You are on page 1of 12

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/51019274

Curcumin and Liver Cancer: A Review

Article  in  Current Pharmaceutical Biotechnology · April 2011


DOI: 10.2174/138920112798868791 · Source: PubMed

CITATIONS READS
135 4,411

3 authors, including:

Bharat Aggarwal Anupam Bishayee


University of Texas MD Anderson Cancer Center Lake Erie College of Osteopathic Medicine Bradenton FL USA
731 PUBLICATIONS   98,410 CITATIONS    220 PUBLICATIONS   7,212 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Journal of Research and Pharmacy Practice View project

Natural compounds with anti-inflammatory and anticancer activity View project

All content following this page was uploaded by Bharat Aggarwal on 05 September 2014.

The user has requested enhancement of the downloaded file.


218 Current Pharmaceutical Biotechnology, 2012, 13, 218-228

Curcumin and Liver Cancer: A Review

Altaf S. Darvesha, Bharat B. Aggarwalb and Anupam Bishayeea,c,*

a
Cancer Therapeutics and Chemoprevention Group, Department of Pharmaceutical Sciences, College of Pharmacy,
Northeast Ohio Medical University, Rootstown, OH 44272, USA; bCytokine Research Laboratory, Department of Ex-
perimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; cDepart-
ment of Internal Medicine, College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA

Abstract: Primary liver cancer, also known as hepatocellular carcinoma (HCC), is one of the most lethal cancers having
worldwide prevalence. Although most HCC cases are reported in the developing countries of Asia and Africa, there has
been an alarming increase in HCC cases in Western Europe as well as United States. Chronic liver diseases, viral hepati-
tis, alcoholism as well as dietary carcinogens, such as aflatoxins and nitrosoamines, contribute to HCC. Liver transplanta-
tion as well as surgical resection at best offer limited treatment options. Thus, there exists a critical need to investigate and
evaluate possible alternative chemopreventive and therapeutic strategies which may be effective in the control of liver
cancer. HCC, most often, develops and progresses in a milieu of oxidative stress and inflammation. Phytochemicals, such
as dietary polyphenols endowed with potent antioxidant as well as anti-inflammatory properties, provide a suitable alter-
native in affording alleviation of HCC. Curcumin, the principal polyphenolic curcuminoid, obtained from the turmeric
rhizome Curcuma longa has long been used to cure several chronic ailments, such as neoplastic and neurodegenerative
diseases. Studies suggest that curcumin may have antitumor, antioxidant, and anti-inflammatory properties. This article
reviews the effects of curcumin in preclinical in vitro and in vivo models of HCC with particular emphasis to its antioxi-
dant, apoptotic and anti-inflammatory effects as well as involvement in various molecular signaling mechanisms. This re-
view also discusses potential challenges involved in the use of curcumin in HCC, such as bioavailability, pharmacokinet-
ics, drug delivery as well as paucity of clinical studies.
Keywords: Chemoprevention, curcumin, inflammation, liver cancer, oxidative stress, therapy.

INTRODUCTION treatment options for HCC. Several factors, such as tumor


size, multifocality along with vascular invasion limit the
Primary liver cancer, essentially hepatocellular carci-
option of surgical resection in only about 20% of HCC pa-
noma (HCC), remains the fifth largest cause of cancer in tients. The recurrence rates of post-surgery HCC are also as
men and the eighth largest in women [1-3]. HCC remains the
high as 50% [14]. Although, liver transplantation is useful in
third leading cause of cancer-related deaths worldwide with
treatment of early-stage HCC, it is available only to a small
about 700,000 deaths reported annually [4, 5]. HCC is a ma-
number of HCC patients primarily due to scarcity of donor
lignancy with extremely grim prospects and a five-year sur-
organs. The effectiveness of the transplant option is also se-
vival rate reported below 9% [6]. HCC incidence has shown
verely limited due to the swift and frequent reappearance of
a precipitous increase worldwide especially in the develop- HCC in the transplanted liver [11, 15]. Several alternative
ing countries of Asia, such as China, as well as sub-Saharan
treatment approaches, such as Yttrium-90 intra-arterial de-
Africa. An alarming increase in HCC cases has also been
livery as microspheres, arterial chemoembolization, micro-
reported in parts of Central and Southern Europe as well as
wave coagulation, intra-tumor ethanol injection as well as
the North American continent [7, 8]. The past three decades
radiofrequency ablation, have limited usefulness, being ap-
has witnessed a dramatic increase in the occurrence of hu-
plicable only to patients with localized liver tumors [11, 16-
man HCC in the United States. Current statistics predict ap- 20]. Sorafenib, a vascular endothelial growth factor receptor
proximately 25,000 new cases and 19,000 deaths due to liver
and tyrosine kinase inhibitor, is currently approved in the
cancer in 2010 in this country [9]. Although, the majority of
United States for the treatment of unresectable HCC.
HCC cases are a result of infections due to the hepatitis B
Although sorafenib has been shown to prolong the medial
and C viruses, risk factors such as obesity, iron overload,
survival time by almost three months in patients with ad-
both alcoholic and non-alcoholic cirrhosis, as well as dietary
vanced HCC, its therapeutic usefulness is limited due to se-
hepatocarcinogenes, such as aflatoxins and nitrosoamines, vere adverse effects including a considerable risk of hemor-
have also been implicated as important key causes of HCC
rhage [21, 22] as well as unprecedented high cost [23]. In
[5, 10-13]. Both surgery and liver transplant offer limited
view of the aforementioned limitations of the current treat-
ment options available for HCC, novel approaches, espe-
*Address correspondence to this author at the Department of Pharmaceuti- cially chemoprevention, has been suggested as the para-
cal and Administrative Sciences, School of Pharmacy, 1600 East Hill Street, mount approach in lowering the occurrence and mortality
Signal Hill, CA 90755, USA; Tel: +562 988 2278, Ext. 2038; Fax: +562 988 associated with HCC [24, 25].
1791; E-mail: abishayee@auhs.edu

1389-2010/12 $58.00+.00 © 2012 Bentham Science Publishers


Curcumin and Liver Cancer Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 219

Primary liver cancer occurs in the milieu of oxidative 90% of it with the remainder exported to be utilized by the
stress and inflammation (reviewed in ref. [26]). Hepatic in- food industry as additive, preservative, flavoring and color-
flammation, resulting from hepatropic viral infections, ing agent [59]. Turmeric has customarily been used in tradi-
chronic hepatitis, cirrhosis, as well as exposure to toxic hepa- tional Ayurvedic medicine, primarily in South Asia, for
tocarcinogens, represents an early malignant step with the thousands of years as an antiseptic, antibacterial, antiin-
consequent epigenetic events occurring as a consequence of flammatory agent in the treatment of infections, respiratory
a protracted inflammatory response. Considerable evidence, ailments, swelling and rheumatism [60-64]. Turmeric has
accumulated over the past several years, implicates inflam- also been attributed with antiarthritic, anticarcinogenic, an-
mation-driven processes such as production of cytokines, tilipidogenic, cardioprotective, hepatoprotective, hypogly-
chemokines, as well as both reactive oxygen and nitrogen cemic and thrombosuppressive properties [65-70]. Polyphe-
species which contribute to the process of hepatocarcino- nolic, yellow-colored curcuminoids are the primary chemical
genesis [27-31]. Oxidative stress, due to environmental in- constituents present in turmeric. Along with curcumin (cur-
sults including hepatotoxicants, generates reactive oxygen cumin I), the principal curcuminoid, three other curcumi-
species such as superoxide anion and the hydroxyl radical noids, namely demethoxycurcumin (curcumin II), bisde-
and is a predisposing factor to hepatocarcinogenesis [32-34]. methoxycurcumin (curcumin III) and the recently identified
Besides the involvement of the oxidative stress and the in- cyclocurcumin are present in turmeric. Commercial curcu-
flammatory cascade, various molecular and signaling min is a mixture of curcuminoids containing approximately
mechanisms have been proposed in the pathogenesis of HCC 77% curcumin, 18% demethoxycurcumin and 5% bisde-
[35, 36]. methoxycurcumin [58, 68, 69, 71].
Phytochemicals, obtained from both dietary and non- Curcumin, the most important and abundant curcuminoid
dietary sources, have shown potential usefulness as therapeu- is chemically known as diferuloylmethane Fig. (1), and was
tic as well as chemopreventive agents for several chronic first isolated in 1815 and crystallized in 1870 followed by
maladies such as cardiovascular, metabolic, neoplastic and structural elucidation in 1910. Curcumin, intensely yellow at
neurodegenerative diseases [37-44]. Dietary components, acidic pH and red at basic pH, exists primarily enolic as well
present in fruits, vegetables, nuts and spices, have demon- as -diketonic forms which is extremely critical for its prop-
strated significant potential in their ability to suppress car- erty as a free radical scavenger [62, 72]. Curcumin has been
cinogenesis in pre-clinical models and prevent and delay the widely studied over the past two decades and has been at-
occurrence of cancer in high-risk populations [45-48]. Of tributed with therapeutic treatment in a wide array of chronic
particular interest are a multitude of polyphenolic com- disorders, such as cardiovascular and neurodegenerative dis-
pounds found in the aforementioned dietary resources. Die- eases, diabetes, autoimmune disorders, namely allergy,
tary polyphenols, such as anthocyanidins obtained from ber- asthama rheumatoid arthritis and psoriasis, hepatoprotection
ries, catechins present in green tea, curcumin in turmeric, as well as cancer. A multitude of both pre-clinical and clini-
ellagic acid present in pomegranates, lycopene found in to- cal studies have highlighted the significant anti-cancer poten-
matoes, resveratrol in peanuts, grapes and red wine, quer- tial of curcumin (reviewed in refs. [68-71, 73, 74]).
cetin present in apples and red onions and many others, have
shown considerable promise as both preventive and thera-
peutic agents in cancer affecting several organ systems such
as breast, liver, lung, prostate and skin [40, 47, 49-51]. Poly-
phenolic compounds exert anticarcinogenic effects primarily
due to their antioxidant and anti-inflammatory properties as
well as their ability to modulate diverse molecular signaling
mechanisms implicated in carcinogenesis [45, 52-55]. Glau-
ert et al. [56] have recently reviewed the preventive potential Fig. (1) Chemical structure of curcumin.
of dietary antioxidants such as vitamin C, vitamin E, sele-
nium as well as several phytochemicals in preclinical animal
models of HCC as well as human studies. Mann and col- Antioxidant and Anti-Inflammatory Properties of Cur-
leagues [57] have also documented the promising chemopre- cumin
ventive and therapeutic properties of several phytochemicals,
Curcumin owes its therapeutic effects to its potent anti-
including dietary polyphenols in hepatocarcinogenesis. In
oxidant, anti-inflammatory properties as well as its ability to
this article, we review the potential role of curcumin as a
modulate an array of signaling mechanisms [74-77]. Oxida-
chemopreventive and therapeutic agent for HCC.
tive stress plays an important role in the pathogenesis of sev-
eral chronic ailments such as neurodegenerative diseases and
CURCUMIN
cancer. Free radical pathology has been implicated in the
Turmeric (Curcuma longa), also known as Indian saffron process of carcinogenesis [43, 78]. Several studies have
is a rhizomatous herbaceous plant of the ginger family, demonstrated the potent free-radical scavenging ability of
Zingiberaceae, native to the Indian sub-continent, Southeast curcuminoid compounds. Curcumin as well as its curcumi-
Asia as well as several other tropical countries. The dried noid analogs have been shown to attenuate free-radical me-
rhizome is ground to yield a yellow powder, also known as diated lipid peroxidation in several experimental systems
yellow ginger, yellow root or natural yellow, and is widely [79-81]. The phenolic hydroxyl group, the methoxy group as
used as a spice in ethnic South and Southeast Asian cuisine well as the 1,3 -diketone moiety have all been shown to be
[58]. India, the major producer of turmeric, consumes about
220 Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 Darvesh et al.

