Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Current Cancer Drug Targets, 2005, 5, 117-129 117

Induction of Apoptosis by Curcumin and Its Implications for Cancer Therapy


D. Karunagaran*, R. Rashmi and T. R. Santhosh Kumar

Cancer Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014 India
Abstract: Curcumin (diferuloyl methane), the yellow pigment in turmeric (Curcuma longa), is a potent
chemopreventive agent that inhibits cancer cells proliferation by arresting them at various phases of the cell
cycle depending upon the cell type. Curcumin-induced apoptosis mainly involves the mitochondria-mediated
pathway in various cancer cells of different tissues of origin. In some cell types like thymocytes, curcumin
induces apoptosis-like changes whereas in many other normal and primary cells curcumin is either inactive or
inhibits proliferation, but does not appear to induce apoptosis. These together with reports that curcumin
protects cells against apoptosis induced by other agents, underscore the need for further understanding of the
multiple mechanisms of cell death unleashed by curcumin. Tumor cells often evade apoptosis by expressing
several antiapoptotic proteins, down-regulation and mutation of proapoptotic genes and alterations in
signaling pathways that give them survival advantage and thereby allow them to resist therapy-induced
apoptosis. Many researchers including ourselves, have demonstrated the involvement of several pro and
antiapoptotic molecules in curcumin-induced apoptosis, and ways to sensitize chemoresistant cancer cells to
curcumin treatment. This review describes the mechanisms of curcumin-induced apoptosis currently known,
and suggests several potential strategies that include down-regulation of antiapoptotic proteins by antisense
oligonucleotides, use of proapoptotic peptides and combination therapy, and other novel approaches against
chemoresistant tumors. Several factors including pharmacological safety, scope for improvement of structure
and function of curcumin and its ability to attack multiple targets are in favor of curcumin being developed as a
drug for prevention and therapy of various cancers.
Keywords: Apoptosis, curcumin, Bcl-2, hsp 70, Bcl-XL, Ku70, antisense, cancer.

INTRODUCTION carcinogenesis may also promote apoptosis, thereby


producing selective pressure to override apoptosis. Finally,
Curcumin (diferuloyl methane) is a naturally occurring
it is well documented that most cytotoxic anticancer agents
yellow pigment isolated from the rhizome of the perennial
induce apoptosis, raising the intriguing possibility that
herb Curcuma longa that has been cultivated for centuries in
defects in apoptotic programs reduce treatment sensitivity
several Asian countries (India, Nepal, China and Indonesia)
and contribute to treatment failure. Hence, it is not
[1, 2]. Turmeric, the powdered form of the rhizome, is
surprising that several laboratories consider apoptosis as the
widely used in these countries in foods (spice and coloring
major mechanism by which most chemopreventive and
agent), traditional medicines (to treat a variety of ailments),
chemotherapeutic agents act in arresting cell cycle
textile and pharmaceutical industries as well as in Hindu
progression and/or triggering programmed cell death [13-15].
religious ceremonies [3-5]. Curcumin possesses
Apoptosis is accompanied by certain well-characterized
antiinflammatory and antioxidant properties [6-8]. It is also
cellular changes like membrane blebbing, mitochondrial
a potent chemopreventive agent inhibiting tumor promotion
membrane potential alteration, fragmentation of DNA into
against skin, oral, intestinal and colon carcinogenesis [9-11].
oligonucleosomal fragments and activation of cysteine
Many studies using curcumin focused on its
proteases called caspases finally culminating in cell death
antiinflammatory, antioxidant, anticarcinogenic, antiviral,
[16]. Curcumin has been shown to induce apoptosis in a
and antiinfective activities. In addition, the wound healing
variety of cells by several workers in the last decade, but no
and detoxifying properties of curcumin have also received
comprehensive review focused on the antiproliferative and
considerable attention [12]. Over the last decade, remarkable
apoptosis-inducing activities of curcumin is available so far
advances in our understanding of cancer biology have led us
although others have reviewed various biological activities
to the realization that apoptosis and the genes that control it
of curcumin [8, 12, 17-20]. Much progress has been made in
have a profound effect on the malignant phenotype although
understanding the molecular mechanism of apoptosis
apoptosis or programmed cell death is primarily a cell
induced by curcumin in cancer cells and its safety records are
suicide program critical for development, tissue
conducive for its therapeutic use at the preclinical/clinical
homeostasis, and protection against pathogens. It is now
stage for various cancers. At the same time, it is apparent
known that some oncogenic mutations disrupt apoptosis,
that most of the currently available drugs are not effective
leading to tumor initiation, progression or metastasis. In
against a subset of solid tumors because of the emergence of
contrast, other oncogenic changes during multistage
acquired chemoresistance. It will be quite interesting to see
whether the new experimental drugs like curcumin can be
*Address correspondence to this author at the Cancer Biology Laboratory, directed against such cancers with necessary interventional
Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 approaches. Our recent insight into how chemoresistance
014 India; Tel: 91-471-2347975; Fax: 91-471-2348096; E-mail: develops and the identification of new players in acquired
dkarunagaran@hotmail.com drug resistance offer great potential to design appropriate

1568-0096/05 $50.00+.00 © 2005 Bentham Science Publishers Ltd.


118 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

interventional strategies to render curcumin-based therapy umbilical vein endothelial cells (HUVEC) as revealed by
more effective. The review places emphasis on novel [3H]-thymidine incorporation in a dose-dependent manner
apoptosis-based potential targets that can be exploited to without significantly affecting the viability of the cells.
sensitize drug-resistant cancers with this drug. Addition of curcumin to HUVEC resulted in an
accumulation of > 46% of the cells in early S-phase, as
determined by flow cytometric analysis. Curcumin caused a
CHEMICAL STRUCTURE AND METABOLOITES significant loss of thymidine kinase activity, which may be
OF CURCUMIN one of the possible mechanism(s) for the inhibition of DNA
Commercial grade curcumin (77% curcumin, 17% synthesis activity of HUVEC by curcumin [36].
demethoxycurcumin and 3% bisdemethoxycurcumin) In response to lower concentrations of curcumin, mouse
contains pure curcumin shown in Fig. (1) with a molecular embryonal carcinoma PCC4 cells ceased to proliferate and
weight of 368.37 and demethoxycurcumin, which are showed cell cycle arrest at G1 phase after 4 hours of
equally potent as inhibitors of phorbol ester-induced tumor treatment, followed by their differentiation, which is
promotion in mouse skin whereas bisdemethoxycurcumin is characterized by an increase in nuclear/cytoplasmic ratio [39].
relatively less active [21]. Curcumin is insoluble in water Curcumin inhibits the proliferation of head and neck
but soluble in ethanol, dimethylsulfoxide and other organic squamous cell carcinoma MDA 686LN cells by arresting the
solvents. When curcumin was incubated in 0.1M phosphate cell cycle in G1 /S phase [30]. In vascular smooth-muscle
buffer and serum-free medium, pH 7.2 at 370C, about 90% cells, this compound causes a G0/G1 cell-cycle arrest [38].
decomposed within 30 min. However, in cell culture Gautam et al. investigated the antiproliferative effects of
medium containing 10% fetal calf serum about 50% of curcumin against a variety of transformed and
curcumin still remained stable after 8h. Trans-6- (4'-hydroxy- nontransformed cell types. At equimolar concentrations
3'-methoxyphenyl)-2,4-dioxo-5-hexenal was predicted to be ranging from 6.25 to 50 µM, curcumin inhibited DNA
the major degradation product; vanillin, ferulic acid and synthesis in five leukemia lines, three nontransformed
feruloyl methane were identified as minor degradation hematopoietic progenitor cell populations, and four
products and the amount of vanillin increased with nontransformed fibroblastic cell lines in a concentration-
incubation time [22]. Curcumin has poor bioavailability in dependent manner. Curcumin also inhibited the cellular
animals and humans due to its poor absorption and rapid growth of both transformed and nontransformed cells in
metabolism in the liver and intestinal wall [23-26]. The clonogenic assays. Without discriminating between
major metabolites in suspensions of human and rat transformed or nontransformed cells, the inhibition of cell
hepatocytes were identified as hexahydrocurcumin and proliferation by curcumin was not always associated with
hexahydrocurcuminol [25]. programmed cell death [44]. Similarly inhibition of cell
O O proliferation by curcumin in many breast cancer cells was
H3CO OCH3 not associated with apoptosis [41, 45]. Khar et al reported
that curcumin had no effect on the growth of nontransformed
cell lines, which showed neither superoxide generation nor
HO OH the induction of a stress response [46]. Interestingly,
Fig. (1). Chemical structure of curcumin curcumin had no growth-inhibitory effect on normal rat
hepatocytes and normal lung fibroblasts [47-49]. Similarly,
curcumin did not inhibit growth in primary culture of mouse
ANTIPROLIFERATIVE ACTIONS OF CURCUMIN embryonic fibroblast C3H 10T1/2, rat embryonic fibroblast,
and human foreskin fibroblast cells in a concentration- and
Cell cycle regulatory proteins and checkpoints are
time-dependent manner [43].
downstream elements of cellular signaling cascades crucial
for cell proliferation. Curcumin inhibits the proliferation of a The mechanisms by which many cell types are arrested in
wide variety of cancer cells derived from breast, blood, the G2/M-phase of the cell cycle upon curcumin treatment
colon, liver, kidney, prostate and skin through its effects on are not known. MCF-7 breast cancer cells treated with 10-20
different phases of cell cycle (Table 1). Human acute µM curcumin for 24-48 h are arrested in M-phase, and DNA
myeloid leukemia cells (HL-60) are highly susceptible to synthesis is almost completely inhibited, and the arrested
curcumin-mediated inhibition of cell proliferation in a dose- mitotic cells exhibit monopolar spindles, and chromosomes
and time-dependent manner leading to typical apoptosis. In do not undergo normal anaphase movements. After 48 h,
HL-60 cells treated with 25 µM curcumin for 48 h, 60.71± most cells eventually leave M-phase, and many form
1.20% cells were arrested in G2 /M phase of cell cycle at multiple micronuclei instead of individual daughter nuclei.
first, then in G0 /G 1 phase and DNA synthesis was halted These observations indicate that curcumin induces the
resulting in apoptosis instead of differentiation [27] with assembly of aberrant, monopolar mitotic spindles that are
estimated IC50 values of 19.5 µM [42]. In HT-29 (human impaired in their ability to segregate chromosomes. The
colon cancer) cells curcumin causes cell cycle arrest in G2/M production of cells with extensive micronucleation after
phase but not apoptosis [33] whereas another study reported curcumin treatment suggests that at least some of the
induction of apoptosis by curcumin in these cells [43]. cytostatic effects of this phytochemical are due to its ability
Curcumin inhibits cell proliferation in gastric (KATO-III) to disrupt normal mitosis, and raises the possibility that
and colon (HCT-116) cancer cells and induces G2/M arrest in curcumin may promote genetic instability under some
HCT-116 cells and cyclin B level remains unaffected, circumstances [50]. Antiproliferative effects of curcumin thus
whereas cyclin D and E levels decline with curcumin in both appear to be cell-type dependent and presumably the
cell lines [32]. Curcumin inhibits DNA synthesis in human experimental conditions and/or designs employed by
Implications for Cancer Therapy Current Cancer Drug Targets, 2005, Vol. 5, No. 2 119

