit the anticancer effects of the proteasome inhibitor bortezomib. Blood. 2008;112(9):3835–46. doi: 10.1182/blood-2008-04-150227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jana NR, Dikshit P, Goswami A, Nukina N. Inhibition of proteasomal function by curcumin induces apoptosis through mitochondrial pathway. J Biol Chem. 2004;279(12):11680–5. doi: 10.1074/jbc.M310369200. [DOI] [PubMed] [Google Scholar]
- 44.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(18):7283–92. doi: 10.1158/0008-5472.CAN-07-6246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wan SB, Yang H, Zhou Z, Cui QC, Chen D, Kanwar J, Mohammad I, Dou QP, Chan TH. Evaluation of curcumin acetates and amino acid conjugates as proteasome inhibitors. Int J Mol Med. 2010;26(4):447–55. doi: 10.3892/ijmm_00000484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sung B, Kunnumakkara AB, Sethi G, Anand P, Guha S, Aggarwal BB. Curcumin circumvents chemoresistance in vitro and potentiates the effect of thalidomide and bortezomib against human multiple myeloma in nude mice model. Mol Cancer Ther. 2009;8(4):959–70. doi: 10.1158/1535-7163.MCT-08-0905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nam S, Smith DM, Dou QP. Tannic acid potently inhibits tumor cell proteasome activity, increases p27 and Bax expression, and induces G1 arrest and apoptosis. Cancer Epidemiol Biomarkers Prev. 2001;10(10):1083–8. [PubMed] [Google Scholar]
- 48.Marienfeld C, Tadlock L, Yamagiwa Y, Patel T. Inhibition of cholangiocarcinoma growth by tannic acid. Hepatology. 2003;37(5):1097–104. doi: 10.1053/jhep.2003.50192. [DOI] [PubMed] [Google Scholar]
- 49.Longley DB, Johnston PG. Molecular mechanisms of drug resistance. J Pathol. 2005;205(2):275–92. doi: 10.1002/path.1706. [DOI] [PubMed] [Google Scholar]
- 50.Cusack JC, Jr, Liu R, Xia L, Chao TH, Pien C, Niu W, Palombella VJ, Neuteboom ST, Palladino MA. NPI-0052 enhances tumoricidal response to conventional cancer therapy in a colon cancer model. Clin Cancer Res. 2006;12(22):6758–64. doi: 10.1158/1078-0432.CCR-06-1151. [DOI] [PubMed] [Google Scholar]
- 51.Fennell DA, Chacko A, Mutti L. BCL-2 family regulation by the 20S proteasome inhibitor bortezomib. Oncogene. 2008;27(9):1189–97. doi: 10.1038/sj.onc.1210744. [DOI] [PubMed] [Google Scholar]
- 52.Zhang Y, Shi Y, Li X, Du R, Luo G, Xia L, Du W, Chen B, Zhai H, Wu K, Fan D. Proteasome inhibitor MG132 reverses multidrug resistance of gastric cancer through enhancing apoptosis and inhibiting P-gp. Cancer Biol Ther. 2008;7(4):540–6. doi: 10.4161/cbt.7.4.5483. [DOI] [PubMed] [Google Scholar]
- 53.Li B, Dou QP. Bax degradation by the ubiquitin/proteasome-dependent pathway: involvement in tumor survival and progression. Proc Natl Acad Sci U S A. 2000;97(8):3850–5. doi: 10.1073/pnas.070047997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dean M. ABC transporters, drug resistance, and cancer stem cells. J Mammary Gland Biol Neoplasia. 2009;14(1):3–9. doi: 10.1007/s10911-009-9109-9. [DOI] [PubMed] [Google Scholar]
- 55.Choudhuri S, Klaassen CD. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int J Toxicol. 2006;25(4):231–59. doi: 10.1080/10915810600746023. [DOI] [PubMed] [Google Scholar]
- 56.Loo TW, Clarke DM. The human multidrug resistance P-glycoprotein is inactive when its maturation is inhibited: potential for a role in cancer chemotherapy. FASEB J. 1999;13(13):1724–32. doi: 10.1096/fasebj.13.13.1724. [DOI] [PubMed] [Google Scholar]