important structural features in the antioxidant properties of showed that novel difluoro Knoevenagel curcumin conden-
curcuminoids [82]. sates, their Schiff bases as well as copper complexes causes
both growth inhibition and induction of apoptosis in colon
The inflammatory cascade has been held as a key media-
and pancreatic cancer cell lines. Salicyl curcumin inhibited
tor of the pathogenesis of several chronic illnesses such as
autoimmune, cardiovascular, neurodegenerative as well as tumor specific angiogenesis in C57BL/6 mice injected with
B16F-10 melanoma cells [119].
neoplastic diseases [83-86]. Kundu and Surh [30] in their
elegant review discuss the critical and multifaceted role of Several clinical trials conducted over the past several
inflammation in carcinogenesis. Curcumin has been shown years, as well as currently ongoing, have studied the chemo-
to possess potent antiinflammatory properties which contrib- preventive and therapeutic effects of curcumin in cancer of
ute to its therapeutic effects in the aforementioned illnesses various organ systems such as colon, gall bladder, liver, pan-
[73, 87, 88]. Pioneering research by the Aggarwal laboratory creas, as well as the gastrointestinal tract. Although trial re-
has highlighted the powerful anti-inflammatory properties of sults remain encouraging, there exists a critical need to criti-
curcumin [73, 89, 90]. One of the key findings to emerge cally evaluate the clinical effects of curcumin. Several inves-
almost a decade ago is the curcumin-mediated suppression of tigators are of the opinion that encouraging in vitro as well as
nuclear factor-B (NF-B), the master switch in the inflam- in vivo pre-clinical data has not been reflected in the clinical
matory cascade [91]. NF-B activation is known to regulate results in curcumin studies essentially due to poor bioavail-
several key inflammatory mediators such as cytokines, ability. Hence, it is important to utilize the various aforemen-
chemokines and kinases, which have been shown to play a tioned novel drug delivery systems to enhance curcumin’s
critical role in the pathogenesis of most chronic illnesses [85, bioavailability, which may result in improved clinical thera-
92, 93]. The curcumin-mediated attenuation of the NF-B peutic effects [68, 76, 120].
activated inflammatory cascade is an extremely critical
mechanism of its widespread therapeutic profile [94]. Sev- Toxicity Studies
eral investigators have delineated the mechanisms involved
Curcumin is an extremely well tolerated, bioactive and
in curcumin-mediated attenuation of NF-B expression and
nontoxic evident from it being an integral component of the
the NF-B gene products which suppress apoptosis and me-
South Asian diet for thousands of years [68, 121, 122].
diate cell proliferation, invasion and angiogenesis [95-99].
However, it has been opined that the high doses administered
Curcumin treatment also leads to suppression of several in-
in clinical trials remain a cause for potential concern since
flammatory cytokines such as interleukin (IL)-1, IL-1, IL-6,
IL-8, chemokines as well as tumor necrosis factor- and cy- the genotoxic and long-term effects of curcumin administra-
tion have not been systematically investigated [123]. Also,
clooxygenases [95, 100-102]. Curcumin also affects a pleth-
there exist rare anecdotal reports of curcumin-induced ad-
ora of signaling mechanisms implicated in process of car-
verse effects. Curcumin-induced allergic contact dermatitis
cinogenesis. Curcumin interacts with multiple cell signaling
and urticaria has also been reported in humans [124-126].
proteins such as transcriptional factors, protein kinases, cy-
tokine signaling receptors, growth factors, adhesion mole-
cules as well as anti-apoptotic proteins. The aforementioned Role of Synergy
molecular effects of curcumin lead to inhibition of cell pro- The role of synergy in the chemopreventive and thera-
liferation, cell invasion and apoptosis causing suppression of peutic effects of dietary polyphenols has been a subject of
metastasis (reviewed in ref. [75]). recent investigation [127-130]. Besides curcumin, turmeric
also contains three known curcuminoids. Sandur and co-
Pharmacokinetics and Bioavailability of Curcumin workers [131] have systematically evaluated the antioxidant,
The pharmacokinetic profile of curcumin has been a sub- antiinflammatory and antiproliferative effects of curcumin as
well as other curcuminoids. Although, curcumin displayed
ject of detailed investigation over the past few decades [103-
higher activity than the other curcuminoids, the combination
107]. Studies reveal poor absorption and rapid metabolism to
of all the curcuminoids was more potent than any individual
form glucoronoid conjugates which result in limited
curcuminoid, including curcumin, indicating a role of syn-
bioavailability of curcumin. The Aggarwal laboratory has
ergy in the pharmacological effects of curcuminoids.
contributed several excellent articles which review the phar-
macokinetics and bioavailability of curcumin [68, 69, 73, 76,
CURCUMIN AND LIVER CANCER: IN VITRO STUD-
108]. Several strategies have been utilized to enhance the
IES
poor bioavailability of curcumin. Shoba and colleagues [109]
demonstrated that co-administration of piperine, a hepatic Numerous studies, over the past several years, have
and intestinal inhibitor of glucoronidation, significantly en- evaluated the effects of curcumin and its analogs in several
hanced curcumin bioavailability. Use of nanoparticle tech- rodent as well as human hepatoma cells. In one of the initial
nology, liposomal formulations, phosphotidylcholine and studies, Lin et al. [132] showed that curcumin decreased the
cyclodextrin complexes, as well as synthetic analogs of cur- secretion of matrix metalloproteinase-9 (MMP-9) and conse-
cumin resulted in superior bioavailability [110-116]. Several quently inhibited both the migration as well as invasion of
recent studies have investigated the potential anti-cancer SK-Hep-1 cells (Table 1). The anti-invasive and anti-
properties of synthetic curcumin derivatives. John and co- migratory effects of curcumin have also been shown in
workers [117] demonstrated the enhanced cytotoxic and anti- CBO140C12 cells. In this study, Ohasi and colleagues [133]
tumor activity of four synthetic curcuminoids as well as their showed that curcumin-mediated decrease in MMP-9 secretion
copper complexes. In a recent study, Padhye et al. [118] was accompanied by a significant inhibition of the adhesion
Curcumin and Liver Cancer Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 221

Table 1. Biological Effects of Curcumin in Preclinical In Vitro Models of Liver Cancer

Biological Effects Mechanisms of Action Concentration Reference

Curcumin inhibited the migration as well as invasion of


MMP-9 secretion 10 M Lin et al. [132]
HCC-SK-Hep-1 cells.

MMP-9 secretion,
Curcumin abrogated the invasion and migration of
fibronectin migration, 0.1-10 M Ohasi et al. [133]
CBO140C12 cells.
laminin migration

Curcumin inhibited histone acetylation in Hep3B cells. HAT, ROS 50, 100 M Kang et al. [134]

Curcumin showed antiproliferating effects in Bel7402, apoptosis,


1-32 M Cui et al. [135]
SGC7901 and HL60 cells. telomerase activity

Curcumin inhibited cell survival in Hepa1-6 cells with


10-40 g/ml Jia et al. [136]
downregulated VEGF-A expression.

Curcumin attenuated cell growth in Huh7, Hep3B, HepG2,


SK-Hep-1, QGY-7703 cells in which the G2/M arrest was apoptosis 25 M Wang et al. [137]
previously abrogated.