Table 1. Effects of Curcumin on the Cell Cycle

Cell type Arrested cell cycle phase Cell cycle-related mechanisms References

HL-60 (Human acute myeloid G2/M first, then in G0/G1 Inhibition of DNA synthesis [27]
leukemia) phase

Human multiple myeloma cells G1/S Down regulation of cyclin D1, inhibition of IKK and NF-κB [28]

CA46 cells (human Burkitt's G0/G1 or G2/M and S Inhibition of DNA synthesis [29]
lymphoma)

MDA 686LN (human head and neck G1/S Down regulation of cyclin D1, inhibition of IKK and NF-κB [30]
squamous cell carcinoma)

HCT-116 (human colon cancer) G2/M P53-and p21-independent [31]


Down regulation of cyclin D and E but not B [32]
Activation of cdc2

HT-29 and HCT-15 (human colon G2/M Prostaglandin independent [33]


adenocarcinoma)

COLO205 (colorectal carcinoma) G1 Inhibition of Ca2+ dependent endonuclease, reduction of p53 [34]
gene expression

Lovo (colon cancer) S, G2/M inhibition of DNA synthesis [35]

HUVEC (human umbilical vein S inhibition of DNA synthesis [36]


endothelial cells)

ECV304 (immortalized human G0/G1 and/or G2/M up-regulation of p21WAF1/CIP1, p27KIP1, and p53 [37]
umbilical vein endothelial cells)

A7r5 (Rat aortic smooth muscle cell G0/G1 and S inhibition of protein tyrosine kinase activity, protein kinase C [38]
line) activity, c-myc mRNA expression and bcl-2 mRNA expression

PCC4 (mouse embryonal carcinoma G1 differentiation characterized by increase of [39]


cells) nuclear/cytoplasmic ratio

MCF-7/TH (multidrug-resistant human G2/M and subG0/G1 Reduction in the expression of Ki67, PCNA and p53 mRNAs [40]
breast carcinoma)

Human breast cancer cells S and G2/M inhibition of ornithine decarboxylase activity [41]

different researchers also contribute to these differences at (Fas associated death domain) and proteolytic activation of
least, in part. Questions on the stability of curcumin procaspase 8, which then induces the activation of
affecting its availability, formation of metabolites and their downstream effector caspases like caspase 3, 6 and 7 [13,
relative contribution for the observed effects of curcumin 51]. In the intrinsic or mitochondrial pathway, cytochrome c
largely remain unanswered. is released from mitochondria into the cytosol resulting in
its high affinity interaction with apoptotic protease
activating factor-1 (Apaf-1) and subsequent activation of
MECHANISMS OF CURCUMIN-INDUCED caspase 9 [52]. The active caspase 9 then activates
APOPTOSIS downstream effector caspases that mediate the execution
Curcumin is known to induce apoptosis in numerous phase exhibiting characteristic features of apoptosis, many of
animal and human cell lines established from malignancies which reflect the proteolytic cleavage of various intracellular
like leukemia, melanoma, breast, lung, prostate, colon, proteins ensuring peaceful elimination of the cell [53-55].
renal, hepatocellular and ovarian carcinomas (Table 2). Apart from cytochrome c, the release of second mitochondria
Curcumin also induces typical characteristics of apoptosis derived activator of caspase (Smac) from mitochondria also
like cell shrinkage, chromatin condensation, and DNA ensures continued caspase activation [56]. Internucleosomal
fragmentation in immortalized mouse embryo fibroblast NIH degradation, a characteristic biochemical feature of apoptosis,
3T3 cells in a concentration- and time-dependent manner results from the cleavage of ICAD (inhibitor of caspase
[43]. Two distinct apoptotic signaling pathways have been activated DNase or DNA fragmentation factor45/DFF45),
characterized in mammalian cells. One of these pathways which releases an endonuclease called CAD (caspase
often called extrinsic or death receptor pathway involves activated DNase).
cytosolic domains of members of the TNFR (tumor necrosis Potential implications in chemoresistance mechanisms
factor receptor) family such as CD 95/Fas and TRAIL-R1 make it highly relevant to ask the question of whether
(TNF-related apoptosis inducing ligand-R1) or TRAIL-R2, curcumin induces apoptosis through extrinsic or intrinsic
which recruit procaspase 8. Ligation of these receptors leads apoptotic pathways specifically in different cancer cells.
to oligomerization, recruitment of cytosolic adaptor FADD
120 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

Table 2. Apoptotic Mechanisms Activated by Curcumin

Cell lines Apoptotic changes induced by Reference Cell lines Apoptotic changes induced Reference
curcumin by curcumin
DNA fragmentation [57] JNK-dependent apoptosis [62]
HL-60 (acute Caspase-3 activation PARP cleavage
myeloid leukemic DNA fragmentation [58] Human colon Activation of casapse-3 and [32]
cell line, human) Cytochrome c release cancer cell caspase-8
Activation of caspase-3 but not [59] lines Down regulation of Bcl-XL [63,64]
caspase-8 Nuclear condensation
Chromatin condensation DNA [60] Release of cytochrome c, [31]
fragmentation AIF and Smac
Down regulation of Mcl-1 [61] Activation of caspase-3,
Up regulation of Bax and Bak caspase-8, caspase-9
Up regulation of p27/kipl, p21/wafl Loss of mitochondrial
and pRbp transmembrane potential
Down regulation of cyclin D3 Cleavage of PARP and
Activation of caspase-8 and caspase- DFF45
3 p53- and p21-independent
PARP cleavage apoptosis
Bid cleavage Impairment of Wnt signaling
Cox-2 inhibition and cell-cell adhesion
Cytochrome c release pathways
Increase in p53 and Bax proteins and [65] Downregulation of NF-κB
Breast cancer cells p53DNA-binding activity Melanoma activation, iNOS and DNA- [68]
(human) Induction of redox signaling [66] (human) dependent protein kinase
Inhibition of H-ras-induced invasive catalytic subunit expression [69]
phenotype [67] Up regulation of p53, p21
Downregulation of Bcl-2 and MMP-2 (CIP1), p27 (Kip1) and
Upregulation of Bax checkpoint kinase 2
Inhibition of telomerase Activation of caspases-3 and
-8 but not caspase-9
Fas receptor aggregation in a
FasL-independent manner
Downregulation of NF-κB
activation and XIAP
expression
p53-dependent apoptosis Down regulation of of egr-1,
Human basal cell DNA fragmentation [70] BKS-2 c-myc, bcl-XL and p53 [71]
carcinoma Up regulation of nuclear p53 protein, (immature B expression
p21 (CIP1/WAF1) and Gadd45 cell
lymphoma)
Stage 4 MYCN- Up-regulation of p53, p21(WAF-
amplified 1/CIP-1) and Bax expression [72] K562 (human Activation of caspases-8 and [73]
neuroblastoma cell leukemia) 9
lines Inhibition of glutathione S-
transferase P1-1 expression
Generation of reactive oxygen
AK-5 (rat intermediates [74,75] MDA 686LN IKK and NF-κB inhibition [30]
histiocytoma) Activation of caspase-3 (human head Reduced expression of Bcl-2,
Loss of mitochondrial membrane and neck cyclin D1, IL-6, COX-2 and
potential squamous cell MMP-9
Cytochrome c release carcinoma)
DNA fragmentation Increased caspase-3
A7r5 (Rat aortic Inhibition of protein tyrosine kinase [38] Vγ9vδ2 T cells expression in nucleus [76]
smooth muscle cell and PKC activities and c-myc and PARP cleavage
line) bcl-2 mRNA expression. AIF translocation to nucleus
Activation of caspase 3
CA46 cells (human DNA fragmentation Decreased [29] Human renal Cleavage of phospholipase [77]
Burkitt's lymphoma) protein and mRNA expression of c- Caki cells C-gamma1
myc, bcl-2, mutant-type p53 Release of cytochrome c
Increased Fas protein and mRNA DNA fragmentation
Dephosphorylation of Akt
Down-regulation of Bcl-2,
Bcl-XL and IAP proteins
Up-regulating Caspase-3 and down- p53-independent mechanism
A2780 (human regulating expression of NF-κB [78] Human lung Decrease in expression of [48]
ovarian cancer) cancer cells p53, bcl-2, and bcl-XL

DNA fragmentation Down regulation of the


Ehrilich’s Ascites Cytochrome c release Activation [79] Human expression of Bcl-2 and Bcl- [80]
Carcinoma cells of caspase 3 prostate xL
Up regulation of Bax cancer cells Activation of caspase-3 and
caspase-8
Implications for Cancer Therapy Current Cancer Drug Targets, 2005, Vol. 5, No. 2 121