- 57.Fujita T, Washio K, Takabatake D, Takahashi H, Yoshitomi S, Tsukuda K, Ishibe Y, Ogasawara Y, Doihara H, Shimizu N. Proteasome inhibitors can alter the signaling pathways and attenuate the P-glycoprotein-mediated multidrug resistance. Int J Cancer. 2005;117(4):670–82. doi: 10.1002/ijc.21063. [DOI] [PubMed] [Google Scholar]
- 58.Lee MH, Hyun DH, Jenner P, Halliwell B. Effect of proteasome inhibition on cellular oxidative damage, antioxidant defences and nitric oxide production. J Neurochem. 2001;78(1):32–41. doi: 10.1046/j.1471-4159.2001.00416.x. [DOI] [PubMed] [Google Scholar]
- 59.Lev N, Melamed E, Offen D. Proteasomal inhibition hypersensitizes differentiated neuroblastoma cells to oxidative damage. Neurosci Lett. 2006;399(1–2):27–32. doi: 10.1016/j.neulet.2005.09.086. [DOI] [PubMed] [Google Scholar]
- 60.Motegi A, Murakawa Y, Takeda S. The vital link between the ubiquitin-proteasome pathway and DNA repair: impact on cancer therapy. Cancer Lett. 2009;283(1):1–9. doi: 10.1016/j.canlet.2008.12.030. [DOI] [PubMed] [Google Scholar]
- 61.Wang QE, Wani MA, Chen J, Zhu Q, Wani G, El-Mahdy MA, Wani AA. Cellular ubiquitination and proteasomal functions positively modulate mammalian nucleotide excision repair. Mol Carcinog. 2005;42(1):53–64. doi: 10.1002/mc.20065. [DOI] [PubMed] [Google Scholar]
- 62.Podlaska A, McIntyre J, Skoneczna A, Sledziewska-Gojska E. The link between 20S proteasome activity and post-replication DNA repair in Saccharomyces cerevisiae. Mol Microbiol. 2003;49(5):1321–32. doi: 10.1046/j.1365-2958.2003.03635.x. [DOI] [PubMed] [Google Scholar]
- 63.Murakawa Y, Sonoda E, Barber LJ, Zeng W, Yokomori K, Kimura H, Niimi A, Lehmann A, Zhao GY, Hochegger H, Boulton SJ, Takeda S. Inhibitors of the proteasome suppress homologous DNA recombination in mammalian cells. Cancer Res. 2007;67(18):8536–43. doi: 10.1158/0008-5472.CAN-07-1166. [DOI] [PubMed] [Google Scholar]
- 64.Jacquemont C, Taniguchi T. Proteasome function is required for DNA damage response and fanconi anemia pathway activation. Cancer Res. 2007;67(15):7395–405. doi: 10.1158/0008-5472.CAN-07-1015. [DOI] [PubMed] [Google Scholar]
- 65.Mimnaugh EG, Yunmbam MK, Li Q, Bonvini P, Hwang SG, Trepel J, Reed E, Neckers L. Prevention of cisplatin-DNA adduct repair and potentiation of cisplatin-induced apoptosis in ovarian carcinoma cells by proteasome inhibitors. Biochem Pharmacol. 2000;60(9):1343–54. doi: 10.1016/s0006-2952(00)00455-x. [DOI] [PubMed] [Google Scholar]
- 66.Kitagawa S, Nabekura T, Kamiyama S. Inhibition of P-glycoprotein function by tea catechins in KB-C2 cells. J Pharm Pharmacol. 2004;56(8):1001–5. doi: 10.1211/0022357044003. [DOI] [PubMed] [Google Scholar]
- 67.Jodoin J, Demeule M, Beliveau R. Inhibition of the multidrug resistance P-glycoprotein activity by green tea polyphenols. Biochim Biophys Acta. 2002;1542(1–3):149–59. doi: 10.1016/s0167-4889(01)00175-6. [DOI] [PubMed] [Google Scholar]
- 68.Farabegoli F, Papi A, Bartolini G, Ostan R, Orlandi M. (−)-Epigallocatechin-3-gallate downregulates Pg-P BCRP in a tamoxifen resistant MCF-7 cell line. Phytomedicine. 2010;17(5):356–62. doi: 10.1016/j.phymed.2010.01.001. [DOI] [PubMed] [Google Scholar]
- 69.Mei Y, Qian F, Wei D, Liu J. Reversal of cancer multidrug resistance by green tea polyphenols. J Pharm Pharmacol. 2004;56(10):1307–14. doi: 10.1211/0022357044364. [DOI] [PubMed] [Google Scholar]
- 70.Qian F, Wei D, Zhang Q, Yang S. Modulation of P-glycoprotein function and reversal of multidrug resistance by (−)-epigallocatechin gallate in human cancer cells. Biomed Pharmacother. 2005;59(3):64–9. doi: 10.1016/j.biopha.2005.01.002. [DOI] [PubMed] [Google Scholar]