Curcumin inhibited growth and produced dose-dependent


damage in both the mitochondrial and nuclear DNA in 8-OHdG, ROS, TBARS 2.5-40 g/ml Cao et al. [138]
HepG2 cells.

Curcumin produced mitochondrial hyperpolarization in


m, cyt. c 2.5-40 g/ml Cao et al. [139]
HepG2 cells.

Curcumin inhibited the proliferation of HEP3B, SK-Hep-1


Notch1 signaling, NICD 10-40 M Ning et al. [141]
and SNU449 cells.

Curcumin and its -diketone modified analogs inhibited


apoptosis 5-25 M Simoni et al. [142]
proliferation in HA22T/VGH cells.

Curcumin as well as tetrahydrocurcumin attenuated the


20-180 mol/L Yoysungnoen et al. [143]
proliferative activity of HepG2 cells.

Curcumin and its novel analog GL63 inhibited growth in


apoptosis, CHOP 5, 10, 20 M Xiao et al. [144]
HepG2 cells.

apoptosis,
Curcumin, alone or in combination with cisplatin and
NF-B (p65),
doxorubicin synergistically exerted cell growth inhibitory 10, 20, 25 M Notarbartolo et al. [145]
COX-2, c-myc,
effects in HA22T/VGH cells.
Bcl-XL, Bcl-XS
Abbreviations: CHOP, C/EBP homologous protein; COX-2, cycloxygenase-2; cyt. c, cytochrome c; m, mitochondrial membrane potential; 8-OHdG, 8-hydroxydeoxyguanosine;
HAT, histone acetyltransferase; MMP-9, matrix metalloproteinase-9; NF-B, nuclear factor-kappaB; NICD, Notch1 intracellular domain; ROS, reactive oxygen species; TBARS,
thiobarbituric acid-reactive substances, VEGF-A, vascular endothelial growth factor-A.

and migration of both fibronectin and laminin. Curcumin, sequent abrogation of the G2/M arrest facilitated curcumin-
akin to other dietary polyphenols, possesses both antioxidant mediated apoptosis in several hepatoma cell lines. The cur-
as well as pro-oxidant properties as evidenced by the curcu- cumin-mediated growth inhibition and DNA damage in both
min-mediated increase in reactive oxygen species (ROS). In mitochondria as well as the nucleus involved pro-oxidant
this study, Kang and co-workers [134] have observed an mechanisms as evidenced by increased ROS and lipid perox-
increase in ROS levels and a decrease in histone acetyltrans- ide levels in HepG2 cells [138]. In a subsequent study, Cao
ferase activity in curcumin-mediated histone hypoacetylation et al. [139] showed that curcumin produced mitochondrial
in Hep3B cells. Cui et al. [135] demonstrated that the anti- hyperpolarizaton, elevated mitochondrial membrane poten-
proliferative effects of curcumin were accompanied by pro- tial and also increased cytochrome c release in HepG2 cells.
nounced apoptosis and attenuation of telomerase activity in Upregulation of the notch1 receptor and signaling pathways
Bel7402, SGC7901 as well as HL60 cells. Curcumin also in hepatocarcinogenesis result in increased cellular prolifera-
inhibited cell survival in Hepa1-6 cells with downregulated tion as well as inhibition of apoptosis [140]. A recent study
expression of the vascular endothelial growth factor-A [136]. by Ning and coworkers [141] showed that curcumin elicited
In an interesting study, Wang and coworkers [137] showed downregulation of the notch1 signaling pathway as well as
that downregulation of the Chk1 protein expression and sub- the notch intracellular domain in HEP3B, SK-Hep-1 and
222 Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 Darvesh et al.

SNU449 cells. Curcumin as well as its analogs have shown stage model to investigate the potential anti-hepatocarcino-
anti-proliferative effects in various hepatoma cells [142- genic properties of curcumin. In these studies, curcumin ad-
144]. Simoni and colleagues [142] showed that curcumin and ministration prevented hepatocellular carcinogenesis in rats.
its -diketone modified analogs stimulated apoptosis and The investigators also reported that curcumin normalized the
produced antiproliferative activity in HA22T/ VGH cells. elevated levels of several enzymes of liver function and bili-
Tetrahydrocurcumin showed comparable results to curcumin rubin, increased glutathione content, elevated antioxidant
with regards to the attenuation of the proliferative activity of enzymes and attenuated lipid peroxidation. Curcumin has
HepG2 cells [143]. In an equivalent study, curcumin as well also been shown to suppress the formation of hepatic ade-
as its novel analog GL63 produced an inhibitory effect on nomas in mice treated with, 2,2'-dihydroxy-di-n-propylnitro-
HepG2 cells. The inhibitory effect was accompanied by in- samine (DHPN) [157].
creased apoptosis and enhanced expression of C/EBP ho- In addition to aforementioned positive reports, several
mologous protein, a pro-apoptotic activating protein [144].
studies have documented the failure of curcumin in protect-
The effects of curcumin in combination with standard che-
ing against chemical hepatocarcinogenesis in rodents. Using
motherapeutic agents have also been investigated. In an in-
a multi-organ hepatocarcinogenesis rat model Takaba and
teresting study, Notarbartalo and colleagues [145] showed
colleagues [158] reported the inability of curcumin to pre-
that curcumin when combined with cisplatin and doxorubicin
vent HCC in rats treated with DNPN, N-ethyl-N-hydroxy-
caused a synergistic cell growth inhibition in HA22T/VGH ethylnitrosamine and 3, 2'-methyl-4-aminobiphenyl. Curcu-
cells. The aforementioned effect was accompanied by an
min also failed to attenuate both the DENA-induced increase
increase in apoptosis and a decrease in NF-B activation as
in formation of glutathione S-transferase (GST)-positive foci
well as a lowering of COX-2 levels.
as well as the DENA/2-amino-3,8-dimethylimidazo[4,5-f]
quinoxaline-induced hepatocarcinogenesis in rats [159, 160].
CURCUMIN AND LIVER CANCER: IN VIVO STUD-
Curcumin treatment did not prevent the GST-positive foci
IES formation in rats treated with DENA and 3,3',4,4'-tetrachlo-
The effects of curcumin and its analogous compounds robiphenyl [161]. In an interesting study, chronic curcumin
have been a subject of considerable investigation over more administration did not prevent the copper-induced HCC in
than the past decade in several chemically-induced as well as rats [126]. A common feature of the aforementioned studies
xenograft preclinical rodent models of HCC. These studies which document the inability of curcumin to protect against
document the potential role of curcumin as well as its ana- chemically-induced hepatocarcinogenesis is the administra-
logs in the prevention and treatment of HCC (Table 2). tion of a relatively low dose of dietary curcumin which may
lead to its pro-oxidant effects. A low dietary dose may also
Chemical Models of Hepatocarcinogenesis offer limited bioavailability of curcumin.
Chemically-induced HCC remains one of the most exten- Xenograft Models of Hepatocarcinogenesis
sively used methods both of the study of the biological
mechanisms of HCC as well as the elucidation of the che- A few investigators have studied the therapeutic effects
mopreventive and therapeutic effects of potential anti-cancer of curcumin in xenograft models of hepatocarcinogenesis.
agents [146-148]. In one of the earliest studies, Soni and Busquets et al. [162], demonstrated that curcumin treatment
colleagues [149] demonstrated that curcumin as well as tur- blocked tumor growth in the Yoshida ascites hepatoma in
meric extract reduced the number of gamma glutamyl rats. Curcumin also suppressed the intrahepatic metastasis in
transpeptidase-positive foci in rats challenged with aflatoxin female mice implanted with CBO140C12 cells [132]. Subse-
B1. Nitrosoamines in general and diethylnitrosoamine quent studies have also demonstrated the curcumin-mediated
(DENA) in particular, either used alone or in presence of a tumor inhibition in mice xenografted with several hepatoma
promoter such as phenobarbital (PB) have been a corner- cells [135, 141]. Although the aforementioned xenograft
stone of pre-clinical liver cancer research [146, 147, 150]. studies highlight the protective role of curcumin, the investi-
Several studies have investigated the pharmacological effects gators do not provide any mechanistic explanation to curcu-
of curcumin and its analogs in DENA-induced HCC. Chuang min’s anti-tumor properties. Yoysungnoen and colleagues
et al. [152, 153] have shown that curcumin protected against [143, 163, 164] in a series of studies document the signifi-
DENA-induced hyperplasia and HCC in rodents. The cant antiangiogenic property of curcumin and its analog in
authors of the aforementioned studies also investigated the nude mice implanted with human HepG2 cells. Curcumin
underlying biochemical mechanisms of curcumin in DENA- treatment decreased both capillary density and vascularity
treated rodents. Curcumin strongly inhibited the DENA- and also decreased expression of the pro-angiogenic vascular
induced increase in the expression of the oncogenic p21ras endothelial growth factor.
and tumor suppressor p53 protein as well as the elevated
levels of proliferating cell nuclear antigen as well as NF-B. CONCLUSION
Thapliyal and coworkers [153] have also documented the
The essential premise for the potential use of curcumin as
attenuation of DENA-induced hepatocarcinogenesis in rats
a chemopreventive and therapeutic agent in HCC rests on its
by dietary curcumin. The suppressive effects of curcumin in
potent antioxidant and anti-inflammatory properties as well
the DENA-initiated and 2-acetylaminofluorene-promoted
as its ability to modulate a multitude of signaling mecha-
alteration of hepatic foci in rats was accompanied by the
nisms. It is clearly evident from the in vitro as well as in vivo
restoration of the normal levels of several hepatic metabolic pre-clinical models that curcumin, along with other curcumi-
enzymes [154]. Sreepriya and Bali [155, 156] utilized the
noids, hold great promise as curative agents for HCC.
well-characterized DENA-initiated and PB-promoted two-
Curcumin and Liver Cancer Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 223

Table 2. Biological Effects of Curcumin in Preclinical In Vivo Models of Liver Cancer

Biological Effects Mechanisms of Action Dose/Duration Reference

Chemical models of hepatocarcinogenesis

Curcumin and turmeric extract reduced GGT+ foci in Curcumin 0.005% and turmeric
Soni et al. [149]
AFB1-treated male Wistar rats. 0.05%, diet; 22 weeks

Curcumin treatment protected against DENA-induced


p21ras, p53, NF-B 200, 600 mg/kg, p.o. Chuang et al. [151]
hyperplasia in male Wistar rats.