Curcumin appears to induce apoptosis in human melanoma both p53 proficient (A549) and p53 null mutant (H1299)
cells through a Fas receptor/caspase-8 pathway and the lung cancer cell lines with a decrease in expression of p53
expression of dominant negative FADD significantly suggesting a p53-independent induction of apoptosis in lung
inhibits curcumin-induced apoptosis [69]. In gastric (KATO- cancer cells [48]. Curcumin treatment causes a reduction in
III) and colon (HCT-116) cancer cells curcumin caused the expression of Ki67, PCNA, and p53 mRNAs in MCF-
cleavage of poly (ADP) ribose polymerase (PARP, a 7/TH, a multidrug-resistant human breast cancer cell line
substrate of caspase-3), activation of caspase-3 and caspase-8 [40]. However, curcumin also induces nuclear p53 protein
initiating the Fas signaling pathway of apoptosis [32]. and its downstream targets, including p21 (CIP1/WAF1)
Induction of Fas ligand in a few tumor cells [46] and and Gadd45 in human basal cell carcinoma cells [70].
increased expression of Fas protein and mRNA in Burkitt's Increase in p53 as well as its DNA-binding activity occurs
lymphoma cells occur upon treatment with curcumin [29]. during curcumin-induced apoptosis of MCF-7 (human breast
To show conclusively that curcumin acts through Fas cancer) cells [65]. Experiments using p53-null MDAH041
signaling, synthesis of FasL should be evident before the cells as well as low and high p53-expressing TR9-7 cells
cells become apoptotic and cytotoxicity should be inhibited also support a p53-dependent pathway for curcumin induced
by manipulations that block FasL-Fas and FADD-procaspase apoptosis in tumor cells [65]. These data suggest that the
8 interactions and cells that lack Fas should fail to respond p53-associated signaling pathway is differentially regulated
to the drug. Unfortunately, such rigorous evidences are by curcumin in a cell type-dependent manner to induce
mostly lacking on the involvement of Fas signaling in apoptosis.
response to curcumin.
Oxidative stress signals induced by the formation of
Curcumin induces membrane permeability, swelling, reactive oxygen species (ROS) and glutathione depletion are
loss of membrane potential and inhibition of ATP synthesis also considered as important activators of apoptosis [13].
mediated by the opening of the permeability transition pore Being lipophilic, curcumin can collapse transmembrane
in isolated rat liver mitochondria [81]. Activation of caspase mitochondrial potential and increase permeability for proton
3 and cleavage of PARP and Bid, release of mitochondrial and interfere with energy the coupling system in the
cytochrome c and activation of caspase 8 (blocked by mitochondria. As a consequence of energy dissipation in
dominant negative FLICE but not by dominant negative mitochondria, cells are likely to suffer from increased levels
FADD) in response to curcumin (25µM) are in favor of of ROS and, sensitivity of many tumor cells to curcumin
mitochondrial pathway for curcumin-induced apoptosis of correlates with the generation of superoxide radicals [46].
HL-60 cells [61]. Surprisingly another report found no Depletion of glutathione by buthionine sulfoximine resulted
activation of caspase 8 in the presence of curcumin (50 µM) in the increased generation of ROS, thereby further
in HL-60 cells [58]. It is not clear whether this striking sensitizing the tumor cells to curcumin [47]. In contrast,
difference is due to the different concentrations of curcumin curcumin is a potent scavenger of a variety of ROS
and/or other experimental conditions used by these including superoxide anion, hydroxyl radical, singlet
researchers. From the available data (Table 2) it appears that oxygen, nitric oxide and peroxynitrite [85, 86]. Curcumin
curcumin-induces apoptosis of many tumor cells mainly by also enhances the activities of antioxidant enzymes [87] and
the mitochondrial pathway and more studies are needed to increases glutathione levels [88], which is expected to
implicate or rule out a role for the extrinsic pathway in protect cells by preventing thiol depletion occurring during
curcumin-induced apoptosis. A mitochondrial flavoprotein apoptosis. In addition, curcumin exhibits both pro- and
called apoptosis-inducing factor (AIF) translocates to the antioxidant activities and the prooxidant DNA cleavage
nucleus when apoptosis is induced and recombinant AIF activity is the consequence of binding of Cu (II) to various
causes chromatin condensation in isolated nuclei and large- sites on the curcumin molecule [89]. Other well-known
scale fragmentation of DNA independent of caspase antioxidants such as N-acetyl L-cysteine, L-ascorbic acid,
activation [82]. Similarly a mitochondrion-specific nuclease alpha-tocopherol, catalase and superoxide dismutase have all
known as endonuclease G translocates to the nucleus during prevented curcumin-induced apoptosis effectively in HL-60
apoptosis and cleaves chromatin DNA into nucleosomal cells [90]. Due to the complexity of redox regulation by
fragments independently of caspases [83]. There is paucity of curcumin as described above, it is very difficult to provide a
information on the role of curcumin in regulating the release mechanistic basis for the induction of apoptosis by curcumin
of caspase-independent proapoptogenic factors such as AIF by this mechanism at the moment.
and endonuclease G although a few laboratories including
Curcumin (50 µM) induces apoptosis-like cell death in
ours have shown the release of AIF in response to curcumin
quiescent rat thymocyte and splenocytes, human peripheral
[63, 64, 76, 84]. It is hoped that these gaps will be filled
blood lymphocytes, proliferating IL-2-dependent T-cells,
soon and as the biochemical and molecular complexities of
human leukemic (MOLT-4) cells and mouse leukemic
the apoptotic pathways are elucidated, new molecular targets
(L1210) cells, and only in HL-60 cells classical apoptosis
for curcumin will be identified.
occurs [57]. Similarly curcumin-induced changes in nuclear
Alteration of p53 (tumor suppressor) is the most morphology, DNA fragmentation, the extent and time-course
common mutation in human cancer, and it functions as a of membrane dynamics are different from typical apoptosis
transcription factor regulating genes (p53 can up regulate induced by dexamethasone in rat thymocytes [91].
Bax and Fas) involved in cell cycle arrest, DNA repair and Apoptosis induced by curcumin in Jurkat cells also differs
apoptosis. Curcumin treatment causes p53- and p21- from "classical apoptosis" by the lack of mitochondrial
independent cell cycle arrest and apoptosis in p53 (+/+), p53 depolarization and of the involvement of caspases [92, 93].
(-/-) and p21 (-/-) derivatives of HCT-116 human colon Jaruga et al. reported that curcumin expands human
cancer cells [31]. In addition, curcumin induces apoptosis in erythrocyte membrane inducing echinocytosis accompanied
122 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

by transient exposure of phosphatidylserine and lipid 2 and Bcl-XL perform their antiapoptotic function by
fluidity changes [94]. These authors caution researchers that inhibiting the release of cytochrome c from mitochondria by
such nonspecific apoptosis-independent effects of curcumin preventing the translocation or activation of Bax or Bak
on cellular membranes would produce artifacts of apoptosis [100, 101]. Bid is a substrate for caspase 8, and after the
measurement, since several methods are based on membrane proteolytic cleavage, it gets truncated to form tBid, which
changes. translocates to mitochondria to oligomerize Bak or Bax
In contrast to the apoptosis-inducing effects described promoting the release of cytochrome c [100, 102]. This
above, dietary curcumin can inhibit chemotherapy-induced connects the death receptor and mitochondrial apoptotic
apoptosis through inhibition of ROS generation and pathways to amplify the signals downstream of caspase
blockade of JNK function [95]. Curcumin also inhibited activation. Apart from Bid-mediated connection between the
camptothecin-, mechlorethamine-, and doxorubicin-induced two apoptotic pathways, additional cross talk can also occur
apoptosis of MCF-7, MDA-MB-231, and BT-474 human such as the caspase 6-mediated activation of procaspase 8
breast cancer cells by up to 70%. Inhibition of programmed [55], but no information on this is available for curcumin-
cell death was time and concentration dependent, but induced apoptosis.
occurred after relatively brief 3-h exposures, or at curcumin Tumor cells proliferate by overexpressing oncogenes such
concentrations of 1 µM [95]. Curcumin inhibited apoptosis as c-myc, and curcumin down regulates the expression of
in dexamethasone-treated rat thymocytes accompanied by survival genes c-myc, egr-1, and bcl-XL in B cell lymphoma
partial suppression of AP-1 activity [96]. The capacity of cells [71]. Curcumin induces apoptosis in lung and prostate
curcumin to inhibit both cell growth and death strongly cancer cell lines with a decrease in expression of Bcl-2, and
implies that these two biological processes share a common Bcl-XL [48, 80]. In smooth muscle cells, significant
pathway at some point and that curcumin affects a common reduction in Bcl-2 m RNA was detected with curcumin [38].
step, presumably involving a a glutathione-independent In HL-60 cells, Bcl2 protein level decreased to 30% after 6 h
mechanism of cell protection [97]. The cytoprotective effect of curcumin treatment and a further 6 h treatment
of curcumin against Shiga toxin-induced injury in cultured subsequently reduced it to 20% [90]. Overexpression of Bcl-
human proximal tubule epithelial cells may be a 2 in HL-60 cells resulted in a delay of curcumin-treated cells
consequence of an increased expression of hsp70 [98]. entering into apoptosis [90]. Similarly, HL-60 cells made to
It is not perfectly clear as to what cellular conditions are overexpress Bcl-2 or Bcl-XL completely blocked curcumin-
contributing to the induction of apoptosis by curcumin as mediated induction of cytochrome c release, caspases 8 and
opposed to the conditions in which curcumin is not able to 3, and cleavage of Bid and PARP [61]. However, Bcl-2 over
induce apoptosis and the conditions that direct it to protect expression fails to protect AK-5 cells against curcumin
cells from apoptosis. Apoptosis-inducing activities of induced apoptosis [103]. Curcumin up regulates Bax but
curcumin derivatives in various cell types have not been Bcl-2 remains unchanged in Ehrlich’s Ascites carcinoma
studied in detail. There is also a need for synthetic cells, bypassing the Bcl-2 checkpoint and overriding its
derivatives of curcumin that induce apoptosis more protective effect on apoptosis [79]. Similarly, the apoptosis
effectively with better bioavailability. suppressor Bcl-2 and promoter Bax were not changed with
curcumin treatment in basal cell carcinoma [70]. In non-
apoptotic cells, Bax is a soluble monomeric protein
IMPLICATIONS FOR CANCER THERAPY diffusely distributed in the cytoplasm [104]. Bax deficiency
renders cancer cells resistant to several anticancer drugs
Basically, cancer therapy aims to kill the cancer cells by acting via the mitochondria or endoplasmic reticulum stress
apoptosis very selectively without affecting the normal cells [105, 106]. Using Bax+/- and Bax-/- human colon cancer
and in this context it is very important to understand the cells, we have found an essential role for Bax in curcumin-
mechanisms responsible for drug-induced apoptosis induced apoptosis (unpublished observations).
especially to choose or improve strategies for treatment.
Tumor progression needs proteins that promote cell Ku 70, a subunit of Ku protein complex involved in
proliferation but additionally requires the expression of DNA repair, plays a major role in keeping the Bax protein in
antiapoptotic proteins and/or the inactivation of proapoptotic inactive conformation even during apoptotic stresses [107].
proteins. Although the molecular mechanisms of High expression of Ku70 observed in cancer cells suggests
antiproliferative and apoptotic effects of curcumin remain additional roles for this protein in cancer progression and
elusive, studies on the expression of specific proteins that chemoresistance [108, 109]. Our laboratory has shown that
regulate apoptosis in various cell types following treatment Bcl-XL and Ku70 protect human colon cancer cells by
with curcumin have provided several clues to formulate inhibiting the release of cytochrome c, Smac and AIF, and
strategies of cancer treatment with this compound. the activation of caspases 9, 8 and 3 triggered by curcumin
[84]. Cells transfected with antisense Bcl-XL or Ku70 were
Different steps in the two apoptotic pathways are highly more sensitive to curcumin through enhanced activation of
regulated and one of the earliest steps is the activation of caspases 9 and 3 and release of cytochrome c, Smac and
caspase 8 that can be inhibited by FLIPs (FADD-like AIF. Curcumin-induced activation of caspase 8 was blocked
interleukin-1-β-converting enzyme like protease inhibitory by Ku70 but not by Bcl-XL. Cells made to overexpress Bax
proteins) [13]. Multi-domain Bcl-2 family of proteins are were highly sensitized to curcumin when Ku70 was down
important regulators of apoptosis that can be grouped into regulated [84].
the antiapoptotic members comprising Bcl-2, Bcl-XL and
Mcl-1, the proapoptotic members such as Bax and Bak and Inhibitors of apoptosis proteins (IAPs) such as XIAP, c-
the BH3 domain only proteins Bim, Bid and Bik [99]. Bcl- IAP1, c-IAP2 and survivin, are another class of regulatory
Implications for Cancer Therapy Current Cancer Drug Targets, 2005, Vol. 5, No. 2 123