- 71.Liang G, Tang A, Lin X, Li L, Zhang S, Huang Z, Tang H, Li QQ. Green tea catechins augment the antitumor activity of doxorubicin in an in vivo mouse model for chemoresistant liver cancer. Int J Oncol. 2010;37(1):111–23. [PubMed] [Google Scholar]
- 72.Chen TC, Wang W, Golden EB, Thomas S, Sivakumar W, Hofman FM, Louie SG, Schonthal AH. Green tea epigallocatechin gallate enhances therapeutic efficacy of temozolomide in orthotopic mouse glioblastoma models. Cancer Lett. 2011;302(2):100–8. doi: 10.1016/j.canlet.2010.11.008. [DOI] [PubMed] [Google Scholar]
- 73.Stearns ME, Amatangelo MD, Varma D, Sell C, Goodyear SM. Combination therapy with epigallocatechin-3-gallate and doxorubicin in human prostate tumor modeling studies: inhibition of metastatic tumor growth in severe combined immunodeficiency mice. Am J Pathol. 2010;177(6):3169–79. doi: 10.2353/ajpath.2010.100330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Luo T, Wang J, Yin Y, Hua H, Jing J, Sun X, Li M, Zhang Y, Jiang Y. (−)-Epigallocatechin gallate sensitizes breast cancer cells to paclitaxel in a murine model of breast carcinoma. Breast Cancer Res. 2010;12(1):R8. doi: 10.1186/bcr2473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Davenport A, Frezza M, Shen M, Ge Y, Huo C, Chan TH, Dou QP. Celastrol and an EGCG pro-drug exhibit potent chemosensitizing activity in human leukemia cells. Int J Mol Med. 2010;25(3):465–70. doi: 10.3892/ijmm_00000366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Singh M, Bhatnagar P, Srivastava AK, Kumar P, Shukla Y, Gupta KC. Enhancement of cancer chemosensitization potential of cisplatin by tea polyphenols poly(lactide-co-glycolide) nanoparticles. J Biomed Nanotechnol. 2011;7(1):202. doi: 10.1166/jbn.2011.1268. [DOI] [PubMed] [Google Scholar]
- 77.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(15):6934–42. doi: 10.1158/0008-5472.CAN-04-4604. [DOI] [PubMed] [Google Scholar]
- 78.Solomon LA, Ali S, Banerjee S, Munkarah AR, Morris RT, Sarkar FH. Sensitization of ovarian cancer cells to cisplatin by genistein: the role of NF-kappaB. J Ovarian Res. 2008;1(1):9. doi: 10.1186/1757-2215-1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Banerjee S, Zhang Y, Ali S, Bhuiyan M, Wang Z, Chiao PJ, Philip PA, Abbruzzese J, Sarkar FH. 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(19):9064–72. doi: 10.1158/0008-5472.CAN-05-1330. [DOI] [PubMed] [Google Scholar]
- 80.Ma Y, Wang J, Liu L, Zhu H, Chen X, Pan S, Sun X, Jiang H. Genistein potentiates the effect of arsenic trioxide against human hepatocellular carcinoma: role of Akt and nuclear factor-kappaB. Cancer Lett. 2011;301(1):75–84. doi: 10.1016/j.canlet.2010.10.022. [DOI] [PubMed] [Google Scholar]
- 81.Banerjee S, Kong D, Azmi AS, Wang Z, Ahmad A, Sethi S, Sarkar FH. Restoring sensitivity to oxaliplatin by a novel approach in gemcitabine-resistant pancreatic cancer cells in vitro and in vivo. Int J Cancer. 2011;128(5):1240–50. doi: 10.1002/ijc.25658. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 82.Zhang B, Shi ZL, Liu B, Yan XB, Feng J, Tao HM. Enhanced anticancer effect of gemcitabine by genistein in osteosarcoma: the role of Akt and nuclear factor-kappaB. Anticancer Drugs. 2010;21(3):288–96. doi: 10.1097/CAD.0b013e328334da17. [DOI] [PubMed] [Google Scholar]
- 83.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(3):123–31. doi: 10.1159/000028993. [DOI] [PubMed] [Google Scholar]
- 84.Shen J, Tai YC, Zhou J, Stephen Wong CH, Cheang PT, Fred Wong WS, Xie Z, Khan M, Han JH, Chen CS. Synergistic antileukemia effect of genistein and chemotherapy in mouse xenograft model and potential mechanism through MAPK signaling. Exp Hematol. 2007;35(1):75–83. doi: 10.1016/j.exphem.2006.09.007. [DOI] [PubMed] [Google Scholar]