Curcumin suppressed the DENA-induced hepatocel- 0.2% diet;


p21ras, PCNA, Cdc2 Chuang et al. [152]
lular carcinoma in male C3H/HeN mice. 38 weeks

Curcumin attenuated DENA-induced GGT+ foci and 0.2, 1, 5%, diet;


Thapliyal et al. [153]
hepatocellular carcinoma in female Wistar rats. 10 weeks

Curcumin prevented the DENA-initiated and 2-AAF-


ATPase, ALP, G6Pase, 200 mg/kg/day, p.o.;
promoted development of altered GGT+/GST+ hepatic Shukla and Arora [154]
GGT, GST-P 5 days
foci in male Wistar rats.

Curcumin significantly blocked the DENA-initiated 100 mg/kg/day, p.o.;


ALT, AST, ALP, BR Sreepriya and Bali [155]
and PB-promoted nodule incidence in male Wistar rats. 14 weeks

Curcumin administration attenuated the DENA/PB- 100 mg/kg/day, p.o.;


GSH, GST, GPX, MDA Sreepriya and Bali [156]
induced hepatic oxidative stress in male Wistar rats. 14 weeks

Curcumin suppressed the DHPN-induced liver ade-


10, 100, 200 M, i.p.; 22 weeks Huang et al. [157]
noma formation in male BALB/c mice.

Curcumin failed to protect against hepatocellular


1% diet;
carcinoma in male F344 rats treated with DHPN, Takaba et al. [158]
36 weeks
EHEN and DMAB.

Curcumin did not inhibit DENA-induced increase in 0.5%, diet;


Hirose et al. [159]
GST-P+ foci in male F344 rats. 6 weeks

Curcumin did not prevent the DENA/MeIQx-induced 0.5%, diet;


Hirose et al. [160]
GST-P+ foci in male F344 rats. 26 weeks

Curcumin did not decrease the preneoplastic GST-P+


0.5% diet;
foci in female Sprague-Dawley rats treated with Tharappel et al. [161]
80 days
DENA and PCB-77.

Curcumin did not protect against copper-induced


0.5% diet;
hepatocellular carcinoma in Long-Evans Cinnamon Frank et al. [126]
100 weeks
rats.

Xenograft models of hepatocarcinogenesis

Curcumin treatment restrained the tumor growth in 20 g/kg/day, i.p.;


Busquets et al. [162]
the Yoshida ascites hepatoma in male Wistar rats. 6 days

Curcumin suppressed the intrahepatic metastasis in


100, 200 mg/kg/day, p.o.;
female B6C3F1 mice implanted with CBO140C12 Ohasi et al. [133]
20 days
cells.

Curcumin significantly attenuated tumor growth in


female BALB/c athymic mice injected with Bel7402, 50, 100, 200 mg/kg/day Cui et al. [135]
SGC7901 and HL60 cells.

Curcumin treatment significantly attenuated tumor


100 mg/kg/day, i.p.;
growth in male nude athymic mice injected with SK- Ning et al. [141]
16 days
Hep-1 cells.

Curcumin produced antiangiogenic activity in male


3000 mg/kg/day, p.o.;
BALB/c nude mice implanted with human HepG2 capillary density Yoysungnoen et al. [163]
14 days
cells.
224 Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 Darvesh et al.

(Table 2) contd….

Biological Effects Mechanisms of Action Dose/Duration Reference

Xenograft models of hepatocarcinogenesis

Curcumin exerted antiangiogenic activity in HepG2 300, 3000 mg/kg/day, p.o.;


COX-2, VEGF Yoysungnoen et al. [164]
cell-implanted male BALB/c nude mice. 14 days

Curcumin and tetrahydrocurcumin prevented tumor


300, 3000 mg/kg/day, p.o.;
angiogenesis in male BALB/c nude mice inoculated capillary vascularity Yoysungnoen et al. [143]
21 days
with human HepG2 cells.
Abbreviations: 2-AAF, 2-acetylaminofluorene; AFB1, aflatoxin B1; ALT, alanine transaminase; ALP, alkaline phosphatase; AST, aspartase transaminase;
ATPase, adenosine triphosphatase; BR, bilirubin; COX-2, cycloxygenase-2; DENA, diethylnitrososamine; DHPN, 2,2'-dihydroxy-di-n-propylnitrosamine;
DMAB, 3,2'-methyl-4-aminobiphenyl; EHEN, N-ethyl-N-hydroxyethylnitrosamine; G6Pase, glucose-6-phosphatase; GGT, gamma-glutamyl transpeptidase;
GPX, glutathione peroxidase; GSH, glutathione; GST, glutathione S-transferase; GST-P, glutathione S-transferase placental form; i.p., intaperitoneal; MDA,
malondialdehyde; MeIQx, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline; NF-B, nuclear factor-kappaB; p.o., per os, PB, phenobarbital; PCB-77, 3,3',4,4'-
tetrachlorobiphenyl; PCNA, proliferating cell nuclear antigen; VEGF, vascular endothelial growth factor.

However, considerable amount of work and significant ef- [5] Schütte, K.; Bornschein, J.; Malfertheiner, P. Hepatocellular carci-
forts would be required before curcumin’s routine clinical noma–epidemiological trends and risk factors. Dig. Dis., 2009, 27,
80-92.
use for HCC. The key to success is translational research – [6] Sherman, M. Hepatocellular carcinoma: epidemiology, risk factors
well designed clinical trials which systematically evaluate and screening. Semin. Liver Dis., 2005, 25, 143-154.
the anti-cancer effects of curcumin against HCC. A major [7] McGlynn, K.A.; Tsao, L.; Hsing, A.W.; Devasa, S.S.; Fraumeni, J.
problem which needs attention is the reduced bioavailability International trends and patterns of primary liver cancer. Int. J.
Cancer, 2001, 94, 290-296.
of curcumin. Low therapeutic concentration of curcumin in [8] Bosch, F.X.; Ribes, J.; Díaz, M.; Cléries, R. Primary liver cancer:
humans has been cited as a reason for lack of sufficient suc- worldwide incidence and trends. Gastroenterology. 2004, 127, S5-
cess clinical trials. There exists a critical need to develop S16.
novel dosage forms of curcumin to enhance its bioavailabil- [9] Jemal, A.; Siefel, R.; Xu, J.; Ward, E. Cancer statistics, 2010. CA
ity and investigate their anti-cancer effects in clinical studies. Cancer J. Clin., 2010, 60, 277-300.
[10] Bartsch, H.; Montesano, R. Relevance of nitrosamines to human
Epidemiological studies, conducted in regions such as South cancer. Carcinogenesis, 1984, 5, 1381-1393.
Asia where turmeric is an integral part of the diet, would [11] Okuda, K. Hepatocellular carcinoma. J. Hepatol., 2000, 32, 225-
provide valuable clues to its potential effects against liver 237.
cancer. Although curcumin is the most widely known and [12] Kensler, T.W.; Egner, P.A.; Wang, J.B. Chemoprevention of hepa-
tocellular carcinoma in aflatoxin endemic areas. Gastroenterology.
effective curcuminoid present in turmeric, this spice also 2004, 127, S310-S318.
contains at least three additional polyphenolic curcuminoids. [13] Ribes, J.; Clèries, R.; Esteban, L.; Moreno, V.; Bosch, F.X. The
Hence, the role of the synergistic effects of curcuminoids influence of alcohol consumption and hepatitis B and C infections
pleads further attention. Although, curcumin is a regular die- on the risk of liver cancer in Europe. J. Hepatol., 2008, 49, 233-
tary component in various countries, and deemed safe and 242.
[14] Llovet, J.M.; Burroughs, A.; Bruix, J. Hepatocellular carcinoma.
nontoxic, its long term effects need to be investigated. The Lancet, 2003, 362, 1907-1917.
high doses used clinically remain a source of concern and a [15] Mazzaferro, V.; Regalia, E.; Doci, R.; Andreola, S.; Pulvirenti, A;
safe dosing regimen needs to be established. In conclusion, Bozzetti, F. Liver transplantation for the treatment of small hepato-
although significant effort is essential prior to its successful cellular carcinoma in patients with cirrhosis. N. Eng. J. Med., 1996,
334, 693-699.
clinical application, curcumin remains a promising chemo- [16] Takayasu, K.; Muramatsu, Y.; Moriyama, N.; Hasegawa, H.; Ma-
preventive and therapeutic agent in the treatment of HCC. kuuchi, M.; Okazaki, N. Clinical and radiological assessment of the
results of hepatectomy for small hepatocellular carcinoma and
ACKNOWLEDGEMENTS therapeutic arterial embolization for postoperative recurrence. Can-
cer, 1989, 64, 1848-1853.
The authors of this manuscript wish to express their sin- [17] Ebara, M.; Ohto, M.; Sugiura, N.; Kita, K.; Yoshikawa, M.; Okuda,
cere thanks to Ms. Karishma A. Samtani for her kind help K. Percutaneous ethanol injection for the treatment of small hepa-
tocellular carcinoma. Study of 95 patients. J. Gastrol. Hepatol.,
with collection of primary literature reviewed in this article. 1990, 5, 616-626.
[18] Livraghi, T.; Giorgio, A.; Marin, G.; Salmi, A.; de Sio, I.; Bolondi,
REFERENCES L. Hepatocellular carcinoma and cirrhosis in 746 patients: long-
term results of percutaneous ethanol injection. Radiology, 1995,
[1] American Cancer Society. Global Cancer Facts & Figures, 2007. 197, 101-108.
[2] El-Serag, H.B.; Rudolph, K.L. Hepatocellular carcinoma: epidemi- [19] Rossi, S.; Di Stasi, M.; Buscarini, E.; Cavanna, I.; Quaretti, P.;
ology and molecular carcinogenesis. Gastroenterology. 2007, 132, Squassante, E. Percutaneous radiofrequency interstitial thermal ab-
2557-2576. lation in the treatment of small hepatocellular carcinoma. Cancer J.
[3] Thun, M.J.; DeLancey, J.O.; Center, M.M.; Jemal, A.; Ward, E.M. Sci. Am., 1995, 1, 73-81.
The global burden of cancer: priorities for prevention. Carcino- [20] Sato, M.; Watanabe, Y.; Ueda, S.; Iseki, S.; Abe, Y.; Sato, N. Mi-
genesis, 2010, 31, 100-110. crowave coagulation therapy for hepatocellular carcinoma. Gastro-
[4] Parkin, D.M.; Bray, F.; Ferlay, J.; Pisani, P. Global cancer statis- enterol., 1996, 110, 1507-1514.
tics, 2002. CA Cancer J. Clin., 2005, 55, 74-108. [21] Llovet, J.M.; Ricci, S.; Mazzaferro, V. Sorafenib in advanced hepa-
tocellular carcinoma. N. Eng. J. Med., 2008, 359, 378-390.
Curcumin and Liver Cancer Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 225