proteins that can bind and inhibit caspases, but IAPs can be resistance to chemo- or radio- therapy [112]. Curcumin-
inhibited by the binding of Smac promoting caspase resistant tumor cell lines showed significantly higher
activation [13]. Bush et al. reported the suppression of production of Hsp70, while the sensitive cells produced only
XIAP in human melanoma cells in response to curcumin basal levels of Hsps [46]. Our laboratory results show that
treatment [69]. Transfection of extrinsic Smac gene results in heat shock partly prevents the curcumin-mediated release of
its over-expression in gastric cancer (MKN-45) cells, which AIF but not that of Smac and cytochrome c from
sensitized these cells to curcumin-induced apoptosis [110]. mitochondria and reduces the activation of caspases 9 and 3
but not 8 induced by curcumin in human colon cancer cells
Recent experimental evidence suggests that heat shock
[63]. These cells were protected from curcumin-induced cell
proteins like hsp70, 90 and 27 can also function as
death by Hsp70 while cells harboring antisense Hsp70
antiapoptotic proteins by suppressing the signaling events of
(Ashsp70) were highly sensitive to curcumin. Loss of
apoptosis [111-113]. High expression of Hsps especially
mitochondrial transmembrane potential induced by curcumin
Hsp27 and Hsp70, in breast, endometrial, or gastric cancer
was further accelerated by antisense Hsp70 expression and
has been associated with metastasis, poor prognosis and

Fig. (2). Known regulators of curcumin-induced apoptosis


124 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

Hsp70 restored it partly. Ashsp70 cells released more PC-3 cells in which Akt is constitutively active [121].
cytochrome c, AIF and Smac from mitochondria upon Blocking peroxisome proliferator-activated receptor-gamma
curcumin treatment than control cells. Hsp70 partly (PPAR-γ) activation abrogated the effects of curcumin on
prevented the release of AIF but not the other proteins. induction of apoptosis and inhibition of the expression of
Activation of caspases 3 and 9 induced by curcumin was extracellular matrix (ECM) genes in activated hepatic stellate
also inhibited by Hsp70, whereas more activation could be cells (HSC) in vitro [122]. Furthermore, curcumin
seen in Ashsp70 cells, although caspase 8 activation was suppresses the gene expression of TGF-β receptors and
unaffected by changes in Hsp70 expression. Curcumin- interrupts the TGF-β signaling pathway in activated HSC,
induced cleavage of PARP and DFF45 was inhibited by mediated by PPAR-γ activation [122]. These results
Hsp70 but enhanced in Ashsp70 cells [64]. Our studies demonstrated that curcumin stimulated PPAR-γ activity in
demonstrated the potential of Hsp70 in protecting SW480 activated HSC in vitro, which is required for curcumin to
cells from curcumin-induced apoptosis and highlighted that reduce cell proliferation, induce apoptosis and suppress
silencing the expression of Hsp70 is an effective approach to ECM gene expression.
augment curcumin-based therapy in cancers that are resistant
Curcumin has the ability to inhibit iNOS induction by
due to Hsp70 expression. It is apparent that many molecules
LPS in the mammary gland and to scavenge NO radicals
regulate curcumin-induced apoptosis by interfering with the
[123]. In ex vivo cultured BALB/c mouse peritoneal
mitochondrial pathway as shown in Fig. (2).
macrophages, 1-20 µM of curcumin reduced the production
Transcription factors such as AP-1 and a number of other of iNOS mRNA in a concentration-dependent manner [124].
signal transduction intermediators including adhesion
The length of telomeres at chromosome ends shortens
molecules, several kinases such as receptor tyrosine kinases,
during each cell cycle, but after reaching a critical shortening
mitogen activated protein kinases (MAPKs) and c-jun N-
the sensors of DNA damage start inducing p53-dependent
terminal kinases (JNKs) influence the course of drug-induced
cell cycle arrest or apoptosis [125]. However, in tumor cells,
apoptosis. Short-term treatment of human epidermoid
increased telomerase activity takes care of teleomere
carcinoma (A431) cells with curcumin inhibited EGF
stabilization [126]. Curcumin inhibits telomerase activity in
receptor intrinsic kinase activity up to 90% in a dose- and
human breast cancer cells (MCF-7) by down regulating the
time-dependent manner, and also inhibited EGF-induced
expression of human telomerase reverse transcritpase [67].
tyrosine phosphorylation of EGF receptors. The observed
early effects of curcumin were mediated via a cellular The growth inhibitory effect of curcumin in breast cancer
mechanism(s), and preceded the period when inhibition of cells was time- and dose-dependent, and correlated with its
cell growth occurred [114]. Curcumin effectively inhibited inhibition of ornithine decarboxylase activity [41].
p185neu tyrosine kinase receptor activity by depleting Curcumin is also known to inhibit PKC [127],
p185neu protein and potently suppressed the growth of cyclooxygenase and lipoxygenase enzymes [128].
multiple breast cancer cell lines [45]. Curcumin enhances the cytotoxicity of chemotherapeutic
Curcumin increases growth arrest and DNA damage- agents in prostate cancer cells by inducing p21
inducible gene 153 (GADD153) mRNA (and also protein) (WAF1/CIP1) and C/EBPbeta expression, and suppressing
expression before the appearance of apoptotic features NF-κB activation [129]. Curcumin enhances the sensitivity
without depending on upstream MAPK [115]. It appears that of prostate tumor cells to TRAIL by inhibiting NF-κB
JNK, but not p38 or extracellular signal-regulated protein activation through blockade of phosphorylation of IκBα and
kinase signaling, plays an important role in curcumin its degradation [130, 131]. Curcumin increases the
mediated apoptosis in human colon cancer cells [62]. sensitivity of ovarian cancer cells (CAOV3 and SKOV3) to
cisplatin [132]. Combining curcumin with 5-fluorouracil
Many cancer cells protect themselves against apoptosis
significantly increased growth inhibition of AGS human
by activating nuclear factor-kappaB (NF-κ B/Rel), a
gastric carcinoma cells compared with either curcumin or 5-
transcription factor that promotes cell survival [116].
FU alone suggesting synergistic actions of the two drugs
Curcumin induces apoptosis by down-regulating NF-κB
[133].
expression in ovarian cancer cells [78] or its activity in
melanoma cells [68]. To understand the role of NF-κB in Aggarwal has recently patented his findings showing that
curcumin-induced apoptosis, we stably transfected relA gene a combination of curcumin and paclitaxel (taxol) can be used
encoding the p65/RelA subunit of NF-κB, into L-929 to treat and inhibit metastasis of breast tumors
(mouse fibrosarcoma) cells and the relA-transfected cells (US2004002499). When tumor cells were pretreated with
were resistant to varying doses of curcumin, whereas the common biologic modulators such as tamoxifen,
parental cells underwent apoptosis in a time- and dose- dexamethasone, and curcumin, the doxorubicin-induced NF-
dependent manner [117]. Recently, it has been reported that κB activation was attenuated significantly [134]. Vinblastine
fresh CD138+ cells derived from multiple myeloma patients sensitivity to multidrug-resistant cervical cells KB-V1 was
express constitutively active NF-κB and STAT3, and increased by curcumin [135]. Curcumin-induced down-
suppression of these transcription factors with curcumin regulation of NF-κB also induced chemosensitivity to
inhibits the survival of the cells leading to apoptosis [118]. vincristine and melphalan in human multiple myeloma cells
Akt, a serine/threonine protein kinase and a downstream [28].
target of phosphatidyl inositol 3-kinase, suppresses Synergistic inhibitory effects of 13-cis retinoic acid and
apoptosis by activating NF-κB [119, 120]. One of the curcumin have been reported on the growth of head and neck
mechanisms of curcumin’s action may be via inhibition of squamous cell carcinoma cells [136]. Combinations of
Akt as it completely inhibited Akt activation in LNCaP and curcumin and retinoic acid have a particularly potent
Implications for Cancer Therapy Current Cancer Drug Targets, 2005, Vol. 5, No. 2 125

Fig. (3). Potential strategies targeting curcumin-induced apoptotic pathway to overcome chemoresistance in cancers

inhibitory effect on the proliferation of HL-60 cells [137]. number of cell cycle genes and p53 [140]. Microarray studies
Down-regulation of Bcl-2 in curcumin plus radiation-treated need to be extended to cell lines of different tissues
PC-3 cells, together, altered the Bcl-2/Bax ratio and this especially because curcumin is known to induce cell-type
caused the enhanced radiosensitization effect [138]. Piperine dependent changes.
enhances the serum concentration, extent of absorption and
Inspite of many in vitro studies on curcumin-induced
bioavailability of curcumin in both rats and humans [26] and
apoptosis, studies conducted in animals and humans have
the biological effects of such a potentially useful
not focused on measuring apoptosis-related parameters.
combination need to be understood in the future. Based on
Fifteen patients with advanced colorectal cancer refractory to
our work and the available literature, we have outlined some
standard chemotherapies received curcuma extract daily for
of the potential strategies that can be employed in curcumin-
up to 4 months. Oral curcumin extract was tolerated well
based therapy in Fig. (3).
and dose-limiting toxicity was not observed [141].
Studying the expression changes of thousands of genes at Curcumin was also used without any notable side-effects or
a given time and/or condition may provide increased insight toxicity in patients suffering from idiopathic inflammatory
into the mechanism of action of curcumin in cancer cells, orbital pseudotumors [142] and phase-II clinical trials were
and increasing our understand of how this compound can conducted with curcumin in patients with urinary bladder
protect against carcinogenesis. Microarray analysis revealed cancer, uterine cervical intraepithelial lesions, oral
the down regulation of 81 genes and up regulation of 71 leukoplasia and intestinal metaplasia of the stomach [143].
genes after curcumin treatment in highly invasive lung Among naturally occurring compounds, curcumin is fit to be
adenocarcinoma cells. Below sublethal concentrations (10 a front-runner in the race to the drug market as it is a
µM), several invasion-related genes were suppressed and pharmacologically safe compound (when administered at
several heat-shock proteins (Hsp27, Hsp70 and Hsp40-like doses up to 10 g/day, human clinical trials indicated no
proteins) were induced by curcumin [139]. Curcumin was dose-limiting toxicity) [17,143].
found to up regulate cdk inhibitor genes, and slightly down
regulate cyclin B1 and cdc2 in human umbilical vein
endothelial cells [37]. Gene expression changes were studied CONCLUSIONS AND FUTURE DIRECTIONS
in human colon cancer cell lines, using cDNA microarrays It is generally believed that chemotherapy eliminates pre
with four thousand human genes, and curcumin modulated a neoplastic or neoplastic cells by the induction of apoptosis,
126 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

