Sei sulla pagina 1di 14

T

he class of steroid-like compounds designated cardiac glycosides includes well-known drugs such as
digoxin, digitoxin, and ouabain. Their continued efficacy in treatment of congestive heart failure and
as anti-arrhythmic agents is well appreciated. Less well known, however, is the emerging role of this
category of compounds in the prevention and/or treatment of proliferative diseases such as cancer. New
findings within the past five years have revealed these compounds to be involved in complex cell-signal
transduction mechanisms, resulting in selective control of human tumor but not normal cellular prolif-
eration. As such, they represent a promising form of targeted cancer chemotherapy. New clinical studies
of their anticancer potential as single or adjuvant treatments may provide insight into these potentially
valuable therapeutic options. This review focuses on recent findings on cellular pharmacology of cardiac
glycosides as they relate to treatment of human cancer and attempts to explain why these agents have been
overlooked in the past.

Robert A. Newman1, Peiying Yang1, Alison D. Pawlus1,


and Keith I. Block2
1
Department of Experimental Therapeutics and Pharmaceutical Development Center,
University of Texas, M. D. Anderson Cancer Center, Houston, TX 77054;
2
Institute for Integrative Cancer Care, Evanston, IL 60201

36
Cardiac Glycosides as Novel Cancer Therapeutic Agents

History of the Use of Cardiac Glycosides toad, known to contain multiple bufodeninolides (another type
for Cancer of cardiac glycoside), including bufalin (Figure 1) (5–7). The
potential use of cardenolide-like compounds for the treatment
The use of cardiac glycoside containing plants for medicinal of cancer, initially investigated forty years ago, was abandoned
purposes was first reported in ancient texts more than 1500 because of the toxicity of these compounds (8, 9). It was only
years ago. They have been used traditionally as arrow poisons, recently, however, that Scandinavian oncologists suggested that the
abortifacients, emetics, diuretics, and heart tonics. It is the latter apoptosis produced by digitalis in human tumor cells occurred at
pharmacologic activity that cardiac glycosides are most com- concentrations that could be achieved without toxicity in humans
monly associated with, and after 200 years, compounds such and, therefore, this agent might be useful for treatment of cancer
as digitalis and digoxin are still prescribed by Western doctors (10–12). In 1979, Stenkvist et al. (13) noted that the altered mor-
for control of congestive heart failure. Their use began after a phology of breast cancer cells from women treated with digitalis
meticulous analysis of a local herbalist’s formula in 1775 by the (who had undergone mastectomy). Women receiving digitalis had
English physician and scientist William Withering. He found that tumor cells with more benign characteristics than those tumor
a patient with “dropsy” (congestive heart failure) improved after cells in patients not receiving this cardiac glycoside. Moreover,
administration of an extract containing foxglove (Digitalis purpurea the cancer recurrence rate of women taking digitalis was lower,
L.) (1). Compounds extracted
from foxglove and oleander R=lactone ring
18
include cardenolides (Figure 17 O
12 H
Lactone Moiety
1), such as digitalis, digoxin, 19
11
H
13
16
O
9 14 15
O O
and oleandrin, which increase 2
1
10 8 Cardenolides R=
cardiac contractility and act 3 5 H 7
OH
4 6 H
as antiarrythmic agents to O O
H H
control atrial fibrillation (2, HO O
O H OH
3). The mechanism of their Steroid
Bufadienolides R=
HO Bufalin
action for the treatment of OH HO
H
congestive heart failure arises O O
from the inhibition of Na+,K+- Glycone
O O OH
ATPase, with a resulting HO
H H
increase in intracellular cal- HO H
cium concentrations. Cardiac H
H O
glycosides, however, have a H OH
narrow therapeutic index, H OH O O O
O OH
limiting their wider applica- O
O O HO
tion to the treatment of other HO H
Ouabain H
diseases, such as cancer. HO H
OCH3 Oleandrin OH
Despite their potential
to cause serious side effects, H OH

application of plant extracts O O


O O H
containing cardiac glycosides O O
HO
for the treatment of malignant OH Digitoxin
OH O
OH O
disease may extend back to
HO H
Arab physicians in the eighth O O
H
century (4). It is not just O
HO H
plants, however, that con- H
tribute to our appreciation of HN S OH H H OH
O
cardiac glycosides possibly O O
H OH O O H
having a role in cancer man-
O O
agement. An ancient Chinese O HO
OH OH Digioxin
OH
medicinal treatment of cancer OH
still in use today involves UNBS-1450 O
application of an extract of Figure 1. Structures of cardiac glycosides with antiproliferative activity. Representative cardenolide and bufadieno-
secretions of the Bufo bufo lide cardiac glycoside compounds are presented.

February 2008
Volume 8, Issue 1 37
Review

Table 1. List of Plants and Animals with Cardiac Glycosides Having Antiproliferative Activities
Plant/Animal Species Cardiac Glycoside(s) In Vitro Cytotoxic Effect Reference
Apocynum cannabinum L. Apocannoside, cymarin Human nasopharynx carcinoma (KB) (66)
(Apocynaceae)
Asclepias curassavica L. Calotropin, 16α-acetoxycalotropin, Human lung carcinoma (A549), breast carcinomas (67)
(Asclepiadaceae) 15β-hydroxycalotropin, calactin, (MCF-7 and MDA-MB-231), and hepatoma (HepG2)
15β-hydroxycalactin, asclepin,
16α-hydroxyasclepin, uscharidin,
uscharin, uzarigenin
Beaumontia brevituba Digitoxigenin, oleandrigenin, digi- Human breast carcinoma (BC1), colon carcinoma (68)
Oliver (Apocynaceae) toxigenin, α-l-cymaroside, digitoxigenin (Col2), fibrosarcoma (HT-1080), nasopharyngeal
β-gentiobiosyl-α-l-cymaroside, Δ16- carcinoma (KB), vinblastine-resistant KB (KB-V1), lung
digitoxigenin β-d-glucosyl-α-l-cymaroside carcinoma (Lu1), and melanoma (Mel2)
Bufo bufo gargarizans L. Bufalin, cinobufagin Prostate carcinomas (LNCaP, DU145, PC3), and (69, 70)
hepatoma (PLC/PRF/5)
Calotropis procera (Ait.) Calotropin, calactin, uscharin, voruscha- Human non-small-cell lung carcinoma (A549), human (71, 72)
R. Br. (Asclepiadaceae) rin, 2’’-oxovoruscharin glioblastomas (Hs683 and U373), human colon
carcinomas (HCT-15 and LoVo), hepatoma (Huh7),
non-hepatoma (COS-1), and colorectal carcinoma
(COLO 320)
Cerbera odollam Gaertner 2′-O-Acetyl cerleaside A, 17α-neriifolin, Human oral epidermoid carcinoma (KB), breast (73)
(Apocynaceae) 17β-neriifolin, cerberin carcinoma (BC), and small-cell lung carcinoma
(NCI-H187)
Coronilla varia L. Hyrcanoside Human lymphocytic leukemia (P-388) and (74)
(Fabaceae) nasopharynx carcinomas (9KB)
Crossopetalum gau- Securigenin-3β-O-β-6-deoxyguloside, Human oral epidermoid carcinoma (KB) (75)
meri (Loes.) Lundell 19-hydroxy-sarmentogenin-3β-O-β-6-
(Celastraceae) deoxyguloside, sarmentogenin-3β-O-
(α-allosyl-(1→4)-β-6-deoxyalloside),
securigenin-3β-O-(α-allosyl-(1→4)-β-6-
deoxyalloside)
Digitalis purpurea L. Digoxin, digitoxin, gitoxin Human prostate carcinomas (LNCaP, DU145, PC3), (76, 77)
(Scrophulariaceae) renal adenocarcinoma (TK-10), breast adenocarci-
Digitalis lanata noma (MCF-7), malignant melanoma (UACC-62),
(Scrophulariaceae) and chronic myelogenous leukemia (K-562)
Elaeodendron sp. Elaeodendrosides Human ovarian carcinoma (A2780) (78)
Euonymus alata (Thunb.) Acovenosigenin A 3-O-α-l- Human oral epidermoid (KB), promyelocytic (79)
Sieb. (Celastraceae) ramnopyranoside, euonymoside A, euo- lymphoma (HL-60), non-small-cell lung carcinoma
nymusoside A (A549), and cervical carcinoma (Hela)
Euonymus sieboldianus Euonymoside A Human lung carcinoma (A549) and ovarian (80)
Blume (Celastraceae) adenocarcinoma (SK-OV- 3)
Maquira calophylla (P.&E.) Maquiroside A Human oral epidermoid carcinoma (KB) (81)
C.C. Berg (Moraceae)
Nerium oleander L. Oleander, oleandrin, cardenolide N-1, Human Jurkat leukaemia (T-cell), histiocytic lymphoma (82, 83)
(Apocynaceae) cardenolide N-4, 3β-O-(β-d-sarmentosyl)- (U-937), promyelocytic lymphoma (HL-60), cervical
16β-acetoxy-14-hydroxy-5β,14β-card- carcinoma (Hela), breast carcinoma (MCF-7), pros-
20-(22)-enolide, 16β-acetoxy-3β,14- tate carcinomas (LNCap, DU145, PC3), malignant
dihydroxy-5β,14β-card-20-(22)-enolide fibroblast (VA-13), and liver carcinoma (HepG2)