[22] Je, Y.; Schutz, F.A.B.; Choueiri, T.K. Risk of bleeding with vascu- [48] Stan, S.D.; Kar, S.; Stoner, G.D.; Singh, S.V. Bioactive food com-
lar endothelial growth factor receptor tyrosine-kinase inhibitors ponents and cancer risk reduction. J. Cell. Biochem., 2008, 104,
sunitinib and sorafenib: a systematic review and meta-analysis of 339-356.
clinical trials. Lancet Oncol., 2009, 10, 967-974. [49] Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenes, L.
[23] Lu, S.C. Where are we in the chemoprevention of hepatocellular Polyphenols: food sources and bioavailability. Am. J. Clin, Nutr.,
carcinoma? Hepatol., 2010, 51, 734-736. 2004, 79, 727-747.
[24] Okuno, M.; Kojima, S.; Moriwaki, H. Chemoprevention of hepato- [50] Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Remesy, C.
cellular carcinoma: concept, progress and perspectives. J. Gastro- Bioavailability and bioefficacy of polyphenols in humans. I. Re-
enterol. Hepatol., 2001, 16, 1329-1335. view of 97 bioavailability studies. Am. J. Clin. Nutr., 2005, 81,
[25] Kensler, T.W.; Quain, G.S.; Chen, J.G.; Groopman, J.D. Transla- 230S-242S.
tional strategies for cancer prevention in liver. J. Natl. Cancer Inst., [51] Lee, K.W.; Lee, H.J. The roles of polyphenols in cancer chemopre-
2003, 3, 321-329. vention. Biofactors, 2006, 26, 105-121.
[26] Bishayee, A.; Politis, T.; Darvesh, A.S. Resveratrol in the chemo- [52] Surh, Y.J. Cancer chemoprevention with dietary phytochemicals.
prevenion and treatment of hepatocellular carcinoma. Cancer Nat. Rev. Cancer, 2003, 3, 768-780.
Treat. Rev., 2010, 36, 43-53. [53] Milner, J.A. Molecular targets for bioactive food components. J.
[27] Laurent-Puig, P.; Zucman-Rossi, J. Genetics of hepatocellular Nutr., 2004, 134, 2492S-2498S.
tumors. Oncogene, 2006, 25, 3778-3786. [54] Rahman, I.; Biswas, S.K.; Kirkham, P.A. Regulation of inflamma-
[28] Prieto, J. Inflammation, HCC and sex: IL-6 in the centre of the tion and redox signaling by dietary polyphenols. Biochem. Phar-
triangle. J. Hepatol., 2008, 48, 380-381. macol., 2005, 72, 1439-1452.
[29] Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related [55] Kim, Y.S.; Young, M.R.; Bobe, G.; Colbum, N.H.; Milner, J.A.
inflammation. Nature, 2008, 454, 436-444. Bioactive food components, inflammatory targets, and cancer pre-
[30] Kundu, J.K.; Surh, Y.-J. Inflammation: gearing the journey to vention. Cancer Prev. Res., 2009, 2, 200-208.
cancer. Mut. Res., 2008, 659, 15-30. [56] Glauert, H.P.; Mason-Calfee, K.; Stemm, D.V.; Tharappel, J.C.
[31] Berasain, C.; Casillo, J.; Perugorria, M.J.; Latasa, M.U.; Prieto, J.; Dietary antioxidants in the prevention of hepatocarcinogenesis: A
Avila, M.A. Inflammation and liver cancer: new molecular links. review. Mol. Nutr. Food Res., 2010, 54, 875-896.
Ann. N.Y. Acad. Sci., 2009, 1155, 206-221. [57] Mann, C.D.; Neal, C.P.; Garcea, G.; Manson, M.M.; Dennison,
[32] Klaunig, J.E.; Kamendulis, L.M. The oxidative stress in carcino- A.R.; Berry, D.P. Phytochemicals as potential chemopreventive
genesis. Annu. Rev. Pharmacol. Toxicol., 2004, 44, 239-267. and chemotherapeutic agents in hepatocarcinogenesis. Eur. J. Can-
[33] Kawanishi, S.; Hiraku, Y.; Pinlaor, S.; Ma, N. Oxidative stress and cer Prev., 2009, 18, 13-25.
nitrative DNA damage in animals and patients with inflammatory [58] Ravindran, P.N. Turmeric-the golden spice of life. In: Turmeric:
diseases in relation to inflammation-related carcinogenesis. Biol. The genus Curcuma. Taylor and Francis Group: London, 2006, pp.
Chem., 2006, 387, 365-372. 1-14.
[34] Gius, D.; Spitz, D.R. Redox signaling in cancer biology. Antioxid. [59] Ploto, A. Post-production management for improved market access
Redox Signal, 2006, 8, 1249-1252. for herbs and spices – turmeric. Food and Agriculture Organization
[35] Pang, R.; Tse, E.; Poon, R.T.P. Molecular pathways in hepatocellu- of the United Nations (FAO), 2003.
lar carcinoma. Cancer Lett., 2006, 240, 157-169. [60] Ammon, H.P.; Wahl, M.A. Pharmacology of Curcuma longa.
[36] Wong, C.-M.; Ng, I.O.L. Molecular pathogenesis of hepatocellular Planta Med., 1991, 57, 1-7.
carcinoma. Liver Int., 2007, 160-174. [61] Araujo, C.C.; Leon, L.L. Biological activities of Curcuma longa L.
[37] Lee, K.W.; Lee, H.J.; Lee, C.Y. Vitamins, phytochemicals, diets, Mem. Inst. Oswaldo Cruz., 2001, 96, 723-728.
and their implementation in cancer chemoprevention. Crit. Rev. [62] Aggarwal, B.B.; Kumar, A.; Bharti, A.C. Anticancer potential of
Food Sci. Nutr., 2004, 44, 437-452. curcumin: preclinical and clinical studies. Anticancer Res., 2003,
[38] Aggarwal, B.B.; Shishodia, S. Molecular targets of dietary agents 23, 363-398.
for prevention and therapy of cancer. Biochem. Pharmacol., 2006, [63] Aggarwal, B.B.; Bhatt, I.D.; Ichikawa, H.; Ahn, K.S.; Sethi, G.;
71, 1397-1421. Sandur, S.K. Curcumin-biological and medicinal properties. In:
[39] Aggarwal, B.B.; Ichikawa, H.; Garodia, P.; Weersinghe, P.; Sethi, Turmeric: The genus Curcuma. Taylor and Francis Group: London,
G.; Bhatt, I.D.; Pandey, M.K.; Shishodia, S.; Nair, M.G. From tra- 2006, pp. 297-368.
ditional ayurvedic medicine to modern medicine: identification to [64] Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcu-
therapeutic targets for suppression of inflammation and cancer. min: the Indian solid gold. Adv. Exp. Med. Biol., 2007, 595, 1-75.
Exp. Opin. Ther. Targets, 2006, 10, 87-118. [65] Kiso, Y.; Suzuki, Y.; Watanabe, N.; Oshima, Y.; Hikino, H. Anti-
[40] Ullah, M.F.; Khan, M.W. Food as Medicine: Potential therapeutic hepatotoxic principles of Curcuma longa rhizomes. Planta Med.,
tendencies of plant derived polyphenolic compounds. Asian Pacific 1983, 49, 185-187.
J. Cancer Prev., 2008, 9, 187-196. [66] Srivastava, R.; Dikshit, M.; Srimal, R.C.; Dhawan, B.N. Anti-
[41] Moiseeva, E.P.; Manson, M.M. Dietary chemopreventive phyto- thrombotic effect of curcumin. Thromb. Res., 1985, 40, 413-417.
chemicals: Too little or too much. Cancer Prev. Res., 2009, 2, 611- [67] Babu, P.S.; Srinivasa, K. Infulence of dietary curcumin and choles-
616. terol on the progression of experimentally induced diabetes in al-
[42] Manach, C.; Hubert, J.; Llorach, R.; Scalbert, A. The complex links bino rats. Mol. Cell Biochem., 1995, 152, 13-21.
between dietary phytochemicals and human health deciphered by [68] Goel, A.; Kunnumakkara, A.B.; Aggarwal, B.B. Curcumin as
metabolomics. Mol. Nutr. Food Res., 2009, 53, 1303-1315. ‘Curecumin”: from kitchen to clinic. Biochem. Pharmacol., 2008,
[43] Bishayee, A.; Darvesh, A.S. Oxidative stress in cancer and neu- 75, 787-809.
rodegenerative diseases: prevention and treatment by dietary anti- [69] Goel, A.; Jhurani, S.; Aggarwal, B.B. Multi-targeted therapy by
oxidants. In: Free Radicals: Formation, Types and Effects, Eds: curcumin: how spicy is it? Mol. Nutr. Food Res., 2008, 52, 1010-
Kozyrev, D., Slutsky, V., Nova Science Publishers, Inc, Haup- 1030.
pauge, NY, 2010, 1-55. [70] Rivera-Espinoza, Y.; Muriel, P. Pharmacological actions of curcu-
[44] Darvesh, A.S.; Carroll, R.T.; Bishayee, A.; Geldenhuys, W.J.; Van min in liver diseases or damage. Liver Int., 2009, 1457-1466.
der Schyf, C.J. Oxidative stress and Alzheimer’s disease: dietary [71] Anand, P.; Sundaram, C.; Jhurani, S.; Kunnumakkara, A.B.; Ag-
polyphenols as potential therapeutic agents. Exp. Rev. Neurother., garwal, B.B. Curcumin and cancer: an “old-age” disease with an
2010, 10, 729-745. “age-old” solution. Cancer Lett., 2008, 267, 133-164.
[45] Johnson, I.T. Phytochemicals and cancer. Proc. Nutr. Soc.; 2007, 6, [72] Shen, L.; Ji, H.F. Theoretical study on physiochemical properties of
207-215. curcumin. Spectrochim. Acta A. Mol. Biomol. Spectrom., 2007, 67,
[46] Russo, G.L. Ins and outs of dietary phytochemicals in cancer che- 619-623.
moprevention. Biochem. Pharmacol.; 2007, 74, 533-544. [73] Aggarwal, B.B.; Harikumar, K.B. Potential therapeutic effects of
[47] Khan, N.; Afaq, F.; Mukhtar, H. Cancer chemoprevention through curcumin, the anti-inflammatory agent, against neurodegenerative,
dietary antioxidants: progress and promise. Antioxid. Redox. Sig- cardiovascular, pulmonary, metabolic, autoimmune and neoplastic
nal., 2008, 10, 475-510. diseases. Int. J. Biochem. Cell Biol., 2009, 41, 40-59.
[74] Sethi, G.; Sung, B.; Aggarwal, B.B. The role of curcumin in mod-
ern medicine. In: Herbal Drugs: Ethnomedicine to Modern Medi-
226 Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 Darvesh et al.