but the other possibility that some forms of chemotherapy- prospects for future clinical use. A review. Arch. Gerontol.
induced cell death cannot be readily classified as apoptosis Geriatr. 2002, 34, 37-46.
[9] Huang, M. T.; Smart, R. C.; Wong, C. Q.; Conney, A. H.
or necrosis exists [144, 145]. Lysosomes have recently Inhibitory effect of curcumin, chlorogenic acid, caffeic acid, and
emerged as the central players of apoptotic cell death in ferulic acid on tumor promotion in mouse skin by 12-O-
addition to mitochondria and lysosomal proteases from the tetradecanoylphorbol-13-acetate. Cancer Res. 1988, 48, 5941-
cathepsin family frequently translocate from lysosomal 5946.
[10] Huang, M. T.; Lou, Y. R.; Ma, W.; Newmark, H. L.; Reuhl, K. R.;
lumen to the cytosol to promote cell death. Cathepsin B and Conney, A. H. Inhibitory effects of dietary curcumin on
Cathepsin D act as the main executors of caspase- forestomach, duodenal, and colon carcinogenesis in mice. Cancer
independent cell death induced by TNF-α and several Res. 1994, 54, 5841-5847.
chemotherapeutic agents [146, 147]. With the present [11] Kelloff, G. J.; Boone, C. W.; Crowell, J. A.; Steele, V. E.; Lubet,
knowledge it is not very clear whether curcumin being a R.; Sigman, C. C. Chemopreventive drug development:
perspectives and progress. Cancer Epidemiol. Biomarkers Prev.
lipophilic compound known to induce apoptotic like 1994, 3, 85-98.
changes independent of caspases can amplify lysosomal [12] Joe, B.; Vijaykumar, M.; Lokesh, B. R. Biological properties of
mediated cathepsin release. Novel apoptotic related proteins curcumin-cellular and molecular mechanisms of action. Crit. Rev.
in this and other unknown mechanisms regulated by Food Sci. Nutr. 2004, 44, 97-111.
[13] Igney, F. H.; Krammer, P. H. Death and anti-death: tumour
curcumin can be rapidly identified with a proteomic resistance to apoptosis. Nat. Rev. Cancer 2002, 2, 277-288.
approach and such studies will also provide a strong [14] Johnstone, R. W.; Ruefli, A. A.; Lowe, S. W. Apoptosis: a link
rationale for the selection of therapeutic targets for between cancer genetics and chemotherapy. Cell 2002, 108, 153-
personalized treatment regimens. Presumably all the 164.
potential approaches highlighted in this review may not [15] Lowe, S. W.; Lin, A. W. Apoptosis in cancer. Carcinogenesis
2000, 21, 485-495.
reach the clinic but serve only to demonstrate the proof of [16] Nagata, S. Apoptosis by death factor. Cell 1997, 88, 355-365.
concept, but it is high time that the knowledge gained is [17] Aggarwal, B. B.; Kumar, A.; Bharti, A. C. Anticancer potential of
translated into clinical applications. Time-tested curcumin: preclinical and clinical studies. Anticancer Res. 2003,
pharmacological safety, multiple molecular targets favorable 23, 363-398.
[18] Chainani-Wu, N. Safety and anti-inflammatory activity of
to treat a genetic disease like cancer with multiple defects, curcumin: a component of tumeric (Curcuma longa). J. Altern.
scope for improving bioavailability and/or biological Complement. Med. 2003, 9, 161-168.
activity (through synthetic curcuminoids of desired activity [19] Leu, T. H.; Maa, M. C. The molecular mechanisms for the
aided by molecular modeling) vouch in favor of curcumin to antitumorigenic effect of curcumin. Curr. Med. Chem. Anti-
Cancer Agents 2002, 2, 357-370.
be developed as a drug for prevention and therapy of various [20] Li, J. K.; Lin-Shia, S. Y. Mechanisms of cancer chemoprevention
cancers either alone or in combination with standard drugs or by curcumin. Proc. Natl. Sci. Counc. Repub. China B 2001, 25,
novel approaches. 59-66.
[21] Huang, M. T.; Newmark, H. L.; Frenkel, K. Inhibitory effects of
curcumin on tumorigenesis in mice. J. Cell. Biochem. Suppl. 1997,
ACKNOWLEDGEMENTS 27, 26-34.
[22] Wang, Y. J.; Pan, M. H.; Cheng, A. L.; Lin, L. I.; Ho, Y. S.; Hsieh,
Work in the authors’ laboratory was supported by C. Y.; Lin, J. K. Stability of curcumin in buffer solutions and
funding from the Kerala State Council for Science, characterization of its degradation products. J. Pharm. Biomed.
Anal. 1997, 15, 1867-1876.
Technology and the Environment (to D. K.), a grant from [23] Ravindranath, V.; Chandrasekhara, N. Absorption and tissue
the Life Sciences Research Board, Defence Research distribution of curcumin in rats. Toxicology 1980, 16, 259-265.
Development Organization, Government of India (to D. K. [24] Ravindranath, V.; Chandrasekhara, N. Metabolism of curcumin--
and S. K.) and a Senior Research Fellowship (to R. R.) of studies with [3H]curcumin. Toxicology 1981, 22, 337-344.
the Council of Scientific and Industrial Research, [25] Ireson, C.; Orr, S.; Jones, D. J.; Verschoyle, R.; Lim, C. K.; Luo, J.
L.; Howells, L.; Plummer, S.; Jukes, R.; Williams, M.; Steward, W.
Government of India. The authors thank Goodwin Jinesh for P.; Gescher, A. Characterization of metabolites of the
technical help. chemopreventive agent curcumin in human and rat hepatocytes
and in the rat in vivo, and evaluation of their ability to inhibit
phorbol ester-induced prostaglandin E2 production. Cancer Res.
REFERENCES 2001, 61, 1058-1064.
[26] Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.;
[1] Araujo, C. C.; Leon, L. L. Biological activities of Curcuma longa Srinivas, P. S. Influence of piperine on the pharmacokinetics of
L. Mem. Inst. Oswaldo Cruz. 2001, 96, 723-728. curcumin in animals and human volunteers. Planta Med. 1998, 64,
[2] Ammon, H. P.; Wahl, M. A. Pharmacology of Curcuma longa. 353-356.
Planta. Med. 1991, 57, 1-7. [27] Wu, Y.; Chen, Y.; He, M. The influence of curcumin on the cell
[3] Chattopadhyay, I.; Biswas, K.; Bandyopadhyay, U.; Banerjee, R. cycle of HL-60 cells and contrast study. J. Tongji Med. Univ.
K. Turmeric and curcumin: Biological actions and medicinal 2000, 20, 123-125.
applications. Curr. Sci. 2004, 87, 44-53. [28] Bharti, A. C.; Donato, N.; Singh, S.; Aggarwal, B. B. Curcumin
[4] Charles, V.; Charles, S. X. The use and efficacy of Azadirachta (diferuloylmethane) down-regulates the constitutive activation of
indica ADR ('Neem') and Curcuma longa ('Turmeric') in scabies. nuclear factor-kappa B and IkappaBalpha kinase in human
A pilot study. Trop. Geogr. Med. 1992, 44, 178-181. multiple myeloma cells, leading to suppression of proliferation
[5] Srimal, R. C.; Dhawan, B. N. Pharmacology of diferuloyl methane and induction of apoptosis. Blood 2003, 101, 1053-1062.
(curcumin), a non-steroidal anti-inflammatory agent. J. Pharm. [29] Wu, Y.; Chen, Y.; Xu, J.; Lu, L. Anticancer activities of curcumin
Pharmacol. 1973, 25, 447-452. on human Burkitt's lymphoma. Zhonghua Zhong Liu Za Zhi 2002,
[6] Ruby, A. J.; Kuttan, G.; Babu, K. D.; Rajasekharan, K. N.; Kuttan, 24, 348-352.
R. Anti-tumour and antioxidant activity of natural curcuminoids. [30] Aggarwal, S.; Takada, Y.; Singh, S.; Myers, J. N.; Aggarwal, B.
Cancer Lett. 1995, 94, 79-83. B. Inhibition of growth and survival of human head and neck
[7] Rao, T. S.; Basu, N.; Siddiqui, H. H. Anti-inflammatory activity of squamous cell carcinoma cells by curcumin via modulation of
curcumin analogues. Indian J. Med. Res. 1982, 75, 574-578. nuclear factor-kappaB signaling. Int. J. Cancer 2004, 111, 679-
[8] Miquel, J.; Bernd, A.; Sempere, J. M.; Diaz-Alperi, J.; Ramirez, 692.
A. The curcuma antioxidants: pharmacological effects and
Implications for Cancer Therapy Current Cancer Drug Targets, 2005, Vol. 5, No. 2 127