- 85.Goel A, Aggarwal BB. Curcumin, the golden spice from Indian saffron, is a chemosensitizer and radiosensitizer for tumors and chemoprotector and radioprotector for normal organs. Nutr Cancer. 2010;62(7):919–30. doi: 10.1080/01635581.2010.509835. [DOI] [PubMed] [Google Scholar]
- 86.Anuchapreeda S, Leechanachai P, Smith MM, Ambudkar SV, Limtrakul PN. Modulation of P-glycoprotein expression and function by curcumin in multidrug-resistant human KB cells. Biochem Pharmacol. 2002;64(4):573–82. doi: 10.1016/s0006-2952(02)01224-8. [DOI] [PubMed] [Google Scholar]
- 87.Tang XQ, Bi H, Feng JQ, Cao JG. Effect of curcumin on multidrug resistance in resistant human gastric carcinoma cell line SGC7901/VCR. Acta Pharmacol Sin. 2005;26(8):1009–16. doi: 10.1111/j.1745-7254.2005.00149.x. [DOI] [PubMed] [Google Scholar]
- 88.Chearwae W, Shukla S, Limtrakul P, Ambudkar SV. Modulation of the function of the multidrug resistance-linked ATP-binding cassette transporter ABCG2 by the cancer chemopreventive agent curcumin. Mol Cancer Ther. 2006;5(8):1995–2006. doi: 10.1158/1535-7163.MCT-06-0087. [DOI] [PubMed] [Google Scholar]
- 89.Shukla S, Zaher H, Hartz A, Bauer B, Ware JA, Ambudkar SV. Curcumin inhibits the activity of ABCG2/BCRP1, a multidrug resistance-linked ABC drug transporter in mice. Pharm Res. 2009;26(2):480–7. doi: 10.1007/s11095-008-9735-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Limtrakul P, Chearwae W, Shukla S, Phisalphong C, Ambudkar SV. Modulation of function of three ABC drug transporters, P-glycoprotein (ABCB1), mitoxantrone resistance protein (ABCG2) and multidrug resistance protein 1 (ABCC1) by tetrahydrocurcumin, a major metabolite of curcumin. Mol Cell Biochem. 2007;296(1–2):85–95. doi: 10.1007/s11010-006-9302-8. [DOI] [PubMed] [Google Scholar]
- 91.Dhandapani KM, Mahesh VB, Brann DW. Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFkappaB transcription factors. J Neurochem. 2007;102(2):522–38. doi: 10.1111/j.1471-4159.2007.04633.x. [DOI] [PubMed] [Google Scholar]
- 92.Kang HJ, Lee SH, Price JE, Kim LS. Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB in breast cancer cells and potentiates the growth inhibitory effect of paclitaxel in a breast cancer nude mice model. Breast J. 2009;15(3):223–9. doi: 10.1111/j.1524-4741.2009.00709.x. [DOI] [PubMed] [Google Scholar]
- 93.Duarte VM, Han E, Veena MS, Salvado A, Suh JD, Liang LJ, Faull KF, Srivatsan ES, Wang MB. Curcumin enhances the effect of cisplatin in suppression of head and neck squamous cell carcinoma via inhibition of IKKbeta protein of the NFkappaB pathway. Mol Cancer Ther. 2010;9(10):2665–75. doi: 10.1158/1535-7163.MCT-10-0064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yoon MJ, Kim EH, Lim JH, Kwon TK, Choi KS. Superoxide anion and proteasomal dysfunction contribute to curcumin-induced paraptosis of malignant breast cancer cells. Free Radic Biol Med. 2010;48(5):713–26. doi: 10.1016/j.freeradbiomed.2009.12.016. [DOI] [PubMed] [Google Scholar]
- 95.Laurie SA, Miller VA, Grant SC, Kris MG, Ng KK. Phase I study of green tea extract in patients with advanced lung cancer. Cancer Chemother Pharmacol. 2005;55(1):33–8. doi: 10.1007/s00280-004-0859-1. [DOI] [PubMed] [Google Scholar]
- 96.Jatoi A, Ellison N, Burch PA, Sloan JA, Dakhil SR, Novotny P, Tan W, Fitch TR, Rowland KM, Young CY, Flynn PJ. A phase II trial of green tea in the treatment of patients with androgen independent metastatic prostate carcinoma. Cancer. 2003;97(6):1442–6. doi: 10.1002/cncr.11200. [DOI] [PubMed] [Google Scholar]