38
Cardiac Glycosides as Novel Cancer Therapeutic Agents

Table 1. continued
Nierembergia aristata D. 17-epi-11α-hydroxy-6,7- Human breast carcinoma (BC1), fibrosarcoma (HT), (84)
Don (Solanaceae) dehydrostrophanthidin-3- lung cancer (LU1), melanoma (Mel2), colon carci-
O-β-boivinopyranoside; noma (Col2), oral epidermoid (KB), drug resistant KB
6,7-dehydrostrophanthidin- with and without vinblastine, epidermoid carcinoma
3-O-β-boivinopyranoside; (A-431), prostate carcinoma (LNCaP), hormone-
6,7-dehydrostrophanthidin-3-O-β- dependent breast carcinoma (ZR-75-1), and glioma
oleandropyranoside (U373)
Ornithogalum umbellatum Convallatoxin Human oral epidermoid carcinoma (KB) (85)
L. (Hyacinthaceae)
Pergularia tomentosa L. 3′-O-β-d-glucopyranosylcalactin, 12-dehy- Kaposi’s sarcoma (KS) (86)
(Asclepiadaceae) droxyghalakinoside, 6′-dehydroxygha-
lakinoside, ghalakinoside, calactin
Periploca graeca L. Periplocin isomers Human prostate carcinoma (PC-3) (87)
(Asclepiadaceae)
Rhodea japonica (Thunb.) Rhodexin A Human leukemia (K562) (88)
Roth. (Liliaceae)
Saussurea stella Maxim. 3-O-β-d-fucopyranosylstrophanthidin, Human gastric cancer (BGC-823) and hepatoma (89)
(Asteraceae) 3-O-β-d-quinovopyranosylperiplogenin, (Bel-7402)
3-O-β-d-glucopyranosyl-(1→4)-α-l-
rhamnopyranosylcannogenin, 3-O-β-d-
xylopyranosylperiplogenin, 3-O-β-d-
quinovopyranosylstrophanthidin,
3-O-β-d-xylopyranosylstrophanthidin,
3-O-β-d-fucopyranosylperiplogenin,
3-O-α-l-rhamnopyranosylcannogenol,
convallatoxin, 3-O-α-l-
rhamnpyranosylacovenosigenin A
Streblus asper Lour. Stebloside, mansonin Oral human epidermoid carcinoma (KB) (90)
(Moraceae)
Streptocaulon juven- Periplogenin digitoxoside, Human fibrosarcoma (HT-1080) (91)
tas (Lour.) Merr. Periplocymarin, digitoxigenin
(Asclepiadaceae) 3-O-(O-β-d-glucopyranosyl-(1→6)-
O-β-d-glucopyranosyl-(1→4)-β-d-
digitoxopyranoside, echujin, corchoru-
soside C
Streptocaulon griffithii 3-O-(β-glucopyranosyl)acovenosigenin A Human gastrointestinal cancer (HCG-27), lung carci- (92)
Hook.f. (Asclepiadaceae) noma (A549), breast carcinoma (MCF-7), and cervi-
cal carcinoma (HeLa)
Strophanthus gratus Ouabain Human prostate carcinomas (LNCaP, DU145, PC3) (76)
Thevetia ahouia (L.) A. Neriifolin, 3′-O-methylevomonoside, National Cancer Institute’s human disease oriented (93)
DC. (Apocynaceae) 2′-acetylneriifolin 60-cell line tumor screening panel
Thevetia peruviana Thevetin A and B, thevetoside Human hepatoma (SMMC-7721), gastric carcinoma (93)
(Pers.) K. Schum. (SGC-7901), and cervical carcinoma (HeLa)
(Apocynaceae)

Urginea maritime (L.) Proscillaridin A, scillaren A Human breast carcinoma (MCF-7) (94–96)
Baker (Liliaceae)

February 2008
Volume 8, Issue 1 39
Review

suggesting an important beneficial anticancer effect of this cardiac 1). The purpose of the present review, therefore, is to examine
glycoside (14). the hypothesis already expressed by some (10, 14–20), that use of
Within the past ten years, there has been a substantial selected cardiac glycosides may represent a worthwhile approach
increase in the number of studies observing the effects of cardiac toward control of malignant cell proliferation even despite their
glycosides on the growth of human malignant tumor cells. A narrow therapeutic index. This is all the more timely because
review of the literature indicates a surprising variety of plants and promising clinical trials of cardiac glycosides and extracts contain-
even animals whose extracts and isolated cardiac glycoside com- ing them have recently been initiated.
pounds have been cited for their antiproliferative effects (Table

Table 2. Reported Mechanisms of Cardiac Glycoside–Mediated Inhibition of Tumor Cell Proliferation


Compound Proposed antiproliferative mechanism(s) Reference
Oleandrin Alteration of membrane fluidity (15, 35, 97, 100)
Decreased activation of nuclear transcription factors NF-κB, (19, 98)
JNK, and AP-1
Increased intracellular calcium (17, 50)
Increased expression of FasL (99)
Increased ROS production, oxidative injury, and mitochondrial injury (50, 51, 58)
Decreased phosphorylation of Akt (17, 36, 57)
Inhibition of cellular transport of tumor growth factors (FGF-2) (100)
Down regulation of IL-8 receptors (99)
Initiates Apo2L/TRAIL apoptosis via increased expression of death receptors 4 and 5 (18, 48)
Activation of calcineurin and nuclear transcription factor NF-AT (99)
Bufalin Increased activation of MAPKs (101–103)
Decreased cAMP content (5)
Inhibition of topoisomerases I and II (17, 104, 105)
Induction of differentiation in human myeloid leukemia (106, 107)
Downregulation of cyclin A, Bcl-2 and Bcl-xL; Increased expression of p21 and Bax (7, 104, 108)
Ouabain, Digitoxin Loss of mitochondrial membrane potential; increase Par-4 expression (17, 109)
Increased Ca2+ uptake (15, 16, 110, 111)
Acts as an estrogen receptor antagonist (15, 112)
Sustained ROS production (29, 31, 111)
Regulates expression of cell tight junctions and adhesion molecules (16, 17, 55)
Selective protein kinase C activation leading to differentiation (17, 113)
Increased activation of MAPKs (16, 113, 114)
Reduction in anti-apoptotic proteins Bcl-xL and Bcl-2 (114–116)
Increased cytochrome c release and caspase activation (28, 114, 116)
Inhibition of topoisomerase I (28, 61, 117, 118)
Block activation of the TNF-α/NF-κB signaling pathway (59, 119)
UNBS1450 Decreased heat shock protein (Hsp70) (59, 120)
Increased permeabilization of lyososomal membrane (120)
Block activation of the TNF-α/NF-κB signaling pathway (59)

NF-κB, Nuclear Factor-kappaB; JNK, c-Jun NH2-terminal kinase; AP-1, Activator Protein-1; FasL, Fas ligand; ROS, reactive oxygen species;
FGF-2, Fibroblast Growth Factor 2; IL-8, Interleukin-8; TNF-α, Tumor Necrosis Factor–α; TRAIL, (TNF)-related apoptosis-inducing ligand; NF-AT,
Nuclear Factor of Activated T cells; MAPKs, mitogen-activated protein kinases.

40
Cardiac Glycosides as Novel Cancer Therapeutic Agents

Na+,K+-ATPase: Beyond Cell Membrane cinogen 1,2-dimethyldrazine (24). There have also been reports
Exchange of Na+ and K+ of increased expression of particular subunits of Na+,K+-ATPase
in gastric (25) and bladder cancers (26). In addition, alterations
Na+,K+-ATPase, as an energy-transducing ion pump, has been in overall Na+,K+-ATPase activity and relative subunit abundance
studied extensively since its discovery in 1957 (21). This enzyme were observed in a highly invasive form of human renal carci-
consists of two types of subunits, designated α and β, in addition noma cells (27), non-small cell lung cancer (28), and carcinoma
to a single-transmembrane-spanning protein, FXYD––named for cell lines obtained from a number of other tissues (29). It would
the conserved amino acids in its signature motif: (Phe-Xxx-Tyr- appear, however, that simply looking at enzyme subunit content
Asp). The α subunit, responsible for binding of Mg2+, ATP, Na+, K+, or relative activity in malignant and non-malignant tissue may not
and cardiac glycosides, is considered the catalytic subunit of the provide adequate insight into the role of this enzyme in cancer.
enzyme. The β subunit is a glycoprotein that seems to act as an This can now be interpreted in the light of newly proposed conse-
adhesion molecule that regulates gap junction proteins; is involved quences of cardiac glycoside binding to Na+,K+-ATPase.
in structural and functional maturation of the holoenzyme; facili-
tates transport of the α subunit to the plasma membrane and main- Proposed Mechanism(s) of
tenance of the enzyme in the lateral membrane of epithelial cells Cardiac Glycoside–Mediated
(15–17). The function of the FXYD protein involves regulation of Antiproliferative Effects
the enzyme function, thus adapting the kinetic properties of active
Na+ and K+ transport to the specific needs of different cells (22, 23). An explanation of the role of Na+,K+-ATPase in complex cell
Four α subunit variants, as well as three β, and seven FXYD sub- signaling pathways, many of which are of critical importance
unit variants have been identified (17). The well-established func- to malignant cell proliferation, has been put forth by Xie and
tion of Na+,K+-ATPase is to use ATP as an energy source to drive colleagues (30–32). They have shown that binding of cardiac
excess Na+ out of cells in exchange for K+, thereby maintaining an glycosides (e.g., ouabain) to Na+,K+-ATPase triggers a complex
essential ionic and osmotic
Apo2/TRAIL
balance. Binding of certain α Upregulation of death receptors
(J)
subunits by cardiac glycosides (A) Altered membrane
DR4 fluidty
DR5 Oleandrin,
inhibits ATP binding and dis- bufalin,
TNF-A
C
av
rupts the ability of the enzyme eo
lin
digitoxin
TNFR

to perform this exchange in an FGF-2 (I)