[31] Jaiswal, A. S.; Marlow, B. P.; Gupta, N.; Narayan, S. Beta- [51] Ashkenazi, A.; Dixit, V. M. Death receptors: signaling and
catenin-mediated transactivation and cell-cell adhesion pathways modulation. Science 1998, 281, 1305-1308.
are important in curcumin (diferuylmethane)-induced growth [52] Liu, X.; Kim, C. N.; Yang, J.; Jemmerson, R.; Wang, X. Induction
arrest and apoptosis in colon cancer cells. Oncogene 2002, 21, of apoptotic program in cell-free extracts: requirement for dATP
8414-8427. and cytochrome c. Cell 1996, 86, 147-157.
[32] Moragoda, L.; Jaszewski, R.; Majumdar, A. P. Curcumin induced [53] Li, P.; Nijhawan, D.; Budihardjo, I.; Srinivasula, S. M.; Ahmad,
modulation of cell cycle and apoptosis in gastric and colon cancer M.; Alnemri, E. S.; Wang, X. Cytochrome c and dATP-dependent
cells. Anticancer Res. 2001, 21, 873-878. formation of Apaf-1/caspase-9 complex initiates an apoptotic
[33] Hanif, R.; Qiao, L.; Shiff, S. J.; Rigas, B. Curcumin, a natural plant protease cascade. Cell 1997, 91, 479-489.
phenolic food additive, inhibits cell proliferation and induces cell [54] Zou, H.; Li, Y.; Liu, X.; Wang, X. An APAF-1 cytochrome c
cycle changes in colon adenocarcinoma cell lines by a multimeric complex is a functional apoptosome that activates
prostaglandin-independent pathway. J. Lab. Clin. Med. 1997, 130, procaspase-9. J. Biol. Chem. 1999, 274, 11549-11556.
576-584. [55] Slee, E. A.; Harte, M. T.; Kluck, R. M.; Wolf, B. B.; Casiano, C.
[34] Chen, Y. C.; Kuo, T. C.; Lin-Shiau, S. Y.; Lin, J. K. Induction of A.; Newmeyer, D. D.; Wang, H. G.; Reed, J. C.; Nicholson, D.
HSP70 gene expression by modulation of Ca(+2) ion and cellular W.; Alnemri, E. S.; Green, D. R.; Martin, S. J. Ordering the
p53 protein by curcumin in colorectal carcinoma cells. Mol. cytochrome c-initiated caspase cascade: hierarchical activation
Carcinogenesis 1996, 17, 224-234. of caspases-2, -3, -6, -7, -8, and -10 in a caspase-9-dependent
[35] Chen, H.; Zhang, Z. S.; Zhang, Y. L.; Zhou, D. Y. Curcumin manner. J. Cell Biol. 1999, 144, 281-292.
inhibits cell proliferation by interfering with the cell cycle and [56] Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial
inducing apoptosis in colon carcinoma cells. Anticancer Res. protein that promotes cytochrome c-dependent caspase activation
1999, 19, 3675-3680. by eliminating IAP inhibition. Cell 2000, 102, 33-42.
[36] Singh, A. K.; Sidhu, G. S.; Deepa, T.; Maheshwari, R. K. [57] Bielak-Zmijewska, A.; Koronkiewicz, M.; Skierski, J.; Piwocka,
Curcumin inhibits the proliferation and cell cycle progression of K.; Radziszewska, E.; Sikora, E. Effect of curcumin on the
human umbilical vein endothelial cell. Cancer Lett. 1996, 107, apoptosis of rodent and human nonproliferating and proliferating
109-115. lymphoid cells. Nutr. Cancer 2000, 38, 131-138.
[37] Park, M. J.; Kim, E. H.; Park, I. C.; Lee, H. C.; Woo, S. H.; Lee, J. [58] Bielak-Mijewska, A.; Piwocka, K.; Magalska, A.; Sikora, E. P-
Y.; Hong, Y. J.; Rhee, C. H.; Choi, S. H.; Shim, B. S.; Lee, S. H.; glycoprotein expression does not change the apoptotic pathway
Hong, S. I. Curcumin inhibits cell cycle progression of induced by curcumin in HL-60 cells. Cancer Chemother.
immortalized human umbilical vein endothelial (ECV304) cells by Pharmacol. 2004, 53, 179-185.
up-regulating cyclin-dependent kinase inhibitor, p21WAF1/CIP1, [59] Roy, M.; Chakraborty, S.; Siddiqi, M.; Bhattacharya, R. K.
p27KIP1 and p53. Int. J. Oncol. 2002, 21, 379-383. Induction of Apoptosis in Tumor Cells by Natural Phenolic
[38] Chen, H. W.; Huang, H. C. Effect of curcumin on cell cycle Compounds. Asian Pac. J. Cancer Prev. 2002, 3, 61-67.
progression and apoptosis in vascular smooth muscle cells. Br. J. [60] Chen, Y.; Wu, Y.; He, J.; Chen, W. The experimental and clinical
Pharmacol. 1998, 124, 1029-1040. study on the effect of curcumin on cell cycle proteins and
[39] Batth, B. K.; Tripathi, R.; Srinivas, U. K. Curcumin-induced regulating proteins of apoptosis in acute myelogenous leukemia.
differentiation of mouse embryonal carcinoma PCC4 cells. J. Huazhong Univ. Sci. Technol. Med. Sci. 2002, 22, 295-298.
Differentiation 2001, 68, 133-140. [61] Anto, R. J.; Mukhopadhyay, A.; Denning, K.; Aggarwal, B. B.
[40] Ramachandran, C.; You, W. Differential sensitivity of human Curcumin (diferuloylmethane) induces apoptosis through
mammary epithelial and breast carcinoma cell lines to curcumin. activation of caspase-8, BID cleavage and cytochrome c release:
Breast Cancer Res. Treat. 1999, 54, 269-278. its suppression by ectopic expression of Bcl-2 and Bcl-xl.
[41] Mehta, K.; Pantazis, P.; McQueen, T.; Aggarwal, B. B. Carcinogenesis 2002, 23, 143-150.
Antiproliferative effect of curcumin (diferuloylmethane) against [62] Collett, G. P.; Campbell, F. C. Curcumin induces c-jun N-terminal
human breast tumor cell lines. Anticancer Drugs 1997, 8, 470- kinase-dependent apoptosis in HCT116 human colon cancer cells.
481. Carcinogenesis 2004, 25, 2183-2189.
[42] Pan, M. H.; Chang, W. L.; Lin-Shiau, S. Y.; Ho, C. T.; Lin, J. K. [63] Rashmi, R.; Santhosh Kumar, T. R.; Karunagaran, D. Human
Induction of apoptosis by garcinol and curcumin through colon cancer cells differ in their sensitivity to curcumin-induced
cytochrome c release and activation of caspases in human apoptosis and heat shock protects them by inhibiting the release of
leukemia HL-60 cells. J. Agric. Food Chem. 2001, 49, 1464-1474. apoptosis-inducing factor and caspases. FEBS Lett. 2003, 538, 19-
[43] Jiang, M. C.; Yang-Yen, H. F.; Yen, J. J.; Lin, J. K. Curcumin 24.
induces apoptosis in immortalized NIH 3T3 and malignant cancer [64] Rashmi, R.; Kumar, S.; Karunagaran, D. Ectopic expression of
cell lines. Nutr. Cancer 1996, 26, 111-120. Hsp70 confers resistance and silencing its expression sensitizes
[44] Gautam, S. C.; Xu, Y. X.; Pindolia, K. R.; Janakiraman, N.; human colon cancer cells to curcumin-induced apoptosis.
Chapman, R. A. Nonselective inhibition of proliferation of Carcinogenesis 2004, 25, 179-187.
transformed and nontransformed cells by the anticancer agent [65] Choudhuri, T.; Pal, S.; Agwarwal, M. L.; Das, T.; Sa, G. Curcumin
curcumin (diferuloylmethane). Biochem. Pharmacol. 1998, 55, induces apoptosis in human breast cancer cells through p53-
1333-1337. dependent Bax induction. FEBS Lett 2002, 512, 334-340.
[45] Hong, R. L.; Spohn, W. H.; Hung, M. C. Curcumin inhibits [66] Kim, M. S.; Kang, H. J.; Moon, A. Inhibition of invasion and
tyrosine kinase activity of p185neu and also depletes p185neu. induction of apoptosis by curcumin in H-ras-transformed
Clin. Cancer Res. 1999, 5, 1884-1891. MCF10A human breast epithelial cells. Arch. Pharm. Res. 2001,
[46] Khar, A.; Ali, A. M.; Pardhasaradhi, B. V.; Varalakshmi, C. H.; 24, 349-354.
Anjum, R.; Kumari, A. L. Induction of stress response renders [67] Ramachandran, C.; Fonseca, H. B.; Jhabvala, P.; Escalon, E. A.;
human tumor cell lines resistant to curcumin-mediated apoptosis: Melnick, S. J. Curcumin inhibits telomerase activity through
role of reactive oxygen intermediates. Cell Stress Chaperones human telomerase reverse transcritpase in MCF-7 breast cancer
2001, 6, 368-376. cell line. Cancer Lett 2002, 184, 1-6.
[47] Syng-Ai, C.; Kumari, A. L.; Khar, A. Effect of curcumin on [68] Zheng, M.; Ekmekcioglu, S.; Walch, E. T.; Tang, C. H.; Grimm,
normal and tumor cells: Role of glutathione and bcl-2. Mol. E. A. Inhibition of nuclear factor-kappaB and nitric oxide by
Cancer Ther. 2004, 3, 1101-1108. curcumin induces G2/M cell cycle arrest and apoptosis in human
[48] Radhakrishna Pillai, G.; Srivastava, A. S.; Hassanein, T. I.; melanoma cells. Melanoma Res 2004, 14, 165-171.
Chauhan, D. P.; Carrier, E. Induction of apoptosis in human lung [69] Bush, J. A.; Cheung, K. J., Jr.; Li, G. Curcumin induces apoptosis
cancer cells by curcumin. Cancer Lett. 2004, 208, 163-170. in human melanoma cells through a Fas receptor/caspase-8
[49] Tourkina, E.; Gooz, P.; Oates, J. C.; Ludwicka-Bradley, A.; pathway independent of p53. Exp. Cell Res. 2001, 271, 305-314.
Silver, R. M.; Hoffman, S. Curcumin-induced apoptosis in [70] Jee, S. H.; Shen, S. C.; Tseng, C. R.; Chiu, H. C.; Kuo, M. L.
scleroderma lung fibroblasts: role of protein kinase cepsilon. Am. Curcumin induces a p53-dependent apoptosis in human basal cell
J. Respir. Cell. Mol. Biol. 2004, 31, 28-35. carcinoma cells. J. Invest. Dermatol. 1998, 111, 656-661.
[50] Holy, J. M. Curcumin disrupts mitotic spindle structure and [71] Han, S. S.; Chung, S. T.; Robertson, D. A.; Ranjan, D.; Bondada,
induces micronucleation in MCF-7 breast cancer cells. Mutat. S. Curcumin causes the growth arrest and apoptosis of B cell
Res. 2002, 518, 71-84.
128 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