- 97.Choan E, Segal R, Jonker D, Malone S, Reaume N, Eapen L, Gallant V. A prospective clinical trial of green tea for hormone refractory prostate cancer: an evaluation of the complementary/alternative therapy approach. Urol Oncol. 2005;23(2):108–13. doi: 10.1016/j.urolonc.2004.10.008. [DOI] [PubMed] [Google Scholar]
- 98.Landis-Piwowar KR, Huo C, Chen D, Milacic V, Shi G, Chan TH, Dou QP. A novel prodrug of the green tea polyphenol (−)-epigallocatechin-3-gallate as a potential anticancer agent. Cancer Res. 2007;67(9):4303–10. doi: 10.1158/0008-5472.CAN-06-4699. [DOI] [PubMed] [Google Scholar]
- 99.Yang H, Sun DK, Chen D, Cui QC, Gu YY, Jiang T, Chen W, Wan SB, Dou QP. Antitumor activity of novel fluoro-substituted (−)-epigallocatechin-3-gallate analogs. Cancer Lett. 2010;292(1):48–53. doi: 10.1016/j.canlet.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lee SC, Chan WK, Lee TW, Lam WH, Wang X, Chan TH, Wong YC. Effect of a prodrug of the green tea polyphenol (−)-epigallocatechin-3-gallate on the growth of androgen-independent prostate cancer in vivo. Nutr Cancer. 2008;60(4):483–91. doi: 10.1080/01635580801947674. [DOI] [PubMed] [Google Scholar]
- 101.Shanafelt TD, Call TG, Zent CS, LaPlant B, Bowen DA, Roos M, Secreto CR, Ghosh AK, Kabat BF, Lee MJ, Yang CS, Jelinek DF, Erlichman C, Kay NE. Phase I trial of daily oral Polyphenon E in patients with asymptomatic Rai stage 0 to II chronic lymphocytic leukemia. J Clin Oncol. 2009;27(23):3808–14. doi: 10.1200/JCO.2008.21.1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.El-Rayes B, Philip P, Sarkar F, Shields A, Ferris A, Hess K, Kaseb A, Javle M, Varadhachary G, Wolff R, Abbruzzese J. A phase II study of isoflavones, erlotinib, and gemcitabine in advanced pancreatic cancer. Investigational New Drugs. 2010:1–6. doi: 10.1007/s10637-010-9386-6. [DOI] [PubMed] [Google Scholar]
- 103.Silasi DA, Alvero AB, Rutherford TJ, Brown D, Mor G. Phenoxodiol: pharmacology and clinical experience in cancer monotherapy and in combination with chemotherapeutic drugs. Expert Opin Pharmacother. 2009;10(6):1059–67. doi: 10.1517/14656560902837980. [DOI] [PubMed] [Google Scholar]
- 104.Kelly MG, Mor G, Husband A, O’Malley DM, Baker L, Azodi M, Schwartz PE, Rutherford TJ. Phase II Evaluation of Phenoxodiol in Combination With Cisplatin or Paclitaxel in Women With Platinum/Taxane-Refractory/Resistant Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancers. Int J Gynecol Cancer. 2011 doi: 10.1097/IGC.0b013e3182126f05. [DOI] [PubMed] [Google Scholar]
- 105.Kanai M, Yoshimura K, Asada M, Imaizumi A, Suzuki C, Matsumoto S, Nishimura T, Mori Y, Masui T, Kawaguchi Y, Yanagihara K, Yazumi S, Chiba T, Guha S, Aggarwal B. A phase I/II study of gemcitabine-based chemotherapy plus curcumin for patients with gemcitabine-resistant pancreatic cancer. Cancer Chemotherapy and Pharmacology. 2010:1–8. doi: 10.1007/s00280-010-1470-2. [DOI] [PubMed] [Google Scholar]
- 106.Bayet-Robert M, Kwiatkowski F, Leheurteur M, Gachon F, Planchat E, Abrial C, Mouret-Reynier MA, Durando X, Barthomeuf C, Chollet P. Phase I dose escalation trial of docetaxel plus curcumin in patients with advanced and metastatic breast cancer. Cancer Biol Ther. 2010;9(1):8–14. doi: 10.4161/cbt.9.1.10392. [DOI] [PubMed] [Google Scholar]
- 107.Shim SH. 20S proteasome inhibitory activity of flavonoids isolated from Spatholobus suberectus. Phytother Res. 2011;25(4):615–8. doi: 10.1002/ptr.3342. [DOI] [PubMed] [Google Scholar]