FADD EGFR Export inhibited
efficient manner. This, in turn, Caspase (H)
B
results in an enhanced entry Activation B
TRADD Lysosomal membrane
Caveolin permeabilization
of calcium into cells, which, Apoptosis PLC A A RIP/TRAF2
IP
Apoptosis
(N)
in the event of failing cardiac ER/SC PI3K
3 Src
(F)
Ca 2+
(G) JNK
myofibrils, helps produce a Ras
ASK-1
Raf Cytochrome c release
more efficient myocardial con-
2+ NF-+B (M) LC3
traction and improves cardiac Ca
Akt (E) (K)
(B) i MEK
ROS
pump activity. PKC
Akt P (D)
Mitochondrial
Erk 1/2 Autophagy
What, then, is the evi- Apoptosis pErk 1/2
(L) Mitochondria
injury and
membrane condensation
dence supporting the hypoth- Nuclear
membrane
potential
AP-1 Gene activation
esis that Na+,K+-ATPase may differentiation
Bcl-XL, BcI-2
(C) topoisomerase I and IIa

be an important target for


cancer therapy? For the past Figure 2. The Na+,K+-ATPase signalosome complex. The binding of selected cardiac glycosides (CGs)––such as ole-
andrin, bufalin, and digitoxin––to Na+,K+-ATPase results in complex but well-documented changes in cell signaling events.
ten years, published stud-
The “signalosome” complex includes the enzyme, Na+,K+-ATPase as well as Src, phosphoinositide-3 kinase (PI3K), and
ies have suggested a role for phospholipase C each of which, in turn, sets into action complex signaling events that can result in tumor cell death
Na+,K+-ATPase in regulation through either apoptosis or autophagy-related mechanisms. Administration of CGs can (A) increase the (cell surface)
expression of death receptors (DR4, DR5) and activate caspase activity; (B) result in increased intracellular calcium con-
of cell growth and expres-
centrations, which, in turn, (C) decreases the expression of transcription factors such as Activator Protein-1 (AP-1). CG
sion of various genes beyond treatment can also (D) inhibit activation (i.e., block the phosphorylation) of Akt, which normally blocks apoptosis; (E) inhibit
that of ion transport. Davies activation of the transcription factor Nuclear Factor-kappaB (NF-κB); (F) activate the Ras pathway, leading to increase
activity of Raf–MAPK pathway; (G) activate Src; (H) inhibit tumor necrosis factor (TNF)-mediated activation of NF-κB by
et al., for example, observed
inhibiting the binding of tumor necrosis factor receptor 1–associated death domain protein (TRADD) to the cellular mem-
altered Na+,K+-ATPase activ- brane; (I) inhibit extracellular transport of tumor growth factors, such as fibroblast growth factor-2 (FGF-2); (J) alter mem-
ity in premalignant mucosa brane fluidity which, in turn, may inhibit Fas-related signaling; (K) lead to the production of reactive oxygen species (ROS)
with subsequent injury to mitochondria; (L) produce a decrease in mitochondria membrane potential and a decrease in
months before tumor devel-
quantity of anti-apoptotic proteins Bcl-XL and Bcl-2 and topoisomerases I and II; and (M) cause mitochondrial condensa-
opment induced by the car- tion and loss of function, that, in turn, can lead to autophagic processes and cell death (N). Adapted from (30).

February 2008
Volume 8, Issue 1 41
Review

A 120
tion has been compiled (Table 2 and Figure 2), and there are
Panc-02
BxPC3 several excellent reviews on this subject (15–17). There are uni-
100
MiaPaca fying themes that link mechanisms involving the water-soluble
PANC-1
Percent of control cell growth

(ouabain) and relatively lipid-soluble (oleandrin, bufalin, and


80
digitoxin) cardiac glycosides, including activation of ERK1/2;
increased expression of the cell cycle inhibitor p21Cip1 and con-
60 15.6 nM 210 nM
IC50 sequent inhibition of cell cycle progression (through decreased
expression of cyclin proteins); inhibition of transcription factors,
40
5.6 nM such as Nuclear Factor-kappaB (NF-κB) and Activator Protein-1
(AP-1); inhibition of Akt (a protein serine–threonine kinase) and
20 related critical components of the phosphoinositide-3 kinase
(PI3K) pathway; initiation of death receptor–mediated apoptosis;
0 sustained ROS production with consequent mitochondrial injury;
0 100 200 300 400 500 600
Concentration (nM) and inhibition of topoisomerases and reduction in expression of
anti-apoptotic proteins, such as Bcl-xL and Bcl-2. Although the
known primary target of cardiac glycosides is Na+,K+-ATPase, not
MiaPaca

B
Panc-02

PANC-1
BxPC3

all of the reported mechanisms of antiproliferative action (Table 2)


are related to inhibition of this important enzyme. That is, cardiac
A1 glycosides such as bufalin and oleandrin should be thought of as
pleiotropic or multi-mechanistic anticancer agents. Few, if any, of
A3 these published reports on mechanism of action of cardiac glyco-
sides, however, address reasons for the well-established differential
B-Actin in cardiac glycoside-mediated effects on human- vs rodent-derived
Figure 3. Relative human and rodent tumor cell sensitivity to olean- malignant cell lines.
drin correlates with Na+,K+-ATPase subunit composition. A. Mouse
pancreatic cancer cells that lack expression of the α-3 subunit (Panc-02) are
non-responsive to oleandrin whereas human tumor cell lines MiaPaca and
Species-Dependent Sensitivity and Selective
Panc-1 (that contain high expression of α-3 relative to α-1) are extremely Human Tumor Cell Response to Cardiac
responsive. The human pancreatic tumor cell line BXPC3 that contains a Glycosides: Important Mechanistic Clues
very low level of the α-3 subunit, as per immunoblot analysis (B), is some-
what resistant to cytotoxic effects of oleandrin. These data suggest that it is
the lack of α3 in rodent tumor cell lines that explains their resistance to car- An unusual attribute of compounds such as oleandrin, bufalin,
diac glycosides. In addition, the data suggest that it is the relative α3:α1 ratio and digitoxin is that they are, on the one hand, almost completely
correlates with human tumor cell sensitivity to lipid soluble cardiac glycosides nontoxic to rodent (mouse, rat, and hamster)-derived tumor cell
such as oleandrin.
lines but potently inhibit proliferation of monkey and human
signaling cascade that is initiated by interacting with neighboring tumor cell lines at nanomolar concentrations (33– 36). These
membrane proteins and organized cytosolic cascades of signaling results have been confirmed across a wide spectrum of human
molecules. These signaling complexes send messages to intracellu- and rodent tumor cell lines, including those of hematologic and
lar organelles via the activation of the protein tyrosine kinase Src, solid tumor derivation. This species-dependent disparity in tumor
transactivation of epidermal growth factor receptor (EGFR) by Src, cell sensitivity to a proposed antitumor agent is unusual. It may, in
activation of Ras and the p42/44 mitogen-activated protein kinases fact, have contributed to the conclusion in the 1970s that cardiac
[MAPKs, also termed extracellular-regulated protein kinases 1 and glycosides were without efficacy because murine P388 and L1210
2 (ERK1/2)], and increased generation of reactive oxygen species lymphocytic leukemia cell lines were, at the time, the only cell
(ROS) by mitochondria. Activation of these cellular pathways is lines available for anticancer drug development. The magnitude
also linked with translocation of Na+,K+-ATPase, through endo- of difference in response of murine as compared to human tumor
cytosis, to the nucleus (17). In fact, Xie and colleagues (17, 30) cell lines suggests that a fundamental difference in drug targeting
have referred to this as the Na+,K+-ATPase-Src-caveolin “signalo- exists and, thus, serves as a probe or model for re-examination of
some” complex (Figure 2). The view of Na+,K+-ATPase as a simple the pharmacologic role of this class of compounds in treatment of
ion-exchange pump situated solely at the cell membrane is thus human cancers.
outmoded. Further research on this enzyme, including its role in Whereas the differences between human and murine
regulation of cell proliferation and its inhibition through cardiac responses to cardiac glycosides are interesting and should be stud-
glycosides, is clearly warranted. ied further, the differential effects of cardiac glycosieds on human
The diverse mechanisms reported to specifically be involved tumor vs normal cells are essential to their usefulness as a therapy.
in cardiac glycoside-mediated control of malignant cell prolifera- Several published studies have confirmed the observation that car-