lymphoma by downregulation of egr-1, c-myc, bcl-XL, NF-kappa [90] Kuo, M. L.; Huang, T. S.; Lin, J. K. Curcumin, an antioxidant and
B, and p53. Clin. Immunol. 1999, 93, 152-161. anti-tumor promoter, induces apoptosis in human leukemia cells.
[72] Liontas, A.; Yeger, H. Curcumin and resveratrol induce apoptosis Biochim. Biophys. Acta 1996, 1317, 95-100.
and nuclear translocation and activation of p53 in human [91] Jaruga, E.; Salvioli, S.; Dobrucki, J.; Chrul, S.; Bandorowicz-
neuroblastoma. Anticancer Res. 2004, 24, 987-998. Pikula, J.; Sikora, E.; Franceschi, C.; Cossarizza, A.; Bartosz, G.
[73] Duvoix, A.; Morceau, F.; Delhalle, S.; Schmitz, M.; Apoptosis-like, reversible changes in plasma membrane
Schnekenburger, M.; Galteau, M. M.; Dicato, M.; Diederich, M. asymmetry and permeability, and transient modifications in
Induction of apoptosis by curcumin: mediation by glutathione S- mitochondrial membrane potential induced by curcumin in rat
transferase P1-1 inhibition. Biochem. Pharmacol. 2003, 66, 1475- thymocytes. FEBS Lett. 1998, 433, 287-293.
1483. [92] Piwocka, K.; Zablocki, K.; Wieckowski, M. R.; Skierski, J.; Feiga,
[74] Khar, A.; Ali, A. M.; Pardhasaradhi, B. V.; Begum, Z.; Anjum, R. I.; Szopa, J.; Drela, N.; Wojtczak, L.; Sikora, E. A novel
Antitumor activity of curcumin is mediated through the induction apoptosis-like pathway, independent of mitochondria and
of apoptosis in AK-5 tumor cells. FEBS Lett. 1999, 445, 165-168. caspases, induced by curcumin in human lymphoblastoid T
[75] Bhaumik, S.; Anjum, R.; Rangaraj, N.; Pardhasaradhi, B. V.; (Jurkat) cells. Exp. Cell Res. 1999, 249, 299-307.
Khar, A. Curcumin mediated apoptosis in AK-5 tumor cells [93] Piwocka, K.; Jaruga, E.; Skierski, J.; Gradzka, I.; Sikora, E.
involves the production of reactive oxygen intermediates. FEBS Effect of glutathione depletion on caspase-3 independent
Lett. 1999, 456, 311-314. apoptosis pathway induced by curcumin in Jurkat cells. Free
[76] Cipriani, B.; Borsellino, G.; Knowles, H.; Tramonti, D.; Cavaliere, Radic. Biol. Med. 2001, 31, 670-678.
F.; Bernardi, G.; Battistini, L.; Brosnan, C. F. Curcumin inhibits [94] Jaruga, E.; Sokal, A.; Chrul, S.; Bartosz, G. Apoptosis-
activation of Vgamma9Vdelta2 T cells by phosphoantigens and independent alterations in membrane dynamics induced by
induces apoptosis involving apoptosis-inducing factor and large curcumin. Exp. Cell Res. 1998, 245, 303-312.
scale DNA fragmentation. J. Immunol. 2001, 167, 3454-3462. [95] Somasundaram, S.; Edmund, N. A.; Moore, D. T.; Small, G. W.;
[77] Woo, J. H.; Kim, Y. H.; Choi, Y. J.; Kim, D. G.; Lee, K. S.; Bae, J. Shi, Y. Y.; Orlowski, R. Z. Dietary curcumin inhibits
H.; Min do, S.; Chang, J. S.; Jeong, Y. J.; Lee, Y. H.; Park, J. W.; chemotherapy-induced apoptosis in models of human breast
Kwon, T. K. Molecular mechanisms of curcumin-induced cancer. Cancer Res 2002, 62, 3868-3875.
cytotoxicity: induction of apoptosis through generation of reactive [96] Sikora, E.; Bielak-Zmijewska, A.; Piwocka, K.; Skierski, J.;
oxygen species, down-regulation of Bcl-XL and IAP, the release Radziszewska, E. Inhibition of proliferation and apoptosis of
of cytochrome c and inhibition of Akt. Carcinogenesis 2003, 24, human and rat T lymphocytes by curcumin, a curry pigment.
1199-1208. Biochem. Pharmacol. 1997, 54, 899-907.
[78] Zheng, L. D.; Tong, Q. S.; Wu, C. H. [Inhibitory effects of [97] Jaruga, E.; Bielak-Zmijewska, A.; Sikora, E.; Skierski, J.;
curcumin on apoptosis of human ovary cancer cell line A2780 Radziszewska, E.; Piwocka, K.; Bartosz, G. Glutathione-
and its molecular mechanism]. Ai. Zheng 2002, 21, 1296-1300. independent mechanism of apoptosis inhibition by curcumin in rat
[79] Pal, S.; Choudhuri, T.; Chattopadhyay, S.; Bhattacharya, A.; Datta, thymocytes. Biochem. Pharmacol. 1998, 56, 961-965.
G. K.; Das, T.; Sa, G. Mechanisms of curcumin-induced apoptosis [98] Sood, A.; Mathew, R.; Trachtman, H. Cytoprotective effect of
of Ehrlich's ascites carcinoma cells. Biochem. Biophys. Res. curcumin in human proximal tubule epithelial cells exposed to
Commun. 2001, 288, 658-665. shiga toxin. Biochem. Biophys. Res. Commun. 2001, 283, 36-41.
[80] Mukhopadhyay, A.; Bueso-Ramos, C.; Chatterjee, D.; Pantazis, P.; [99] Martinou, J. C.; Green, D. R. Breaking the mitochondrial barrier.
Aggarwal, B. B. Curcumin downregulates cell survival Nat. Rev. Mol. Cell Biol. 2001, 2, 63-67.
mechanisms in human prostate cancer cell lines. Oncogene 2001, [100] Korsmeyer, S. J.; Wei, M. C.; Saito, M.; Weiler, S.; Oh, K. J.;
20, 7597-7609. Schlesinger, P. H. Pro-apoptotic cascade activates BID, which
[81] Morin, D.; Barthelemy, S.; Zini, R.; Labidalle, S.; Tillement, J. P. oligomerizes BAK or BAX into pores that result in the release of
Curcumin induces the mitochondrial permeability transition pore cytochrome c. Cell Death Differ. 2000, 7, 1166-1173.
mediated by membrane protein thiol oxidation. FEBS Lett. 2001, [101] Chao, D. T.; Korsmeyer, S. J. BCL-2 family: regulators of cell
495, 131-136. death. Annu. Rev. Immunol. 1998, 16, 395-419.
[82] Susin, S. A.; Lorenzo, H. K.; Zamzami, N.; Marzo, I.; Snow, B. [102] Li, H.; Zhu, H.; Xu, C. J.; Yuan, J. Cleavage of BID by caspase 8
E.; Brothers, G. M.; Mangion, J.; Jacotot, E.; Costantini, P.; mediates the mitochondrial damage in the Fas pathway of
Loeffler, M.; Larochette, N.; Goodlett, D. R.; Aebersold, R.; apoptosis. Cell 1998, 94, 491-501.
Siderovski, D. P.; Penninger, J. M.; Kroemer, G. Molecular [103] Anjum, R.; Khar, A. Differential regulation of apoptosis in AK-5
characterization of mitochondrial apoptosis-inducing factor. tumor cells by the proto-oncogene Bcl-2: presence of Bcl-2
Nature 1999, 397, 441-446. dependent and independent pathways. FEBS Lett. 2001, 499, 166-
[83] Li, L. Y.; Luo, X.; Wang, X. Endonuclease G is an apoptotic 170.
DNase when released from mitochondria. Nature 2001, 412, 95- [104] Hsu, Y. T.; Youle, R. J. Bax in murine thymus is a soluble
99. monomeric protein that displays differential detergent-induced
[84] Rashmi, R.; Kumar, S.; Karunagaran, D. Ectopic expression of conformations. J. Biol. Chem. 1998, 273, 10777-83.
Bcl-xL or Ku70 protects human colon cancer cells (SW480) [105] Theodorakis, P.; Lomonosova, E.; Chinnadurai, G. Critical
against curcumin-induced apoptosis while their downregulation requirement of BAX for manifestation of apoptosis induced by
potentiates it. Carcinogenesis 2004, 25, 1867-1877. multiple stimuli in human epithelial cancer cells. Cancer Res.
[85] Fujisawa, S.; Atsumi, T.; Ishihara, M.; Kadoma, Y. Cytotoxicity, 2002, 62, 3373-3376.
ROS-generation activity and radical-scavenging activity of [106] Yamaguchi, H.; Bhalla, K.; Wang, H. G. Bax plays a pivotal role
curcumin and related compounds. Anticancer Res 2004, 24, 563- in thapsigargin-induced apoptosis of human colon cancer
569. HCT116 cells by controlling Smac/Diablo and Omi/HtrA2 release
[86] Subramanian, M.; Sreejayan; Rao, M. N.; Devasagayam, T. P.; from mitochondria. Cancer Res. 2003, 63, 1483-1489.
Singh, B. B. Diminution of singlet oxygen-induced DNA damage [107] Sawada, M.; Sun, W.; Hayes, P.; Leskov, K.; Boothman, D. A.;
by curcumin and related antioxidants. Mutat. Res. 1994, 311, 249- Matsuyama, S. Ku70 suppresses the apoptotic translocation of
255. Bax to mitochondria. Nat. Cell Biol. 2003, 5, 320-329.
[87] Reddy, A. C.; Lokesh, B. R. Effect of dietary turmeric (Curcuma [108] Wilson, C. R.; Davidson, S. E.; Margison, G. P.; Jackson, S. P.;
longa) on iron-induced lipid peroxidation in the rat liver. Food Hendry, J. H.; West, C. M. Expression of Ku70 correlates with
Chem. Toxicol. 1994, 32, 279-283. survival in carcinoma of the cervix. Br. J. Cancer 2000, 83, 1702-
[88] White, E. L.; Ross, L. J.; Schmid, S. M.; Kelloff, G. J.; Steele, V. 1706.
E.; Hill, D. L. Screening of potential cancer preventing chemicals [109] Zhao, H. J.; Hosoi, Y.; Miyachi, H.; Ishii, K.; Yoshida, M.;
for induction of glutathione in rat liver cells. Oncol. Rep. 1998, 5, Nemoto, K.; Takai, Y.; Yamada, S.; Suzuki, N.; Ono, T. DNA-
507-512. dependent protein kinase activity correlates with Ku70 expression
[89] Ahsan, H.; Parveen, N.; Khan, N. U.; Hadi, S. M. Pro-oxidant, and radiation sensitivity in esophageal cancer cell lines. Clin.
anti-oxidant and cleavage activities on DNA of curcumin and its Cancer Res. 2000, 6, 1073-1078.
derivatives demethoxycurcumin and bisdemethoxycurcumin. [110] Zheng, L. D.; Tong, Q. S.; Tao, K. X.; Wang, L.; Zhang, B.
Chem. Biol. Interact. 1999, 121, 161-175. [Effects of Smac gene overexpression on chemotherapeutic
Implications for Cancer Therapy Current Cancer Drug Targets, 2005, Vol. 5, No. 2 129