cine, Ramawat, K.G., Ed., Springler-Verlag Berlin, Heidelberg, expression of cell proliferation and anti-apoptotic and metastatis
2009, 97-113. gene products through suppression of IkappaBalpha kinase and Akt
[75] Calabrese, V.; Bates, T.E.; Mancuso, C.; Cornelius, C.; Ven- activation. Mol. Pharmacol., 2006, 69, 195-206.
timiglia, B.; Cambria, M.T.; Di Renzo, L.; De Lorenzo, A.; [98] Kamat, A.M.; Sethi, S.; Aggarwal, B.B. Curcumin potentiates the
Dinkova-Kostova, A.T. Curcumin and the cellular stress response apoptotic effects of chemotherapeutic agents and cytokines through
in free radical-related diseases. Mol. Nutr. Food Res., 2008, 52, down-regulation of nuclear factor-kappaB and nuclear factor-
1062-1073. kappaB-regulated gene products in IFN-alpha-sensitive and IFN-
[76] Kunnumakkara, A.B.; Anand, P.; Aggarwal, B.B. Curcumin inhib- alpha-resistant human bladder cancer cells. Mol. Cancer Ther.,
its proliferation, invasion, angiogenesis and metastasis of different 2007, 6, 1022-1030.
cancers through interaction with multiple cell signaling proteins. [99] Deeb, D.; Jiang, H.; Gao, X.; Al-Holou, S.; Danyluk, A.L.; Dul-
Cancer Lett., 2008, 269, 199-225. chavsky, S.A. Curcumin sensitizes human prostate cancer cells to
[77] Sarkar, F.H.; Li, Y.; Wang, Z.; Padhye, S. Lesson learned from tumor necrosis factor-related apoptosis-inducing ligand/Apo2L-
nature for the development of novel anti-cancer agents: implication induced apoptosis by suppressing nuclear factor-kappab via inhibi-
of isoflavone, curcumin, and their synthetic analogs. Curr. Pharm. tion of the prosurvival Akt signaling pathway. J. Pharmacol. Exp.
Des., 2010, 16, 1801-1812. Ther., 2007, 321, 616-625.
[78] Valko, M.; Rhodes, C.J.; Moncol, J.; Izaokovic, M.; Mazur, M. [100] Li, L.; Aggarwal, B.B.; Shishodia, S.; Abbruzzese, J.; Kurzrock, R.
Free radicals, metals and antioxidants in oxidative stress-induced Nuclear factor-kappaB and Ikappa-B kinase are constitutively ac-
cancer. Chem. Biol. Interact., 2006, 160, 1-40. tive in human pancreatic cells, and their down-regulation by cur-
[79] Sharma, O.P. Antioxidant activity curcumin and related com- cumin (diferuloylmethane) is associated with the suppression of
pounds. Biochem. Pharmacol., 1976, 25, 1811-1812. proliferation and the induction of apoptosis. Cancer, 2004, 101,
[80] Osawa, T.; Sugiyama, Y.; Inayoshi, M.; Kawakishi, S. Antioxida- 2351-2362.
tive activity of tetrahydrocurcuminoids. Biosci. Biotechnol. Bio- [101] Shakibaei, M.; Schulze-Tanzil, G.; John, T.; Mobasheri, A. Curcu-
chem., 1995, 59, 1609-1612. min protects human chodrocytes from IL-1beta-induced inhibition
[81] Ak, T.; Gulcin, I. Antioxidant and radical scavenging properties of of collagen type II and beta1-integrin expression and activatin of
curcumin. Chem. Biol. Interact., 2008, 174, 27-37. caspase-3: an immunomorphological study. Ann. Anat., 2005, 187,
[82] Sugiyama, Y.; Kawakishi, S.; Osawa, T. Involvement of the beta- 487-497.
diketone moiety in the antioxidative mechanism of tetrahydrocur- [102] Skommer, J.; Wlodkowic, D.; Pelkonen, J. Gene-expression profil-
cumin. Biochem. Pharmacol., 1996, 52, 519-525. ing during curcumin-induced apoptosis reveals downregulation of
[83] Aggarwal, B.B.; Shishodia, S.; Sandur, S.K.; Pandey, M.K.; Sethi, CXCR4. Exp. Hematol., 2007, 35, 85-95.
G. Inflammation and cancer: how hot is the link? Biochem. Phar- [103] Wahlstrom, B.; Blennow, G. A study on the fate of curcumin in the
macol., 2006, 72, 1605-1621. rat. Acta Pharmacol. Toxicol. (Copenh.), 1978, 43, 86-92.
[84] Hansson, G.K.; Robertson, A.K.; Soderberg-Naucler, C. Inflamma- [104] Holder, G.M.; Plummer, J.L.; Ryan, A.J. The metabolism and
tion and atherosclerosis. Annu. Rev. Pathol., 2006, 1, 297-329. excretion of curcumin in the rat. Xenobiotica, 1978, 8, 761-768.
[85] Libby, P. Inflammatory mechanisms: the molecular basis of in- [105] Ravindranath, V.; Chandrasekhara, N. Absorption and tissue distri-
flammation and disease. Nutr. Rev., 2007, 65, S140-S146. bution of curcumin in rats. Toxicol., 1980, 16, 259-265.
[86] Packard, R.R.; Libby, P. Inflammation in atherosclerosis: from [106] Ireson, C.; Orr, S.; Jones, D.J.; Verschoyle, R.; Lim, C.K.; Luo,
vascular biology to biomarker discovery and risk prediction. Clin. J.L. Characterization of metabolites of f the chemopreventive agent
Chem., 2008, 54, 24-38. curcumin in human and rat hepatocytes and in the rat in vivo, and
[87] Murakami, Y.; Ishii, H.; Takeda, N.; Tanaka, S.; Machino, M.; Ito, evaluation of their ability to inhibit phorbol ester-induced prosta-
S.; Fujisawa, S. Comparative anti-inflammatory activities of cur- glandin E2 production. Cancer Res., 2001, 61, 1058-1064.
cumin and tetrahydrocurcumin based on the phenolic O-H bond [107] Garcea, G.; Jones, D.J.; Singh, R.; Dennison, A.R.; Farmer, P.B.;
dissociation enthalpy, ionization potential and quantum chemical Sharma, R.A. Detection of curcumin and its metabolites in hepatic
descriptor. Anticancer Res., 2008, 28, 699-707. tissue and portal blood of patients following oral administration.
[88] Jurenka, J.S. Anti-inflammatory properties of curcumin, a major Br. J. Cancer, 2004, 90, 1011-1015.
constituent of Curcuma longa: a review of preclinical and clinical [108] Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B.
research. Altern. Med. Rev., 2009, 14, 141-153. Bioavailability of curcumin: problems and promises. Mol. Pharm.,
[89] Aggarwal, B.B.; Sung, B. Pharmacological basis for the role of 2007, 4, 807-818.
curcumin in chronic diseases: an age old spice with modern targets. [109] Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srini-
Trends Pharmacol. Sci., 2009, 30, 85-94. vas, P.S. Influence of piperine on the pharmacokinetics of curcu-
[90] Gupta, S.C.; Kim, J.H.; Prasad, H.; Aggarwal, B.B. Regulation of min in animals and human volunteers. Planta Med., 1998, 64, 353-
survival, proliferation, invasion, angiogenesis, and metastasis, of 356.
tumor cells through modulation of inflammatory pathways by nu- [110] Li, L.; braiteh, F.S.; Kurzrock, R. Liposome-encapsulated curcu-
traceuticals. Cancer Metas. Rev., 2010, 29, 405-434. min: in vitro and in vivo effects on proliferation, apoptosis, signal-
[91] Singh, S.; Aggarwal, B.B. Activation of transcription factor NF- ing, and angiogenesis. Cancer, 2005, 104, 1322-1331.
kappa B is suppressed by curcumin (diferuloylmethane). J. Biol. [111] Sun, A.; Shoji, M.: Lu, Y.J.; Liotta, D.C.; Snyder, J.P. Synthesis of
Chem., 1995, 270, 24995-25000. EF24-tripeptide chloromethyl ketone: a novel curcumin-related
[92] Sethi, G.; Sung, B.; Aggarwal, B.B. Nucelar factor-kappaB activa- anticancer drug delivery system. J. Med. Chem., 2006, 49, 3153-
tion: from bench to bedside. Exp. Biol. Med., 2008, 233, 21-31. 3158.
[93] Ralhan, R.; Pandey, M.K.; Aggarwal, B.B. Nuclear factor-kappa B [112] Bisht, S.; Feldmann, G.; Soni, S.; Ravi, R.; Karikar, C.; Maitra, A.
links carcinogenic and chemopreventive agents. Front. Biosci., Polymeric nanoparticle-encapsulated curcumin (“nanocurcumin”):
2009, 1, 45-60. a novel strategy for human cancer therapy. J. Nanobiotechnol.,
[94] Aggarwal, B.B.; Kunnumakkara, A.B.; Harikumar, K.B.; Tharakan, 2007, 5, 3.
S.T.; Sung, B.; Anand, P. Potential of spice derived phytochemicals [113] Marczylo, T.H.; Verschoyle, R.D.; Cooke, D.N.; Morazzoni, P.;
for cancer prevention. Planta Med., 2008, 74, 1560-1569. Steward, W.P.; Gescher, A.J. Comparison of systematic availability
[95] Shishodia, S.; Amin, H.M.; Lai, R.; Aggarwal, B.B. Curcumin of curcumin with that of curcumin formulated with phosphatadiyl-
(diferuloylmethane) inhibits constitutive NF-kappaB activation, in- choline. Cancer Chemother. Pharmacol., 2007, 60, 171-177.
duced G1/S arrest, suppresses proliferation, and induces apoptosis [114] Anand, P.; Nair, H.B.; Sung, B.; Kunnumakkara, A.B.; Yadav,
in mantle cell lymphoma. Biochem. Pharmacol., 2005, 70, 700- V.R.; Tekmal, R.R.; Aggarwal, B.B. Design of curcumin-loaded
713. PLGA nanoparticles formulation with enhanced cellular uptake,
[96] Aggarwal, B.B.; Shishodia, S.; Takeda, Y.; Banerjee, S.; Newman, and increased bioactivity in vitro and superior bioavailability in
R.A.; Bueso-Ramos, C.E.; Price, J.E. Curcumin suppresses the vivo. Biochem. Pharmacol., 2010, 79, 330-338.
paclitaxel-induced nuclear factor-kappaB pathway in breast cancer [115] Nair, H.B.; Sung, B.; Yadav, V.R.; Kannappan, R.; Chaturvedi,
cells and inhibits lung metastasis of human breast cancer in nude M.M.; Aggarwal, B.B. Delivery of anti-inflammatory nutraceuti-
mice. Clin. Cancer Res., 2005, 11, 7490-7498. cals by nanoparticles for the prevention and treatment of cancer.
[97] Aggarwal S.; Ichikawa, H.; Takada, Y.; Sandur, S.K.; Shishodia, Biochem. Pharmacol., 2010, 80, 1833-1843.
S.; Aggarwal, B.B. Curcumin (diferuloylmethane) down-regulates
Curcumin and Liver Cancer Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 227