42
Cardiac Glycosides as Novel Cancer Therapeutic Agents

diac glycosides have a selective effect on malignant but not normal some prognostic value if that patient is to be subsequently treated
cell proliferation. For example, oleandrin suppresses the activation with a cardiac glycoside.
of certain transcription factors and potentiates ceramide-induced Mijatovic et al., on the other hand, suggest that, rather than
apoptosis in human tumor cells but not in normal, primary the α3 subunit, it is the α1 subunit of Na+,K+-ATPase that could
human cells (19). In vitro observations that leukemia cells under- represent a novel anticancer target (47). They have shown that
go apoptosis in the presence of oleandrin and bufalin, but that human lung cancer cell lines overexpressing the α1 subunit were
normal leukocytes do not, are also consistent with the hypothesis sensitive to a few select cardenolides. They noted that the cardiac
of a potentially therapeutic, selective therapeutic effect of cardiac glycosides produced a marked change in the actin cytoskeleton,
glycosides on tumor growth (37–39). Not only do cardiac gly- suggesting this abets tumor cell death. Whether it is altered
cosides appear to be more effective at inhibiting proliferation of expression of α1, as suggested by Mijatovic et al., or an elevation
malignant cells than normal cells, but they also are more effective of α3, as indicated by our own work, or perhaps even a specific
at sensitizing tumor cells to irradiation, which would appear to ratio of α3:α1 that is most important as a predictor of cell sensi-
increase their potential utility in the clinic. Research reported by tivity, remains to be determined. More research, using human tis-
several investigators (40–42) indicates that cardiac glycosides sen- sues and not just cell lines, will no doubt shed light on the poten-
sitize human tumor but not normal cells to subsequent radiation tial importance and perhaps even prognostic value of the enzyme
treatment. These data suggest that it may be possible to exploit subunit composition within individual types of tumors.
differences in the Na+,K+-ATPase pumps of normal as opposed to It is significant that the relative composition of Na+,K+-ATPase
tumor cells to improve the therapeutic index of radiation therapy. subunits may not be static within human tissues. The relative ratio
Modern drug development seeks specific biochemical differ- of α subunits within the enzyme may shift when tissues are trans-
ences between malignant and normal cells that may be critical to formed from a benign to a malignant state. Sakai et al. (46) recent-
survival of cancer cells. One then attempts to develop selective ly showed, for example, that a decrease in the α1 isoform and an
inhibitors to disrupt these pathways. Na+,K+-ATPase, however, is increase in the α3 subunit occurs in colon tissue when a normal
a ubiquitous enzyme present in every mammalian cell. At first phenotype changes to a malignant one. If, as our data suggest, it
appearance, therefore, it would appear to make Na+,K+-ATPase is the relative expression of α3 that is important for determining
an anticancer target of dubious value unless, of course, the target sensitivity of a tissue to inhibition by cardiac glycosides then, in
were found to be fundamentally different in normal versus malig- essence, the report by Sakai et al. suggests that the tumor becomes
nant human cells or between rodent and mammalian cancer cells. a more sensitive target than normal tissue to cardiac glycoside
Our recent data suggest that, in fact, there is a difference in the therapy (46). Given the current as well as proposed clinical trials
basic subunit composition of Na+,K+-ATPase that might explain of cardiac glycosides for treatment of cancer, specific determina-
the differential species-dependent sensitivity to cardiac glycosides. tion of enzyme subunit composition in specific tissue types as
Although human tumor cells and tissues commonly express both well as pathologic characterization may prove to be a timely tool
α1 and α3 subunits, all rodent tumor cell lines we have examined to help optimize the effectiveness of this class of potential cancer
to date only express the α1 subunit (Figure 3). Early reviews of therapeutic agents.
the biochemical properties of Na+,K+-ATPase suggested that cardi-
ac glycoside binding may be equal to all four α subunit isoforms; Cardiac Glycoside-Mediated Cancer Cell
however, more recent studies have shown a clear preferential Death: Autophagy and Apoptosis
binding of cardenolides to the α3 form over that of the α1 or α2
isoforms (43–45). For example, O’Brien et al. (45) cite a 1000-fold Although it is clear that lipid-soluble cardiac glycosides (i.e.,
difference in binding of ouabain to the α3 isoform over that of digitoxin, oleandrin, and bufalin) have a potent ability to pro-
α1. Given the fact that rodent tumor cells possess the α1 subunit, duce human tumor cell death, the mechanisms by which this is
lack expression of α3, and are unresponsive to inhibition of prolif- accomplished are still being defined. Apoptotic cell death medi-
eration with cardiac glycosides, we suggest that the α3 subunit is ated by cardenolides has been demonstrated in a number of cell
critical. The increased expression of α3 over α1 subunits has also lines. Sreenivasan et al., for example, have shown that oleandrin
been noted in human colon colorectal cancer and colon adeno- produced an increase in expression of Fas and Tumor Necrosis
carcinoma cell lines (e.g., KM12-L4, T-84, HT-29, and WiDr), Factor Receptor 1 (TNFR1), resulting in potentiation of apoptosis
whereas no significant expression of the α3 isoform protein was in tumor cells but not in normal primary cells, such as peripheral
noted in the normal kidney and renal tissues (46). Moreover, blood mononuclear cells or neutrophils (48). Fas–Fas ligand and
human tumor cell lines with a low ratio of α3:α1 are relatively TNF–TNFR1 death pathways are important mediators of apopto-
resistant to growth inhibition with cardiac glycosides but those sis (49). Another recent report has shown that oleandrin, bufalin,
tumor cell lines with high α3:α1 ratios are very sensitive (Figures digoxin, and digitoxin initiate apoptosis induced by Apo2L/TNF-
3 and 4). This finding, of course, also suggests that determination related apoptosis-inducing ligand (TRAIL) in non-small-cell lung
of the relative α3:α1 ratio in tumor biopsy specimens may have cancer cells by increasing the expression of death receptors 4 and

February 2008
Volume 8, Issue 1 43
Review

5 (18). Because Apo2L/TRAIL induces apoptosis in tumor cells Cardiac Glycosides and Cancer Prevention
with little if any toxicity to normal cells, this cytokine is of great
interest to cancer researchers. The selective cardenolide activation Recent investigations of potent cardiac glycosides have focused on
of death receptors may very well contribute to the observation their potential application to the treatment of established cancers;
that compounds such as oleandrin are relatively selective in their however, at least one report has suggested that there may also be
cytotoxic activity. a chemopreventive role for this class of agents. That is, Afaq et al.
Oleandrin elicits caspase-associated apoptosis in human have suggested that oleandrin might serve as an effective agent
prostate carcinoma cells (50). Interestingly, however, treatment of for the prevention or treatment of skin cancer (57). Their research
human PANC-1 pancreatic cancer cells produces clear hallmarks investigated the topical application of oleandrin to CD-1 mice to
of autophagy, including formation of autophagosome bodies with counteract the effects of TPA (12-0-tetradecanoylphorbol-13-ace-
damaged mitochondria and expression of light chain-1 protein, an tate), a widely used skin tumor promoter. The topical application
early indicator of autophagosome formation (51). Frese et al. have of TPA to mouse skin or its treatment in certain epidermal cells is
also suggested that the apoptotic potential of cardiac glycosides known to result in several biochemical alterations, changes in cel-
depends on the cell type treated (18). Our data on the differential lular functions, and histological changes leading to dermal tumor
effects of oleandrin on tumor cells, such as pancreatic vs prostate promotion. The data of Afaq et al. clearly show that application of
tumor cells, as compared to oleandrin-treated normal human cells oleandrin to skin prior to TPA administration affords significant
concurs with this. inhibition of TPA-induced skin edema, hyperplasia, epidermal
ornithine decarboxylase (ODC) activity, and protein expression of
Cardiac Glycosides and Estrogen ODC and cyclooxygenase-2 (COX-2), classical markers of inflam-
Receptor Interaction mation and tumor promotion. Their data also show that topical
application of oleandrin prior to TPA inhibits activation of PI3K
Selected cardiac glycosides may be of particular importance in the and phosphorylation of Akt, activation of NF-κB, and degradation
treatment of human breast cancer. Chen et al. (15) have recently and phosphorylation of the inhibitor of NF-κB α protein (IκBα).
suggested several reasons why cardenolides should be developed These authors, therefore, recommend the use of chemopreven-
as anti-breast cancer drugs. These include the facts that: 1) Na+,K+ tive agents (i.e., oleandrin) in formulations such as emollients or
-ATPase is a key player of cell adhesion and is involved in cancer patches for the prevention or treatment of skin cancer (57). This
progression; 2) the enzyme serves as a versatile signal transducer suggestion is all the more relevant when considered in light of our
involving a number of hormones, including estrogens; and 3) the own work which shows a potent ability of oleandrin to inhibit
aberrant expression and activity of this enzyme in breast cancer human melanoma proliferation (58).
implicates an etiologic or at least contributing role of Na+,K+-
ATPase in the development and progression of this malignant In Vivo Efficacy and Development of
disease. For example, there is now strong evidence that Na+,K+- Cardiac Glycosides for Clinical
ATPase plays an important role in the assembly of tight junctions Cancer Therapy
(TJs) and cell adhesion (52–55). Chen et al. convincingly argue
that altered expression and malfunction of Na+,K+-ATPase may Rodent tumor cells fail to respond to cardiac glycosides in vitro.
lead to abnormal TJ structure and, thus, to altered cell adhesion Similarly, it has been very difficult to demonstrate an in vivo
important in the progression of breast cancer. As mentioned pre- response of syngeneic rodent tumors to administration of this class
viously, there are also strong data supporting the role of Na+,K+- of compounds. However, as shown in Figure 4, there is no ques-
ATPase in a complex signalosome involved in transmitting mem- tion that human tumor cell lines are extremely sensitive to treat-
brane signals to the nucleus. ment with cardiac glycosides such as oleandrin and bufalin. Thus,
A series of reports suggests that estrogen receptor (ER) ligands it is possible that human tumor xenografts would respond. Indeed,
(e.g., 17β-estradiol and estrogen-like molecules) can also serve as this is exactly the case, as shown by several investigators. Han et
ligands of Na+,K+-ATPase. Use of 17β-estradiol enhances Na+,K+- al. (7), for example, explored the response of a human hepatocel-
ATPase activity (56) possibly through improvement of the interac- lular carcinoma cell line (BEL-7402) implanted orthotopically
tion of the enzyme with ATP as well as Na+ and K+ ions. Because (i.e., transplantation of cells or tissue into its normal anatomical
the interaction of 17β-estradiol with ERs serves as an important site) in liver tissue to intraperitoneal treatment with bufalin. They
determinant of breast cancer growth, and cardiac glycosides can found that this toad-derived cardiac glycoside produced significant
block this interaction, cardenolides could be considered effective reductions in tumor volumes and a prolongation in life-span of
modulators of estradiol-dependent breast cancer proliferation. the animals. Importantly, no adverse morphological changes were
noted in myocardial, hepatic, or renal tissues. Another interesting
report involved use of the semi-synthetic cardenolide UNBS-1450
against orthotopically implanted human non-small-cell lung cancer