sensitivity of gastric cancer cell line MKN-45]. Ai Zheng 2004, [131] Deeb, D.; Jiang, H.; Gao, X.; Hafner, M. S.; Wong, H.; Divine,
23, 361-366. G.; Chapman, R. A.; Dulchavsky, S. A.; Gautam, S. C. Curcumin
[111] Jaattela, M. Escaping cell death: survival proteins in cancer. Exp. sensitizes prostate cancer cells to tumor necrosis factor-related
Cell Res. 1999, 248, 30-43. apoptosis-inducing ligand/Apo2L by inhibiting nuclear factor-
[112] Garrido, C.; Gurbuxani, S.; Ravagnan, L.; Kroemer, G. Heat kappaB through suppression of IkappaBalpha phosphorylation.
shock proteins: endogenous modulators of apoptotic cell death. Mol. Cancer Ther. 2004, 3, 803-812.
Biochem. Biophys. Res. Commun. 2001, 286, 433-442. [132] Chan, M. M.; Fong, D.; Soprano, K. J.; Holmes, W. F.; Heverling,
[113] Saleh, A.; Srinivasula, S. M.; Balkir, L.; Robbins, P. D.; Alnemri, H. Inhibition of growth and sensitization to cisplatin-mediated
E. S. Negative regulation of the Apaf-1 apoptosome by Hsp70. killing of ovarian cancer cells by polyphenolic chemopreventive
Nat. Cell Biol. 2000, 2, 476-483. agents. J. Cell Physiol. 2003, 194, 63-70.
[114] Korutla, L.; Kumar, R. Inhibitory effect of curcumin on [133] Koo, J. Y.; Kim, H. J.; Jung, K. O.; Park, K. Y. Curcumin inhibits
epidermal growth factor receptor kinase activity in A431 cells. the growth of AGS human gastric carcinoma cells in vitro and
Biochim. Biophys. Acta 1994, 1224, 597-600. shows synergism with 5-fluorouracil. J. Med. Food 2004, 7, 117-
[115] Scott, D. W.; Loo, G. Curcumin-induced GADD153 gene 121.
upregulation in human colon cancer cells. Carcinogenesis 2004, [134] Chuang, S. E.; Yeh, P. Y.; Lu, Y. S.; Lai, G. M.; Liao, C. M.; Gao,
25, 2155-2164. M.; Cheng, A. L. Basal levels and patterns of anticancer drug-
[116] Karin, M.; Cao, Y.; Greten, F. R.; Li, Z. W. NF-kappaB in cancer: induced activation of nuclear factor-kappaB (NF-kappaB), and
from innocent bystander to major culprit. Nature Rev. Cancer its attenuation by tamoxifen, dexamethasone, and curcumin in
2002, 2, 301-310. carcinoma cells. Biochem. Pharmacol. 2002, 63, 1709-1716.
[117] Anto, R. J.; Maliekal, T. T.; Karunagaran, D. L-929 cells [135] Limtrakul, P.; Anuchapreeda, S.; Buddhasukh, D. Modulation of
harboring ectopically expressed RelA resist curcumin-induced human multidrug-resistance MDR-1 gene by natural
apoptosis. J. Biol. Chem. 2000, 275, 15601-15604. curcuminoids. BMC Cancer 2004, 4, 13.
[118] Bharti, A. C.; Shishodia, S.; Reuben, J. M.; Weber, D.; Alexanian, [136. Spingarn, A.; Sacks, P. G.; Kelley, D.; Dannenberg, A. J.;
R.; Raj-Vadhan, S.; Estrov, Z.; Talpaz, M.; Aggarwal, B. B. Schantz, S. P. Synergistic effects of 13-cis retinoic acid and
Nuclear factor-kappaB and STAT3 are constitutively active in arachidonic acid cascade inhibitors on growth of head and neck
CD138+ cells derived from multiple myeloma patients, and squamous cell carcinoma in vitro. Otolaryngol Head Neck Surg.
suppression of these transcription factors leads to apoptosis. Blood 1998, 118, 159-164.
2004, 103, 3175-3184. [137] Liu, Y.; Chang, R. L.; Cui, X. X.; Newmark, H. L.; Conney, A. H.
[119] Romashkova, J. A.; Makarov, S. S. NF-kappaB is a target of AKT Synergistic effects of curcumin on all-trans retinoic acid- and 1
in anti-apoptotic PDGF signalling. Nature 1999, 401, 86-90. alpha,25-dihydroxyvitamin D3-induced differentiation in human
[120] Ozes, O. N.; Mayo, L. D.; Gustin, J. A.; Pfeffer, S. R.; Pfeffer, L. promyelocytic leukemia HL-60 cells. Oncol. Res. 1997, 9, 19-29.
M.; Donner, D. B. NF-kappaB activation by tumour necrosis [138] Chendil, D.; Ranga, R. S.; Meigooni, D.; Sathishkumar, S.; Ahmed,
factor requires the Akt serine-threonine kinase. Nature 1999, 401, M. M. Curcumin confers radiosensitizing effect in prostate
82-85. cancer cell line PC-3. Oncogene 2004, 23, 1599-1607.
[121] Chaudhary, L. R.; Hruska, K. A. Inhibition of cell survival signal [139] Chen, H. W.; Yu, S. L.; Chen, J. J.; Li, H. N.; Lin, Y. C.; Yao, P.
protein kinase B/Akt by curcumin in human prostate cancer cells. L.; Chou, H. Y.; Chien, C. T.; Chen, W. J.; Lee, Y. T.; Yang, P. C.
J. Cell. Biochem. 2003, 89, 1-5. Anti-invasive gene expression profile of curcumin in lung
[122] Zheng, S.; Chen, A. Activation of PPARgamma is required for adenocarcinoma based on a high throughput microarray analysis.
curcumin to induce apoptosisand to inhibit the expression of Mol. Pharmacol. 2004, 65, 99-110.
extracellular matrix genes in hepatic stellate cell in vitro. [140] Van Erk, M. J.; Teuling, E.; Staal, Y. C.; Huybers, S.; Van
Biochem. J. 2004, 384, 149-157. Bladeren, P. J.; Aarts, J. M.; Van Ommen, B. Time- and dose-
[123] Onoda, M.; Inano, H. Effect of curcumin on the production of dependent effects of curcumin on gene expression in human
nitric oxide by cultured rat mammary gland. Nitric Oxide 2000, 4, colon cancer cells. J. Carcinog. 2004, 3, 8.
505-515. [141] Sharma, R. A.; McLelland, H. R.; Hill, K. A.; Ireson, C. R.;
[124] Chan, M. M.; Huang, H. I.; Fenton, M. R.; Fong, D. In vivo Euden, S. A.; Manson, M. M.; Pirmohamed, M.; Marnett, L. J.;
inhibition of nitric oxide synthase gene expression by curcumin, a Gescher, A. J.; Steward, W. P. Pharmacodynamic and
cancer preventive natural product with anti-inflammatory pharmacokinetic study of oral Curcuma extract in patients with
properties. Biochem. Pharmacol. 1998, 55, 1955-1962. colorectal cancer. Clin. Cancer Res. 2001, 7, 1894-1900.
[125] Hahn, W. C.; Stewart, S. A.; Brooks, M. W.; York, S. G.; Eaton, [142] Lal, B.; Kapoor, A. K.; Agrawal, P. K.; Asthana, O. P.; Srimal, R.
E.; Kurachi, A.; Beijersbergen, R. L.; Knoll, J. H.; Meyerson, M.; C. Role of curcumin in idiopathic inflammatory orbital
Weinberg, R. A. Inhibition of telomerase limits the growth of pseudotumours. Phytother. Res. 2000, 14, 443-447.
human cancer cells. Nat. Med. 1999, 5, 1164-1170. [143] Cheng, A. L.; Hsu, C. H.; Lin, J. K.; Hsu, M. M.; Ho, Y. F.; Shen,
[126] Zhang, X.; Mar, V.; Zhou, W.; Harrington, L.; Robinson, M. O. T. S.; Ko, J. Y.; Lin, J. T.; Lin, B. R.; Ming-Shiang, W.; Yu, H. S.;
Telomere shortening and apoptosis in telomerase-inhibited human Jee, S. H.; Chen, G. S.; Chen, T. M.; Chen, C. A.; Lai, M. K.; Pu,
tumor cells. Genes Dev. 1999, 13, 2388-2399. Y. S.; Pan, M. H.; Wang, Y. J.; Tsai, C. C.; Hsieh, C. Y. Phase I
[127] Hasmeda, M.; Polya, G. M. Inhibition of cyclic AMP-dependent clinical trial of curcumin, a chemopreventive agent, in patients
protein kinase by curcumin. Phytochemistry 1996, 42, 599-605. with high-risk or pre-malignant lesions. Anticancer Res 2001, 21,
[128] Huang, M. T.; Lysz, T.; Ferraro, T.; Abidi, T. F.; Laskin, J. D.; 2895-2900.
Conney, A. H. Inhibitory effects of curcumin on in vitro [144] Blagosklonny, M. V. Cell death beyond apoptosis. Leukemia 2000,
lipoxygenase and cyclooxygenase activities in mouse epidermis. 14, 1502-1508.
Cancer Res. 1991, 51, 813-819. [145] Leist, M.; Jaattela, M. Four deaths and a funeral: from caspases to
[129] Hour, T. C.; Chen, J.; Huang, C. Y.; Guan, J. Y.; Lu, S. H.; Pu, Y. alternative mechanisms. Nat. Rev. Mol. Cell Biol. 2001, 2, 589-
S. Curcumin enhances cytotoxicity of chemotherapeutic agents in 598.
prostate cancer cells by inducing p21(WAF1/CIP1) and [146] Broker, L. E.; Huisman, C.; Span, S. W.; Rodriguez, J. A.; Kruyt,
C/EBPbeta expressions and suppressing NF-kappaB activation. F. A.; Giaccone, G. Cathepsin B mediates caspase-independent
Prostate 2002, 51, 211-218. cell death induced by microtubule stabilizing agents in non-small
[130] Deeb, D.; Xu, Y. X.; Jiang, H.; Gao, X.; Janakiraman, N.; cell lung cancer cells. Cancer Res. 2004, 64, 27-30.
Chapman, R. A.; Gautam, S. C. Curcumin (diferuloyl-methane) [147] Foghsgaard, L.; Wissing, D.; Mauch, D.; Lademann, U.;
enhances tumor necrosis factor-related apoptosis-inducing Bastholm, L.; Boes, M.; Elling, F.; Leist, M.; Jaattela, M. Cathepsin
ligand-induced apoptosis in LNCaP prostate cancer cells. Mol. B acts as a dominant execution protease in tumor cell apoptosis
Cancer Ther. 2003, 2, 95-103. induced by tumor necrosis factor. J. Cell Biol. 2001, 153, 999-
1010.
130 Current Cancer Drug Targets, 2005, Vol. 5, No. 2 Karunagaran et al.

You might also like