[116] Yadav, V.R.; Prasad, S.; Kannappan, R.; Ravindran, J.; Chaturvedi, [138] Cao, J., Jia, L.; Zhou, H.-M.; Liu, Y.; Zhong, L.-F. Mitochondrial
M.M.; Vaahtera, L.; Parkkinen, J.; Aggarwal, B.B. Cyclo-dextrin- and nuclear DNA damage induced by curcumin in human hepa-
complexed curcumin exhibits anti-inflammatory and antiprolifera- toma G2 cells. Toxicol. Sci., 2006, 91, 476-483.
tive activities superior to those of curcumin through higher cellular [139] Cao, J.; Liu, Y.; Jia, L.; Zhou, H.-M.; Kong, Y.; Yang, G.; Jiang,
uptake. Biochem. Pharmacol., 2010, 80, 1021-1032. L.P.; Li, Q.J.; Zhong, L.-F. Curcumin induces apoptosis through
[117] John, V.D.; Kuttan, G.; Krishnankutty, K. Anti-tumour studies of mitochondrial hyperpolarization and mtDNA damage in human he-
metal chelates of synthetic curcuminoids. J. Exp. Clin. Cancer patoma G2 cells. Free Rad. Biol. Med., 2007, 43, 968-975.
Res., 2002, 21, 219-224. [140] Roy, M.; Pear, W.S.; Aster, J.C. The multifaceted role of Notch in
[118] Padhye, S.; Yang, H.; Jamadar, A.; Cui, Q.C.; Chavan, D.; cancer. Curr. Opin. Genet. Dev., 2007, 17, 52-59.
Dominiak, K.; McKinney, J.; Banerjee, S.; Dou, Q.P.; Sarkar, F.H. [141] Ning, L.; Wentworth, L.; Chen, H.; Weber, S.M. Down-regulation
New difluoro Knoevenagel condensates of curcumin, their Schiff of notch1 signaling inhibits tumor growth in human hepatocellular
bases and copper complexes as proteasome inhibitors and apoptosis carcinoma. Am. J. Transl. Res., 2009, 1, 358-366.
inducers in cancer cells. Pharm. Res., 2009, 26, 1874-1880. [142] Simoni, D.; Rizzi, M.; Rondanin, R.; Baruchello, R.; Marchetti, P.;
[119] Leyon, P.V.; Kuttan, G. Studies on the role of some synthetic cur- Invidiata, F.P.; Labbozzetta, M.; Poma, P.; Carina, V.; Notarbar-
cuminoid derivatives in the inhibition of tumour specific angio- tolo, M.; Alaimo, A.; D’Alessandro, N. Antitumor effects of cur-
genesis. J. Exp. Clin. Cancer Res., 2003, 22, 77-83. cumin and structurally -diketone modified analogs on multidrug
[120] Bar-Sela, G.; Epelbaum, R.; Schaffer, M. Curcumin as an anti- resistant cancer cells. Bioorg. Med. Chem. Lett., 2008, 18, 845-849.
cancer agent: review of the gap between basic and clinical applica- [143] Yoysungneon, P.; Wirachwong, P.; Changtam, C.; Suksamrarn, A.;
tions. Curr. Med. Chem., 2010, 17, 190-197. Patumraj, S. Anti-cancer and anti-angiogenic effects of curcumin
[121] Aggarwal, B.B.; Takada, Y.; Oommen, O.V. From chemopreven- and tetrahydrocurcumin on implanted hepatocellular carcinoma in
tion to chemotherapy: common targets and common goals. Exp. nude mice. World J. Gastroenterol., 2008, 14, 2003-2009.
Opin. Invest. Drugs, 2004, 13, 1327-1338. [144] Xiao, J.; Chu, Y.; Hu, K.; Wan, J.; Huang, Y.; Jiang, C.; Liang, G.;
[122] Shishodia, S.; Sethi, G.; Aggarwal, B.B. Curcumin: getting back to Li, X. Synthesis and biological analysis of a new curcumin ana-
the roots. Ann. N.Y. Acad. Sci., 2005, 1056, 206-217. logue for enhanced anti-tumor activity in HepG2 cells. Oncol. Rep.,
[123] Burgos-Morón, E.; Calderón-Montano, J.M.; Salvador, J.; Robles, 2010, 23, 1435-1441.
A.; López-Lázaro, M. The dark side of curcumin. Int. J. Cancer, [145] Notarbartolo, M.; Poma, P.; Perri, D.; Dusonchet, L.; Cervello, M.;
2010, 126, 1771-1775. D’Alessandro, N. Antitumor effects of curcumin, alone or in com-
[124] Hata, M; Sasaki, E.; ota, M.; Fujimoto, K.; Yajima, J.; Shichida, T. bination with cisplatin or doxorubicin, on human hepatic cancer
Allergic contact dermatitis from curcumin (turmeric). Contact cells. Analysis of their possible relationship to changes in NF-kB
Dermat., 1997, 36, 107-108. activation levels and in IAP gene expression. Cancer Lett., 2005,
[125] Swierczynska, M.K.; Krecisz, B. occupational skin changes in 224, 53-65.
persons working in contact with food spices. Med. Pr., 1998, 49, [146] Bishayee, A.; Chatterjee, M. Inhibitory effect of vanadium on rat
187-190. liver carcinogenesis initiated with diethylnotisoamine and pro-
[126] Frank, N.; Knauft, J.; Amelung, F.; Nair, J.; Wesch, H.; Bartsch, H. moted with phenobarbital. Br. J. Cancer, 1995, 71, 1214-1220.
No prevention of liver and kidney tumors in Long-Evans Cinna- [147] Bishayee, A; Dhir, N. Resveratrol-mediated chemoprevention of
mon rats by dietary curcumin, but inhibition at other sites and of diethylnitrosoamine-initiated hepatocarcinogenesis: inhibition of
metastases. Mutat. Res., 2003, 523, 127-135. cell proliferation and induction of apoptosis. Chem.-Biol. Interact.,
[127] Liu, R.H. Potential synergy of phytochemicals in cancer preven- 2009, 10, 131-144.
tion: mechanism of action. J. Nutr., 2004, 3479S-3485S. [148] Darvesh, A.S.; Bishayee, A. Selenium in the prevention and treat-
[128] de Kok, T.M.; van Breda, S.G.; Manson, M.M. Mechanisms of ment of hepatocellular carcinoma. Anti-Cancer Agents Med.
combined action of different chemopreventive dietary compounds. Chem., 2010, 10, 338-345.
Eur. J. Nutr., 2008, 47, 51-59. [149] Soni, K.B.; Lahiri, M.; Chackradeo, P.; Bhide, S.V.; Kuttan, R.
[129] Korkina, L.G.; de Luca, C.; Kostyuk, V.A.; Pastore, S. Plant poly- Protective effect of food additives on aflatoxin-induced mutagenic-
phenols and tumors: from mechanisms to therapies, prevention and ity and hepatocarcinogenicity. Cancer Lett., 1997, 115, 129-133.
protection against toxicity of anti-cancer treatments. Curr. Med. [150] Bishayee, A.; Sarkar, A.; Chatterjee, M. Further evidence for che-
Chem., 2009, 16, 3943-3965. mopreventive potential of beta-carotene against experimental car-
[130] Sheepens, A., Tan, K., Paxton, J.W. Improving the oral bioavail- cinogenesis: diethylnitrosamine-initiated and Phenobarbital-
ability of beneficial polyphenols through designed synergies. Genes promoted hepatocarcinogenesis prevented more effected by beta-
Nutr., 2010, 5, 75-87. carotene than by retinoic acid. Nutr. Cancer, 2000, 37, 89-98.
[131] Sandur, S.K.; Pandey, M.K.; Sung, B.; Ahn, K.S.; Murakami, A.; [151] Chuang, S.E.; Cheng, A.-L.; Lin, J.-K.; Kuo, M.-L. Inhibition by
Sethi, G.; Limtrakul, V.; Badmaev, V., Aggarwal, B.B. Curcumin, curcumin of diethylnitrosamine-induced hepatic hyperplasia, in-
demethoxycurcumin, bisdemethoxycurcumin, tetrahydrocurcumin, flammation, cellular gene products and cell-cycle-related proteins
and tumerones differentially regulate anti-inflammatory and anti- in rats. Food Chem. Toxicol., 2000, 38, 991-995.
proliferative responses through a ROS-independent mechanism. [152] Chuang, S.E.; Kuo, M.L.; Hsu, C.H.; Chen, C.R.; Lin, J.K.; Lai,
Carcinogenesis, 2007, 8, 1765-1773. G.M.; Hsieh, C.Y.; Cheng, A.L. Curcumin-containing diet inhibits
[132] Lin, L.-I.; Ke, Y.-F.; Ko, Y.-C.; Lin, J.-K. Curcumin inhibits SK- diethylnitrosamine-induced murine hepatocarcinogenesis. Carcino-
Hep-1 hepatocellular carcinoma cell invasion in vitro and sup- genesis, 2000, 21, 331-335.
presses matric metalloproteinase-9 secretion. Oncology, 1998, 55, [153] Thapliyal, R.; Naresh, K.N.; Rao, K.V.K.; Maru, G.B. Inhibition of
349-353. nitrosoamine-induced hepatocarcinogenesis by dietary turmeric in
[133] Ohashi, Y.; Tsuchiya, Y.; Koizumi, K.; Sakurai, H.; Saiki, Y. Pre- rats. Toxicol. Lett., 2003, 139, 45-54.
vention of intrahepatic metastasis by curcumin in an orthotopic im- [154] Shukla, Y.; Arora, A. Suppression of altered hepatic foci develop-
plantation model. Oncology, 2003, 65, 250-258. ment by curcumin in Wistar rats. Nutr. Cancer, 2003, 45, 53-59.
[134] Kang, J.; Chen, J.; Shi, Y.; Jia, J.; Zhang, Y. Curcumin-induced [155] Sreepriya, M; Bali, G. Chemopreventive effects of embelin and
histone hypoacetylation: the role of reactive oxygen species. Bio- curcumin against N-nitrosodiethylamine/phenobarbital-induced he-
chem. Pharmacol., 2005, 69, 1205-1213. patocarcinogenesis in Wistar rats. Fitoterapia, 2005, 76, 549-555.
[135] Cui, S.-X.; Qu, X.-J.; Xie, Y.-Y.; Zhou, L.; Nakata, M.; Makuuchi, [156] Sreepriya, M.; Bali, G. Effects of administration of embelin and
M.; Tang, W. Curcumin inhibits telomerase activity in human can- curcumin on lipid peroxidation, hepatic glutathione antioxidant de-
cer cell lines. Int. J. Mol. Med., 2006, 18, 227-231. fense and hematopoietic system during N-nitrosodiethylamine/
[136] Jia, L.; Wang, H.; Qu, S.; Miao, X.; Zhang, J. CD147 regulates phenobarbital-induced hepatocarcinogenesis in Wistar rats. Mol.
vascular endothelial growth factor–A expression, tumorigenicity, Cell. Biochem., 2006, 284, 49-55.
and chemosensitivity to curcumin in hepatocellular carcinoma. [157] Huang, A.-C.; Lin, S.-Y.; Su, C.-C.; Lin, S.-S.; Ho, C.-C.; Hsia, T.-
Life, 2008, 60, 57-63. C.; Chiu, T.-H.; Yu, C.-S.; Ip, S.W.; Lin, T.-P.; Chung, J.-G. Ef-
[137] Wang, W.-Z.; Cheng, J.; Luo, J.; Zhuang, S.-M. Abrogation of fects of curcumin on N-bis(2-hydroxypropyl) nitrosamine (DHPN)-
G2/M arrest sensitizes curcumin-resistant hepatoma cells to apop- induced lung and liver tumorigenesis in BALB/c mice in vivo. In
tosis. F.E.B.S. Lett., 2008, 582, 2689-2695. Vivo, 2008, 22, 781-786.
228 Current Pharmaceutical Biotechnology, 2012, Vol. 13, No. 1 Darvesh et al.