44
Cardiac Glycosides as Novel Cancer Therapeutic Agents

BxPC3 PANC-1
shown, however, that single cardenolides, or those derived from
various extracts of plants and animals (Table 1), represent potent
ATPase-A3

compounds with selective effects against human tumor cell lines


and xenografts. For example, UNBS-1450, a semisynthetic carde-
Na,K-

nolide derivative of a cardiac glycoside originally isolated from an


African plant (Calotropis procera), has shown promising activity
against human non-small cell lung carcinomas growing as xeno-
grafts in nude mice (59). UNBS-1450 can be safely administered
Oleandrin

to mice in doses that are twenty-four times greater than that of


ouabain and twelves times greater than that of the parent com-
pound oxovoruscharin (64). With good activity against non-small
cell lung cancers in mice with metastases in the brain and liver,
UNBS-1450 is entering phase I clinical trials in Belgium (17).
Figure 4. Relationship of expression of Na+,K+-ATPase α3 subunit and
binding of oleandrin to human pancreatic cancer cell membranes. To date, there is only a single plant extract, Anvirzel™,
Cells were stained using a mitochondrial dye, Mitotracker, (red color) and a derived from Nerium oleander, that has progressed through a
nuclear Herscht dye (blue). Top row: Cells were also exposed to an antibody Phase I trial in the United States. Anvirzel is a hot-water extract
to the α3 subunit of Na+,K+-ATPase (green color). Bottom row: Cells were
incubated with a fluorescent analog of oleandrin (green color). The presence of oleander whose encouraging preclinical activity (65) led to a
of green staining (top row) indicates the relative presence of the α3 subunit successful investigational new drug application (IND) from the
which is more prevalent in the PANC-1 than the BXPC3 cells. There is a cor- FDA and to a Phase I clinical trial performed at the Cleveland
responding uptake of oleandrin (bottom row) only in those cells (PANC-1)
that express the α3 subunit.
Clinic between 2000 and 2001. No objective responses were
noted which might be because the limited time of exposure to
(59). Chronic oral administration of this compound produced a the product and intramuscular route of administration limited
beneficial therapeutic effect against these orthotopically implanted the total volume of extract that could be administered on a daily
A549 tumor cells. Finally, digoxin inhibits human neuroblastoma basis. A longer time of exposure and a different route of admin-
growth in vitro and significantly reduced the growth of human istration may impact response. Also, no dose-limiting toxicities
neuroblastoma tumor in vivo in mice (60), whereby the antitumor were found. The product known as PBI-05204 was produced in
effect arose, at least in part, from an antiangiogenic effect because response to the need for a formulation and route of administration
digoxin was also found to be effective in the chicken chorioallanto- suitable for adequately exploring the anticancer potential of an
ic membrane (CAM) assay. Thus, although limited, evidence exists oleander extract. PBI-05204 is a modified supercritical CO2 extract
for antitumor activity against human tumor xenografts. of organically grown Nerium oleander that has been especially for-
In this age of targeted therapeutics, however, where one mulated for oral administration to humans. An IND for evaluation
strives for selective effects within tumor cells so as to increase of PBI-05204 as a “botanical drug” was obtained from the FDA in
effectiveness and also minimize toxicity to normal tissues, one September, 2007, and a Phase I clinical trial in patients with solid
might reasonably question why cardiac glycosides with a known tumors has now been initiated at the University of Texas M.D.
narrow therapeutic index would ever be considered for clini- Anderson Cancer Center. Given the expanding knowledge of the
cal development as an anticancer therapy. There is little doubt multiple anticancer mechanisms for cardenolides, the outcome of
that the potential for serious cardiovascular toxicity exists with these early stage clinical trials of potent plant extracts, such as the
many, if not all cardiac glycosides, but the risks appear manage- proposed botanical drug PBI-05204 and simpler single chemical
able because the effective concentrations (i.e., in the nanomolar entities such as UNBS-1450, will be of great interest. The results
range) of these agents to control cancer-cell proliferation are well may hold the key as to whether this class of potent natural prod-
below concentrations that produce cardiac toxicity (61). Because ucts is worthy of further development as primary and/or adjuvant
the cardiovascular toxicities associated with this class of agents therapy for malignant diseases.
are well documented, careful monitoring of plasma concentra-
tions may allow for the continued safe use of digitalis and related Conclusions
compounds. Moreover, specific antibody-based treatments, such
as digoxin-specific Fab antibody fragments (Digibind; DigiTAb), as Our understanding of the spectrum of the pharmacologic activities
well as an older treatment, activated charcoal, are available to res- of cardiac glycosides has increased significantly since the discov-
cue patients receiving accidental over-medication (62, 63). ery of their effectiveness for treatment of congestive heart failure.
The desirable goal of producing a synthetically derived car- It is now recognized that certain cardiac glycosides are involved
diac glycoside with potent ability to inhibit proliferation of human in complex cell signal transduction mechanisms that may have
tumors but without the potential to cause cardiac related toxicity, important consequences in their application to the prevention
unfortunately, has not yet been fully achieved. Investigators have and/or treatment of malignant diseases. Development of clinically

February 2008
Volume 8, Issue 1 45
Review

11. Haux, J., Klepp, O., Spigset O., and Tretli, S. Digitoxin medication and
targeted, antiproliferative cardiac glycosides could be helped by cancer; case control and internal dose-response studies. BMC Cancer 1,
systematic evaluations of several formulations and chemical vari- 11 EPub (2001).
ants. Furthermore, assays of the relative presence of α1 and α3 12. Haux, J., Lam, J., Marthinsen, A.B.L., Strickert, T., and Lundgren, S.
Digitoxin, in non toxic concentrations induces cell death in Jurkat T cells
subunits in clinical samples, for example, may give some direction in vitro. Z. Onkol. 31, 14–20 (1999).
for the assessment of which cancers might be most susceptible 13. Stenkvist, B., Bengtsson, E., Eriksson, O., Holmquist, J., Nordin, B., and
to cardiac glycoside therapy. Further development of synthetic, Westman-Naeser, S. Cardiac glycosides and breast cancer. Lancet 10,
semi-synthetic, or naturally occurring cardiac glycosides, with 563 (1979)

assessment of their toxicity and structure-activity relationships, 14. Stenkvist, B. Is digitalis a therapy for breast cancer? Oncol. Rep. 6,
493–496 (1999).
might expand the possibilities of finding a cardiac glycoside with
15. Chen, J-Q., Contreras, R.G., Wang, R., Fernandez, S.V., Shoshani, L.,
a wider therapeutic index. Because of concerns with toxicity of Russo, I.H., Cereijido, M., and Russo, J. Sodium/potassium ATPase
internally used cardiac glycosides, topical formulations should (Na+,K+-ATPase) and ouabain/related cardiac glycosides: A new para-
digm for development of anti-breast cancer drugs? Breast Cancer Res.
also be considered for skin cancer prevention and/or treatment.
Treat. 96, 1–15 (2006).
Additionally, because chemotherapy has had limited benefits in 16. Nesher, M., Shpolansky, U., Rosen, H., and Lichstein, D. The digitalis-
most advanced malignancies, cardiac glycosides could also be like steroid hormones: New mechanisms of action and biological signifi-
investigated for possible adjuvant therapy. They may, for example, cance. Life Sci. 80, 2093–2107 (2007).

be of benefit in early-stage disease as well as stand-alone therapy 17. Schoner, W. and Scheiner-Bobis, G. Endogenous and exogenous car-
diac glycosides: Their roles in hypertension, salt metabolism and cell
for patients where conventional interventions are either not growth. Am. J. Physiol. Cell Physiol. 293, C509–C536 (2007).
applicable, because of patient tolerance, tumor cell sensitivity, or 18. Frese, S., Frese-Schaper, M., Anne-Catherine, A., Miescher, D.,
where a patient has run out of conventional therapeutic options Zumkehr, B., and Schmid, R.A. Cardiac glycosides initiate Apo2L/TRAIL-
induced apoptosis in non-small cell lung cancer cells by up-regulation of
altogether. Finally, consideration should be given to the use of
death receptors 4 and 5. Cancer Res. 66, 6867–5874 (2006).
selected cardiac glycosides or natural extracts containing them as 19. Sreenivasan, Y., Sarkar, A., and Manna, S.K. Oleandrin suppresses
adjuvant therapy for concomitant use with currently existing ther- activation of nuclear transcription factor-kB and activator protein-1 and
apies. Given that certain Na+,K+-ATPase subunits appear to make potentiates apoptosis induced by ceramide. Biochem. Pharmacol. 66,
2223–2239 (2003).
unique targets that are selectively expressed in tumor as opposed
20. Mijatovic, T., Van Quaquebeke, E., Delest, B., Debeir, O., Darro, F., and
to normal tissue and that the potential for cardiac toxicity is not Kiss, R. Cardiotonic steroids on the road to anti-cancer therapy. Biochim.
shared by many other currently available cancer therapies, cardiac Biophys. Acta 1776, 32–57 (2007).
glycosides should be considered as an therapeutic option for novel 21. Kaplan, J., H. Biochemistry of Na,K-ATPase. Ann. Rev. Biochem. 71,
511–535 (2002).
combination therapy. doi:10.1124/mi.8.1.8
22. Garty, H., and Karlish, S.J. Role of FXYD proteins in ion transport. Ann.
Rev. Physiol. 68, 431–459 (2006).
References 23. Geering, K. FXYD proteins: New regulators of Na-K-ATPase. Amer. J.
1. Huxtable, R.J. The erroneous pharmacology of a cat. Molec. Interven. 1, Physiol. Renal Physiol. 290, F241–F250 (2006).
75–77 (2001). 24. Davies, R.J., Sandle, G.I., and Thompson, S.M. Inhibition of the Na+,K+-
2. Gheorghiade, M., van Velduisen, D.J., and Colucci, W.S. Contemporary ATPase pump during induction of experimental colon cancer. Cancer
use of digoxin in the management of cardiovascular disorders. Biochem. Biophys. 12, 81–94 (1991).
Circulation 113, 2556–2564 (2006). 25. Avila, J., Lecuona, E., Morales, M., Soriano, A., Alonso, T., and Martin-
3. Hamad, E., Mather, P.J., Srinivasan, S., Rubin, S., Whellan, D.J., and Vasallo, P. Opposite expression pattern of the human Na,K-ATPase beta
Feldman, A.M. Pharmacologic therapy of chronic heart failure. Am. J. 1 isoform in stomach and colon adenocarcinomas. Ann. N.Y. Acad. Sci.
Cardiovasc. Drugs 7, 235–248 (2007). 834, 653–655 (1997).
4. Brewer, H. Historical perspectives on health. Early Arabic medicine. J. 26. Espineda, C., Seligson, D.B., Ball, Jr., W., Rao, J., Palotie, A., Horvath,
Roy. Soc. Health 124, 184–187 (2004). S., Huang, Y., Shi, T., and Rajasekaran, A.K. Analysis of the Na,K-
ATPase alpha- and beta-subunit expression profiles of bladder cancer
5. Watabe, M., Masudo, Y., Nakajo, S., Yoshida, T., Kuroiwa, Y., and
using tissue microarrays. Cancer 97, 1859–1868 (2003).
Nakaya, K. The cooperative interaction of two different signaling path-
ways in response to bufalin induces apoptosis in human leukemia U937 27. Rajasekaran, S.A., Ball, Jr., Bander, N.H., Liu, H., Pardee, J.D., and
cells. J. Biol. Chem. 271, 14067–14072 (1996). Rajasekaran, A.K. Reduced expression of beta-subunit of Na,K-ATPase
in human clear-cell renal cell carcinoma. J. Urol. 162, 574–580 (1999).
6. Yeh, J.Y., Huang, W.J., Kan, S.F., and Wang, P.S. Effects of bufalin and
cinobufagin on the proliferation of androgen dependent and independent 28. Factor, P., Senne, C., Dumasius, V., Ridge, K., Jaffe, H.A., Uhal, B., Gao,
prostate cancer cells. Prostate 54, 112–124 (2003). Z., and Sznajder, J.I. Overexpression of the Na+,K+-ATPase alpha1 sub-
unit increases Na+,K+-ATPase function in A549 cells. Am. J. Respir. Cell
7. Han, K.Q., Huang, G., Gu, W., Su, Y.H., Huang, X.Q., and Ling, C.Q.
Mol. Biol. 18, 741–749 (1998).
Anti-tumor activities and apoptosis-regulated mechanisms of bufalin on
the orthotopic transplantation tumor model of human hepatocellular car- 29. Winnicka, W., Bielawski, K., and Bielawska, A. Cardiac glycosides in can-
cinoma in nude mice. World J. Gastroenterol. 13, 3374–3379 (2007). cer research and cancer therapy. Acta Pol. Pharm. 63, 109–115, 2006.
8. Hartwell, J.L. and Abbott, B.J. Antineoplastic principles in plants: Recent 30. Xie, Z. and Cai, T. Na+-K+-ATPase-mediated signal transduction: From
developments in the field. Adv. Pharmacol. 7, 117–209 (1969). protein interaction to cellular function. Mol. Interv. 3, 157–168, 2003.
9. Shiratori, O. Growth inhibitory effects of cardiac glycosides and aglycones 31. Liu, J., Tian, J., Haas, M., Shapiro, J.I., Askari, A., and Xie, Z. Ouabain
on neoplastic cells: In vitro and in vivo studies. Gann 58, 521–528 (1967). interaction with cardiac Na+/K+ ATPase initates signal cascades inde-
pendent of changes in intracellular Na+ and Ca2+. J. Biol. Chem. 275,
10. Haux, J. Digitoxin is a potential anticancer agent for several types of
27838–27844 (2000).
cancer. Med. Hypotheses 53, 543–548 (1999).

46
Cardiac Glycosides as Novel Cancer Therapeutic Agents

32. Xie, Z. Ouabain interaction with cardiac Na+-K+-ATPase reveals that the 52. Rajasekaran, S.A., Hu, J., Gopal, J., Gallemore, R., Ryazantsev, S., Bok,
enzyme can act as a pump and a signal transducer. Cell. Mol. Biol. 47, D., and Rajasekaran, A.K. Na, K-ATPase inhibition alters tight junction
383–390 (2001). structure and permeability in human retinal pigment epithelial cells. Am.
33. Erdmann, E. and Schoner, W. Ouabain-receptor interactions in (Na+-K+) J. Physiol. Cell Physiol. 284, C1497–C1507 (2003).
ATPase preparations from different tissues and species. Biochim. 53. Contreras, R.G., Flores-Maldonado, G., Lazaro., A., Shoshani, L.,
Biophys. Acta 307, 386–398 (1973). Flores-Benitez, D., Larre, I., and Cereijido, M. Ouabain binding to Na+,K+-
34. Gupta, R.S., Chopra A., and Stetsko, D.K. Cellular basis for the species ATPase relaxes cell attachment and sends a specific signal (NACos) to
differences in sensitivity to cardiac glycosides (digitalis). J. Cell. Physiol. the nucleus. J. Membr. Biol. 198, 147–158 (2004).
127, 197–206 (1986). 54. Rajasekaran, S.A., Palmer, L.G., Quan, K., Harper, J.F., Ball, Jr.,W.J.,
35. Pathak, S., Multani A.S., Marayan S., Kumar, V., and Newman, R.A. Bander, N.H., Peralta Soler, A., and Rajasekaran, A.K. Na+,K+-ATPase
AnvirzelTM, an extract of Nerium oleander, induces cell death in human beta-subunit is required for epithelial polarization, suppression of inva-
but not murine cancer cells. Anticancer Drugs 11, 455–463 (2000). sion, and cell motility. Mol. Biol. Cell 12, 279–295 (2001).
36. Raghavendra, P.B., Sreenivasan, Y., and Manna, S.K. Oleandrin induces 55. Rajasekaran, S.A., Palmer, L.G., Moon, S.Y., Peralta Soler, A., Apodaca,
apoptosis in human, but not in murine cells: Dephosphorylation of Akt, G.L., Harper, J.F., Zheng, Y., and Rajasekaran, A.K. Na,K-ATPase activ-
expression of FasL, and alteration of membrane fluidity. Mol. Immunol. ity is required for the formation of tight junctions, desmosomes, and
44, 2292–2302 (2007). induction of polarity in epithelial cells. Mol. Biol. Cell. 12, 3717–3732
(2001).
37. Numazawa, S., Honna, Y., Yamamoto, T., Yoshida, T., and Kuroiwa, Y. A
cardiotonic steroid bufalin-like factor in human plasma induces leukemia 56. Dzurba, A., Ziegelhoffer, A., Vrbjar, N., Styk, J., and Slezak, J. Estradiol
cell differentiation. Leuk. Res. 19, 945–953 (1995). modulates the sodium pump in the heart sarcolemma. Mol. Cell.
Biochem. 176, 113–118 (1997).
38. Zhang, L., Nakaya, K., Yoshida, T., and Kuroiwa, Y. Induction by bufalin
of differentiation of human leukemia cells HL60, U937 and ML1 toward 57. Afaq, F., Saleem, M., Aziz, M.H., and Mukhtar, H. Inhibition of 12-O-tetrade-
macrophage/monocyte-like cells and its potent synergistic effect on the canoylphorbol-13-acetate-induced tumor promotion markers in CD-1 mouse
differentiation of human leukemia cells in combination with other induc- skin by oleandrin. Tox. Applied Pharmacol. 195, 361–369 (2004).
ers. Cancer Res. 52, 4634–4641 (1992). 58. Newman, R.A., Yang, P., Hittelman, W.N. et al. Oleandrin-mediated
39. Verheye-Dua, F.A. and Bohm, L. Influence of apoptosis on the enhance- oxidative stress in human melanoma cells. J. Exp. Therap. Oncol. 5,
ment of radiotoxicity by ouabain. Strahlenther. Onkol. 176, 186–191 (2000) 167–181 (2006).
40. Verheye-Dua, F. and Bohm, L. Na+,K+-ATPase inhibitor, ouabain accen- 59. Mijatovic, T., De Beeck, A.O., Van Quaquebeke, E., Dewelle, E., Darro,
tuates irradiation damage in human tumour cell lines. Radiat. Oncol. F., de Launoit, Y., and Kiss, R. The cardenolide UNBS1450 is able to
Investig. 6, 109–119 (1998). deactivate nuclear factor κB-mediated cytoprotective effects in human
non-small cell lung cancer cells. Mol. Cancer Ther. 5, 391–399 (2006).
41. Lawrence, T.S. Ouabain sensitizes tumor cells but not normal cells to
radiation. Int. J. Radiat. Oncol. Biol. Phys. 15, 953–958 (1998). 60. Svensson, A., Azarbayjani, F., Bäckman, U., Matsumoto, T., and
Christofferson, R. Digoxin inhibits neuroblastoma tumor growth in mice.
42. Nasu, S., Milas, L., Kawabe, S., Raju, U., and Newman, R.A.
Anticancer Res. 25, 207–212 (2005).
Enhancement of radiotherapy by oleandrin is a caspase-3 dependent
process. Cancer Lett. 185, 145–151 (2002). 61. Lopez-Lazra, M., Pastor, N., Azrak, S.S., Ayuso, M.J., Austin, C.A., and
Cortes, F. Digitoxin inhibits the growth of cancer cell lines at concentrations
43. Lucchesi, P.A. and Sweadner, K.J. Postnatal changes in Na, K-ATPase
commonly found in cardiac patients. J. Nat. Prod. 68, 1642–1645 (2005).
isoforms expression in rat cardiac ventricle. Conservation of biphasic
ouabain affinity. J. Biol. Chem. 266, 9327–9331 (1991). 62. Ujhelyi, M.R., Robert, S., Cummings, D.M., Colucci, R.D., Green,
P.J., Sailstad, J., Vlasses, P.H., and Zarowitz, B.J. Influence of digoxin
44. Noel, F., Fagoo, M., and Godfraind, T. A comparison of the affinities of
immune Fab therapy and renal dysfunction on the disposition of free and
rat (Na+,K+)-ATPase isozymes for cardioactive steroids, role of lactone
total digoxin. Ann. Intern. Med. 120, 247 (1994).
ring, sugar moiety ad KCl concentration. Biochem. Pharmacol. 40,
2611–2616 (1990). 63. Dasgupta, A., Wahed, A., Culton, L., Olsen, M., Wells, A., and Actor, J.K.
Activated charcoal is more effective than equilibrium dialysis in removing
45. O’Brien, W.J., Lingrel J.B., and Wallick, E.T. Ouabain binding kinetics of
Chinese medicines Chan Su and Dan Shen from serum and activated
the rat alpha two and alpha 3 isoforms of the sodium-potassium adenos-
charcoal also prevents further absorption of these agents from GI tract
ine triphosphate. Arch. Biochem. Biophys. 310, 32–39 (1994).
of mice: Monitoring the effect in clinical laboratory by measuring digoxin
46. Sakai, H., Suzuki, T., Maeda, M., Takahashi, Y., Horikawa, N., activity in serum. Clin. Chim. Acta 324, 51–59 (2002).
Minamimura, T., Tsukada, K., and Takeguchi, N. Up-regulation of Na+,K+-
64. Van Quaquebeke, E., Simon, G., Andre, A. et al. Identification of a novel
ATPase in α-3-isoform and down-regulation of the α 1-isoform in human
cardenolide (2″-oxovoruscharin) from Calotropis procera and the hemi-
colorectal cancer. FEBS Lett. 563, 151–154 (2004).
synthesis of novel derivatives displaying potent in vivo antitumor activi-
47. Mijatovic, T., Roland, I., Van Quaquebeke, E. et al. The α1 subunit of the ties and high in vivo tolerance; structure-activity relationship analyses. J.
sodium pump could represent a novel target to combat non-small cell Med. Chem. 48, 849–856 (2005).
lung cancers. J. Pathol. 212, 170–179 (2007).
65. Mekhail, T., Kaur, H., Ganapathi, R., Budd, G.T., Elson, P., and
48. Sreenivasan, Y., Raghavendra, P.B., and Manna, S.K. Oleandrin- Bukowski, R.M. Phase 1 trial of AnvirzelTM in patients with refractory
mediated expression of Fas potentiates apoptosis in tumor cells. J. Clin. solid tumors. Invest. New Drugs 24, 423–427 (2006).
Immunol. 26, 308–322 (2006).
66. Kupchan, S.M., Hemingway, R.J., and Doskotch, R.W. Tumor Inhibitors.
49. Mollinedo, F. and Gajate, C. Fas/CD95 death receptor and lipid rafts: IV. Apocannoside and Cymarin, the cytotoxic principles of Apocynum
New targets for apoptosis-directed cancer therapy. Drug Resist. Updat. Cannabinum L. J. Med. Chem. 7, 803–804 (1964).
9, 51–73 (2006).
67. Roy, M.C., Chang, F.R., Huang, H.C., Chiang, M.Y., and Wu, Y.C.
50. McConkey, D.J., Lin, Y., Nutt, L.K., Ozel, H.Z., and Newman, R.A. Cytotoxic principles from the formosan milkweed, Asclepias curassavica.
Cardiac glycosides stimulate Ca2+ increases and apoptosis in androgen- J. Nat. Prod. 68, 1494–1499 (2005).
independent, metastatic human prostate adenocarcinoma cells. Cancer
68. Kaneda, N., Chai, H., Pezzuto, J.M., Kinghorn, A.D., Farnsworth, N.R.,
Res. 60, 3807–3812 (2000).
Tuchinda, P., Udchachon, J., Santisuk, T., and Reutrakul, V. Cytotoxic
51. Newman, R.A., Kondo, Y., Yokoyama, T., Dixon, S., Cartwright, C., Chan, activity of cardenolides from Beaumontia brevituba Stems. Planta
D., Johansen, M., and Yang, P. Autophagic cell death of human pancre- Medica 58, 429–431 (1992).
atic tumor cells mediated by oleandrin, a lipid-soluble cardiac glycoside.
69. Yeh, J.Y., Huang, W.J., Kan, S.F., and Wang, P.S Effects of bufalin and
Integr. Cancer Ther. 6, 354–364 (2007).
cinobufagin on the proliferation of androgen dependent and independent