[158] Takaba, K.; Hirose, M.; Yoshida, Y.; Kimura, J.; Ito, N.; Shirai, T. tetrachlorobiphenyl (PCB-77). Food Chem. Toxicol., 2008, 46,
Effects of n-trtricontane-16,18-dione, curcumin, chlorophyllin, 3467-3474.
dihydroguaiaretic acid, tannic acid and phytic acid on the initiation [162] Busquets, S.; Carb, N.; Almendro, V.; Quiles, M.T.; Lpez-
stage in a rat multi-organ carcinogenesis model. Cancer Lett., Soriano, F.J.; Argilés, J.M. Curcumin, a natural product present in
1997, 113, 39-46. turmeric, decreases tumor growth but does not behave as an anti-
[159] Hirose, M.; Takahashi, S.; Ogawa, K.; Futakuchi, M.; Shirai, T.; cachectic compound in a rat model. Cancer Lett., 2001, 167, 33-38.
Shibutani, M.; Uneyama, C.; Toyoda, K.; Iwata, H. Chemopreven- [163] Yoysungnoen, P.; Wirachwong, P.; Bhattarakosol, P.; Niimi, H.;
tion of heterocyclic amine-induced carcinogenesis by phenolic Patumraj, S. Antiangiogenic activity of curcumin in hepatocellular
compounds in rats. Cancer Lett., 1999, 143, 173-178. carcinoma cells implanted nude mice. Clin. Hemor. Microcirc.,
[160] Hirose, M.; Takahashi, S.; Ogawa, K.; Futakuchi, M.; Shirai. Phe- 2005, 33, 127-135.
nolics: Blocking agents for heterocyclic amine-induced carcino- [164] Yoysungnoen, P.; Wirachwong, P.; Bhattarakosol, P.; Niimi, H.;
genesis. Food Chem. Toxicol., 1999, 37, 985-992. Patumraj, S. Effects of curcumin on tumor angiogenesis and bio-
[161] Tharappel, J.C.; Lehmler, H.-J.; Srinivasan, C.; Robertson, L.W.; markers, COX-2 and VEGF, in hepatocellular carcinoma cell-
Spear, B.T.; Glauert, H.P. Effect of antioxidant phytochemicals on implanted carcinoma cell-implanted nude mice. Clin. Hemor. Mi-
the hepatic tumor promoting activity of 3,3', 4,4'- crocirc., 2006, 34, 109-115.

Received: September 03, 2010 Revised: September 29, 2010 Accepted: October 01, 2010

View publication stats

You might also like