February 2008
Volume 8, Issue 1 47
Review

prostate cancer cells. Prostate 54, 112–124 (2003). asper. J. Nat. Prod. 48, 981–985 (1985).
70. Kamano, Y., Kotake, A., Hashima, H. et al. Structure-cytotoxic activity 91. Ueda, J.Y., Tezuka, Y., Banskota, A.H., Tran, Q.L., Tran, Q.K., Saiki, I., and
relationship for the toad poison bufadienolides. Bioorg. Med. Chem. 6, Kadota, S. Constituents of the Vietnamese medicinal plant Streptocaulon
1103–1115 (1998). juventas and their antiproliferative activity against the human HT-1080
71. Choedon, T., Mathan, G., Arya, S., Kumar, V.L., and Kumar, V. fibrosarcoma cell line. J. Nat. Prod. 66, 1427–1433 (2003).
Anticancer and cytotoxic properties of the latex of Calotropis procera 92. Huang, YT., Chueh, S.C., Teng, C.M., and Guh, J.H. Investigation of oua-
in a transgenic mouse model of hepatocellular carcinoma. World J. bain-induced anticancer effect in human androgen-independent prostate
Gastroenterol. 12, 2517–2522 (2006). cancer PC-3 cells. Biochem. Pharmacol. 67, 727–733 (2004).
72. Smit, H.F., Woerdenbag, H.J., Singh, R.H., Meulenbeld, G.J., Labadie, 93. Decosterd, L. The differential cytotoxicity of cardenolides from Thevetia
R.P., and Zwaving, J.H. Ayurvedic herbal drugs with possible cytostatic ahouia. Phytotherapy Res. 8, 74–77 (1994).
activity. J. Ethnopharm. 47, 75–84 (1995). 94. Iizuka, M., Warashina, T., and Noro, T. Bufadienolides and a new lignan from
73 Laphookhieo, S., Cheenpracha, S., Karalai, C., Chantrapromma, S., the bulbs of Urginea maritima. Chem. Pharm. Bull. 49, 282–286 (2001).
Rat-a-Pa, T., Ponglimanont, C., and Chantrapromma, K. Cytotoxic carde- 95. Bielawski, K., Winnicka, K, and Bielawska, A. Inhibition of DNA topoi-
nolide glycoside from the seeds of Cerbera odollam. Phytochemistry 65, somerases I and II, and growth inhibition of breast cancer MCF-7
507–510 (2004). cells by ouabain, digoxin and Proscillaridin A. Biol. Pharm. Bull. 29,
74 Hembree, J.A., Chang, C.J., McLaughlin, J.L., Peck, G., and Cassady, 1493–1497 (2006).
J.M. Potential antitumor agents: A cytotoxic cardenolide from Coronilla 96. Jha, S. and Sen, S. Quantitation of principal bufadienolides in different
varia. J. Nat. Prod. 42, 293–298 (1979). cytotypes of Urginea indica. Planta Med. 47, 43–45 (1983).
75 Ankli, A., Heilmann, J., Heinrich, M., and Sticher, O. Cytotoxic carde- 97. Manna, S.K., Sreenivasan, Y., and Sarkar, A. Cardiac glycoside inhibits
nolides and antibacterial terpenoids from Crossopetalum gaumeri. IL-8 induced biological responses by downregulating IL-8 receptors
Phytochemistry 54, 531–537 (2000). through altering membrane fluidity. J. Cell. Physiol. 207, 195–207 (2006).
76. Yeh, J.Y., Hunag, W.J., Kan, S.F., and Wang, P.S. Inhibitory effects of 98. Manna, S.K., Sah, N.K., Newman, R.A., Cisneros, A., and Aggarwal,
digitalis on the proliferation of androgen dependent and independent B.B. Oleandrin suppresses activation of nuclear transcription factor-kap-
prostate cancer cells. J. Urol. 166, 1937–1942 (2001). paB, activator protein-1, and c-Jun NH2 terminal kinase. Cancer Res.
77. López-Lázaro, M., Pastor, N., Azrak, S.S., Ayuso, M.J., Austin, C.A., and 60, 3838–3847 (2000).
Cortez, F. Digitoxin inhibits the growth of cancer cell lines at concentrations 99. Raghavendra, R.B., Sreenivasan, Y., Ramesh, G.T., and Manna, S.K.
commonly found in cardiac patients. J. Nat. Prod. 68, 1642–1645 (2005). Cardiac glycoside induces cell death via FasL by activating calcineurin
78. Cao, S., Brodie, P.J., Miller, J.S. et al. Antiproliferative cardenolides of and NF-AT, but apoptosis initially proceeds through activation of cas-
an Elaeodendron sp. from the Madagascar rain forest. J. Nat. Prod. 70, pases. Apoptosis 12, 307–318 (2007).
1064–1067 (2007). 100. Smith, J.A., Madden, T., Vijjeswarapu, M., and Newman, R.A. Inhibition
79. Kitanaka, S., Takido, M., Mizoue, K., and Nakaike, S. Cytotoxic cardenolides of fibroblast growth factor-2 (FGF-2) from the prostate cancer cell lines
from woods of Eunymus alata. Chem. Pharm. Bull. 44, 615–617 (1996). PC3 and DU-145 by Anvirzel and its cardiac glycoside component, ole-
80. Baek, N.I., Lee, Y.H., Park, J.D., Kim, S.I., and Ahn, B.Z. Euonymoside andrin. Biochem. Pharmacol. 62, 469–472 (2001).
A: A new cytotoxic cardenolide glycoside from the bark of Euonymus 101. Kurosawa, M., Numazawa, S., Tani, Y., and Yoshida, T. ERK signaling
sieboldianus. Planta Medica 60, 26–29 (1994). mediates the induction of inflammatory cytokines by bufalin in human
81. Rovinski, J.M., Tewalt, G.L., and Sneden, A.T. Maquiroside A, A new monocytic cells. Am. J. Physiol. 278, C500–C508 (2000).
cytotoxic cardiac glycoside from Maquira calophylla. J. Nat. Prod. 50, 102. Kawazoe, N., Watabe, M., Masuda, Y., Nakajo, S., and Nakaya, K. Tiam
211–216 (1987). 1 is involved in the regulation of bufalin-induced apoptosis in human leu-
82. Raghavendra, P.B., Sreenivasan, Y., Ramesh, G.T., and Manna, S.K. kemia cells. Oncogene 18, 2413–2421 (1999).
Cardiac glycosides induced cell death vai FasL by activating calcineurin 103. Watabe, M., Ito, K., Masuda, Y., Navajo, S., and Nakaya, K. Activation of
and NF-AT, but apoptosis initially proceeds through activation of cas- AP-1 is required for bufalin-induced apoptosis in human leucemia U937
paces. Apoptosis 12, 307–318 (2007). cells. Oncogene 16, 779–787 (1998).
83. Zhao, M., Bai, L., Wang, L. et al. Bioactive cardenolides from the stems 104. Watabe, M., Nakajo, S., Yoshida, T., Kuroiwa, Y., and Nakaya, K.
and twigs of Nerium Oleander. J. Nat. Prod. 70, 1098–1103 (2007). Treatment of U937 cells with bufalin induces the translation of casein
84. Gil, R.R., Lin, L.Z., Chai, H.B., Pezzuto, J.M., and Cordell, G.A. kinase 2 and modulates the activity of topoisomerase II prior to induction
Cardenolides from Nierembergia aristata. J. Nat. Prod. 58, 848–856 (1995). of apoptosis. Cell Growth Differ. 8, 871–879 (1997).
85. Kelly, R.B., Daniels, E.G., and Spaulding, L.B. Cytotoxicity of cardiac 105. Hashimoto, S., Jing, Y., Kawazoe, N., Masuda, Y., Nakajo, S., Yoshida, T.,
principles. J. Med. Chem. 8, 547–548 (1965). Kuroiwa, Y., and Nakaya, K. Bufain reduces the level of topoisomerase II
in human leukemia cells and affects the cytotoxicity of anticancer drugs.
86. Hamed, A.I., Plaza, A., Balestrieri, M.L., Mahalel, U.A., Springuel, I.V.,
Leuk. Res. 21, 875–883 (1997).
Oleszek, W., Pizza, C., and Piacente, S. Cardenolide glycosides from
Pergularia tomentosa and their proapoptotic activity in Kaposi’s sarcoma 106. Kurosawa, M., Tari, Y., Nishimura, S., Numazawa, S., and Yoshida,
cells. J. Nat. Prod. 69, 1319–1322 (2006). T. Distinct PKC isozymes regulate bufalin-induced differentiation and
apoptosis in human monocytic cells. Am. J. Physiol. Cell Physiol. 280,
87. Spera, D., Siciliano, T., DeTommasi, N., Braca, A., and Vessieres, A.
C459–C464 (2001).
Antiproliferative cardenolides from Periploca graeca. Planta Med. 73,
384–387 (2007). 107. Yamada, K., Hino, T., Tomoyasu, S., Honma, Y., and Tsuruoka, N.
Enhancement by bufalin of retinoic acid-induced differentiation of acute pro-
88. Umebayashi, C., Yamamoto, N., Nakao, H., Toi, Y., Chikahisa-
myelocytic leukemia cells in primary culture. Leuk. Res. 22, 589–595 (1998).
Muramatsu, L., Kanemaru, K., Masuda, T., and Oyama, Y. Flow
Cytometric Estimation of cytotoxic activity of Rhodexin A isolated from 108. Nasu, K., Nishida, M., Ueda, T., Takai, N., Bing, S., Narahara, H., and
Rhodea japonica in human leukemia K562 cells. Biol. Pharm. Bull. 26, Miyakawa, I. Bufalin induces apoptosis and the G0/G1 cell cycle arrest
627–630 (2003). of endometriotic stromal cells: A promising agent for the treatment of
endometriosis. Mol. Human Reprod. 11, 817–823 (2005).
89. Wang, T.-M., Hojo, T., Ran, F.X., Wang, R.F., Wang, R.Q., Chen, R.B.,
Cui, J.R., Shang, M.Y., and Cai, S.Q. Cardenolides from Saussurea stella 109. Huang, Y.T., Chueh, S.C., Teng, C.M., and Guh, J.H. Investigation of
with cytotoxicity toward cancer cells. J. Nat. Prod. 70, 1429–1433 (2007). ouabain-induced anticancer effect in human androgen-independent
prostate cancer PC-3 cells. Biochem. Pharmacol. 15, 727–733 (2004).
90. Fiebig, M., Duh, C.Y., Pezzuto, J.M., Kinghorn, A.D., and Farnsworth,
N.R. Plant Anticancer Agents, XLI. Cardiac glycosides from Streblus 110. Kometiani, P., Liu, L. and Askari, A. Digitalis-induced signaling by Na+,K+-

48
Cardiac Glycosides as Novel Cancer Therapeutic Agents

ATPase in human breast cancer cells. Molec. Pharmacol. 67, 929–936 (2005). Keith I. Block, MD, co-founded
111. Liu, J., Tian, J., Haas, M., Shapiro, J.I., Askari, A., and Xie, Z. Ouabain and is Medical/Scientific Director
interaction with cardiac Na+,K+-ATPase initiates signal cascades inde-
pendent of changes in intracellular Na+ and Ca2+ concentrations. J. Biol.
of the Block Center for Integrative
Chem. 275, 27838–27844 (2000). Cancer Treatment. He is a member
112. Contreras, R.G., Flores-Beni Tez, D., Flores-Maldonado, C., Larre, I., of the National Cancer Institute’s
Shoshani, L., and Cereijido, M. Na+,K+-ATPase and hormones: New roles Physician Data Query (PDQ)
for an old enzyme and an old inhibitor. Cell Mol. Biol. 52, 31–40 (2006).
Cancer CAM Editorial Board, the
113. Harwood, S. and Yaqoob, M.M. Ouabain-induced cell signaling. Front.
Biosci. 10, 2011–2017 (2005). editor-in-chief of the peer-reviewed
114. Kulikov, A., Eva, A., Kirch, U., Boldyrev, A., and Scheiner-Bobis, G. journal Integrative Cancer Therapies,
Ouabain activates signaling pathways associated with cell death in and a Clinical Assistant Professor
human neuroblastoma. Biochim. Biophys. Acta 1768, 1691–1702 (2007).
at the College of Medicine at the University of Illinois, Chicago.
115. Glibert, M. and Knox, S. Influence of Bcl-2 overexpression on Na+,K+-
ATPase pump activity: Correlation with radiation-induced programmed In collaboration with the University of Illinois and other univer-
cell death. J. Cell Physiol. 171, 299–304 (1997). sity facilities in the US and Israel, Dr. Block conducts research in
116. Lopez-Lazaro, M. Digitoxin as an anticancer agent with selectivity for nutrition and in the use of natural medicines in cancer treatment.
cancer cells: Possible mechanisms involved. Expert Opin. Ther. Targets
11, 1043–1053 (2007).
117. Johansson, S., Lindholm, P., Gullbo, J., Larsson, R., Bohlim, L., and
Peiying Yang, PhD, (Collaborator)
Claeson, P. Cytotoxicity of digitoxin and related cardiac glycosides in is an Assistant professor in the
human tumor cells. Anticancer Drugs 12, 475–483 (2001). Department of Experimental
118. Bielawski, K., Winnicka, K., and Bielawska, A. Inhibition of DNA topoi- Therapeutics. Dr. Yang’s research
somerase I and II and growth inhibition of breast cancer MCF-7 cells by oua-
bain, digoxin and proscillaridin A. Biol. Pharm. Bull. 29, 1493–1497 (2006). has focused on bioactive lipids
119. Yang, Q., Huang, W., Jozwik, C. et al. Cardiac glycosides inhibit TNF- and natural products in cancer
alpha/NF-κB signaling by blocking recruitment of TNF receptor-associat- development and prevention. She
ed death domain to the TNF receptor. Proc. Natl. Acad. Sci. U.S.A. 102,
developed a rapid, specific, and
9631–9636 (2005).
120. Mijatovic, T., Matthieu, V., Gaussin, J.F., DeNeve, N., Ribaucour, F.,
sensitive method for simultaneous-
VanQuaquebeke, E., Dumont, P., Darro, F., and Kiss, R. Cardenolide- ly determination of arachidonate
induced lysosomal membrane permeabiliazation demonstrates thera- metabolites in various biological matrices. Additionally, Dr. Yang
peutic benefits in experimental human non-small cell lung cancers.
Neoplasia 8, 402–412 (2006).
is interested in the effects of nutritional supplements, such as fish
oil, and Chinese herbal medicine.
Robert A. Newman, PhD,
received his undergraduate train- Alison Pawlus, RPh, PhD, com-
ing at the University of Rhode pleted her graduate training, in
Island and graduate training at the 2007, in Pharmacognosy at the
University of Connecticut, where University of Illinois at Chicago in
he obtained MS and PhD degrees the laboratory of Dr. A. Douglas
in pharmacology and toxicology. Kinghorn. She is a Postdoctoral
His postgraduate work was per- Fellow in the Department of
formed at the Medical School of Experimental Therapeutics at the
Georgia and at the University of University of Texas, M.D. Anderson
Vermont Medical School. After a sabbatical at Stanford University Cancer Center where she studies
in 1983, Dr. Newman joined the faculty of the University of Texas the use of natural products for cancer treatment and prevention.
M.D. Anderson Cancer Center. He is a Professor of Experimental
Therapeutics and holds the D.B. Land Professorship. Dr. Newman
also jointly runs the Pharmacology and Analytical Core lab for the
institution as well as co-directs the Pharmaceutical Development
Center that has introduced more than six compounds into the
clinic over the past seven years. He is the author of over 250 peer-
reviewed publications and several books. His current research
deals with the science of nutraceuticals and understanding how
these can be specifically applied for prevention and treatment of
inflammation and malignant disease. E-mail rnewman@mdander-
son.org; fax (713) 563-9093.

February 2008
Volume 8, Issue 1 49

Potrebbero piacerti anche